101
|
Serna J, Bergwitz C. Importance of Dietary Phosphorus for Bone Metabolism and Healthy Aging. Nutrients 2020; 12:E3001. [PMID: 33007883 PMCID: PMC7599912 DOI: 10.3390/nu12103001] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 09/25/2020] [Accepted: 09/26/2020] [Indexed: 12/13/2022] Open
Abstract
Inorganic phosphate (Pi) plays a critical function in many tissues of the body: for example, as part of the hydroxyapatite in the skeleton and as a substrate for ATP synthesis. Pi is the main source of dietary phosphorus. Reduced bioavailability of Pi or excessive losses in the urine causes rickets and osteomalacia. While critical for health in normal amounts, dietary phosphorus is plentiful in the Western diet and is often added to foods as a preservative. This abundance of phosphorus may reduce longevity due to metabolic changes and tissue calcifications. In this review, we examine how dietary phosphorus is absorbed in the gut, current knowledge about Pi sensing, and endocrine regulation of Pi levels. Moreover, we also examine the roles of Pi in different tissues, the consequences of low and high dietary phosphorus in these tissues, and the implications for healthy aging.
Collapse
Affiliation(s)
- Juan Serna
- Yale College, Yale University, New Haven, CT 06511, USA;
| | - Clemens Bergwitz
- Section of Endocrinology and Metabolism, Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06519, USA
| |
Collapse
|
102
|
Structural Mechanism of Cooperative Regulation of Calcium-Sensing Receptor-Mediated Cellular Signaling. CURRENT OPINION IN PHYSIOLOGY 2020; 17:269-277. [PMID: 33709045 DOI: 10.1016/j.cophys.2020.08.020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Calcaium sensing receptors (CaSRs) play a central role in regulating extracellular calcium (Ca2+) homeostasis and many (patho)physiological processes. This regulation is primarily orchestrated in response to extracellular stimuli via the extracellular domain (ECD). This paper first reviews the modeled structure of the CaSR ECD and the prediction and investigation of the Ca2+ and amino acid binding sites. Several recently solved X-ray structures are then compared to support a proposed CaSR activation model involving functional cooperativity. The review also discusses recent implications for drug development. These studies provide new insights into the molecular basis of diseases and the design of therapeutic agents that target CaSR and other family C G protein-coupled receptors (cGPCRs).
Collapse
|
103
|
Magno AL, Leatherbarrow KM, Brown SJ, Wilson SG, Walsh JP, Ward BK. Functional Analysis of Calcium-Sensing Receptor Variants Identified in Families Provisionally Diagnosed with Familial Hypocalciuric Hypercalcaemia. Calcif Tissue Int 2020; 107:230-239. [PMID: 32638038 DOI: 10.1007/s00223-020-00715-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 06/18/2020] [Indexed: 12/11/2022]
Abstract
Identification of variants in the calcium-sensing receptor (CASR) gene is an important means of distinguishing between familial hypocalciuric hypercalcaemia (FHH) and primary hyperparathyroidism. However, identification and bioinformatics analysis of genetic variants alone is now considered insufficient as definitive proof; additional functional assessment is required to diagnose FHH with certainty. We identified two novel variants, D433Y and C739Y, and one previously reported variant G509R in the CASR of four kindreds provisionally diagnosed with FHH and aimed to functionally characterise these variants to confirm the diagnosis. Variant receptors were cloned as FLAG-tagged constructs into the mammalian expression vector, pcDNA3.1. Wild type and variant receptor constructs were expressed in HEK293 cells and their expression assessed by Western blot analysis and their functionality analysed using an IP-One assay which measures myo-inositol 1-phosphate accumulation following CaSR activation. Western blot analysis showed that the D433Y receptor had diminished mature glycosylated receptor compared with wild type CaSR whereas the G509R receptor had a complete lack of mature receptor. The C739Y receptor was consistently overexpressed. Functional assessment showed the D433Y receptor to be mildly inactivating at physiological calcium concentrations whereas the G509R receptor was inactive at all calcium concentrations. By contrast, the C739Y variant was activating compared to wild type receptor which is inconsistent with it causing FHH. We conclude that functional assessment of CaSR variants using the IP-One assay was useful in the investigation of suspected FHH probands, confirming the D433Y and G509R variants as likely pathogenic/pathogenic, but dismissing the C739Y variant as causing FHH.
Collapse
Affiliation(s)
- Aaron L Magno
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Block C, Level 1, Hospital Avenue, Nedlands, WA, Australia
| | - Kassandra M Leatherbarrow
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Block C, Level 1, Hospital Avenue, Nedlands, WA, Australia
| | - Suzanne J Brown
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Block C, Level 1, Hospital Avenue, Nedlands, WA, Australia
| | - Scott G Wilson
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Block C, Level 1, Hospital Avenue, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA, Australia
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - John P Walsh
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Block C, Level 1, Hospital Avenue, Nedlands, WA, Australia
- Medical School, University of Western Australia, Nedlands, WA, Australia
| | - Bryan K Ward
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Block C, Level 1, Hospital Avenue, Nedlands, WA, Australia.
- Harry Perkins Institute of Medical Research, Centre for Medical Research, QEII Medical Centre, University of Western Australia, Nedlands, WA, Australia.
| |
Collapse
|
104
|
Illuminating the allosteric modulation of the calcium-sensing receptor. Proc Natl Acad Sci U S A 2020; 117:21711-21722. [PMID: 32817431 DOI: 10.1073/pnas.1922231117] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Many membrane receptors are regulated by nutrients. However, how these nutrients control a single receptor remains unknown, even in the case of the well-studied calcium-sensing receptor CaSR, which is regulated by multiple factors, including ions and amino acids. Here, we developed an innovative cell-free Förster resonance energy transfer (FRET)-based conformational CaSR biosensor to clarify the main conformational changes associated with activation. By allowing a perfect control of ambient nutrients, this assay revealed that Ca2+ alone fully stabilizes the active conformation, while amino acids behave as pure positive allosteric modulators. Based on the identification of Ca2+ activation sites, we propose a molecular basis for how these different ligands cooperate to control CaSR activation. Our results provide important information on CaSR function and improve our understanding of the effects of genetic mutations responsible for human diseases. They also provide insights into how a receptor can integrate signals from various nutrients to better adapt to the cell response.
Collapse
|
105
|
Wang C, Jia Q, Sun C, Jing C. Calcium sensing receptor contribute to early brain injury through the CaMKII/NLRP3 pathway after subarachnoid hemorrhage in mice. Biochem Biophys Res Commun 2020; 530:651-657. [PMID: 32768195 DOI: 10.1016/j.bbrc.2020.07.081] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 07/17/2020] [Indexed: 12/26/2022]
Abstract
The subversive role of Calcium sensing receptor (CaSR) in cerebral ischemia and traumatic brain injury has been recently reported. Nevertheless, the role of CaSR in early brain injury (EBI) after subarachnoid hemorrhage (SAH) remains unexplored. Using the endovascular perforation model in mice, this study was aimed at investigating the role and potential mechanism of CaSR in EBI after SAH. Gadolinium trichloride (GdCI3), an agonist of CaSR, and NPS-2143, an inhibitor of CaSR, were administered intraperitoneally. The CaMKII inhibitor KN-93 was injected to intracerebroventricular. We found that CaSR expression was increased and widely expressed in neurons, astrocytes, and microglia after SAH. GdCI3 further deteriorated neurological function, brain edema, neurodegeneration, which were alleviated by NPS-2143. Also, GdCI3 increased the level of CaMKII phosphorylation, and upregulated expression of NLRP3, cleaved caspase-1, and IL-1β, which were attenuated by NPS-2143. Besides, CaMKII inhibitor KN-93 down-regulated the upregulated expression of NLRP3, cleaved caspase-1, and IL-1β induced by GdCI3. In conclusion, CaSR activation promotes early brain injury, which may be related to the CaMKII/NLRP3 signaling pathway.
Collapse
Affiliation(s)
- Chun Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qingbin Jia
- Department of Neurosurgery, Liaocheng People's Hospital, Liaocheng, Shandong, China
| | - Chenjun Sun
- Department of Neurosurgery, Shaoxing Central Hospital, Shaoxing, Zhejiang, China
| | - Chaohui Jing
- Department of Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
106
|
Park J, Fu Z, Frangaj A, Liu J, Mosyak L, Shen T, Slavkovich VN, Ray KM, Taura J, Cao B, Geng Y, Zuo H, Kou Y, Grassucci R, Chen S, Liu Z, Lin X, Williams JP, Rice WJ, Eng ET, Huang RK, Soni RK, Kloss B, Yu Z, Javitch JA, Hendrickson WA, Slesinger PA, Quick M, Graziano J, Yu H, Fiehn O, Clarke OB, Frank J, Fan QR. Structure of human GABA B receptor in an inactive state. Nature 2020; 584:304-309. [PMID: 32581365 PMCID: PMC7725281 DOI: 10.1038/s41586-020-2452-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 05/01/2020] [Indexed: 01/21/2023]
Abstract
The human GABAB receptor-a member of the class C family of G-protein-coupled receptors (GPCRs)-mediates inhibitory neurotransmission and has been implicated in epilepsy, pain and addiction1. A unique GPCR that is known to require heterodimerization for function2-6, the GABAB receptor has two subunits, GABAB1 and GABAB2, that are structurally homologous but perform distinct and complementary functions. GABAB1 recognizes orthosteric ligands7,8, while GABAB2 couples with G proteins9-14. Each subunit is characterized by an extracellular Venus flytrap (VFT) module, a descending peptide linker, a seven-helix transmembrane domain and a cytoplasmic tail15. Although the VFT heterodimer structure has been resolved16, the structure of the full-length receptor and its transmembrane signalling mechanism remain unknown. Here we present a near full-length structure of the GABAB receptor, captured in an inactive state by cryo-electron microscopy. Our structure reveals several ligands that preassociate with the receptor, including two large endogenous phospholipids that are embedded within the transmembrane domains to maintain receptor integrity and modulate receptor function. We also identify a previously unknown heterodimer interface between transmembrane helices 3 and 5 of both subunits, which serves as a signature of the inactive conformation. A unique 'intersubunit latch' within this transmembrane interface maintains the inactive state, and its disruption leads to constitutive receptor activity.
Collapse
Affiliation(s)
- Jinseo Park
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Ziao Fu
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Aurel Frangaj
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Jonathan Liu
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Lidia Mosyak
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Tong Shen
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA, USA
| | - Vesna N Slavkovich
- Department of Environmental Health Sciences, Columbia University, New York, NY, USA
| | - Kimberly M Ray
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Jaume Taura
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Baohua Cao
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Yong Geng
- Department of Pharmacology, Columbia University, New York, NY, USA
- Key Laboratory of Receptor Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hao Zuo
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Yongjun Kou
- Key Laboratory of Receptor Research, Center for Structure and Function of Drug Targets, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Robert Grassucci
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Shaoxia Chen
- MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Zheng Liu
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
| | - Xin Lin
- Department of Psychiatry, Columbia University, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Justin P Williams
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - William J Rice
- National Resource for Automated Molecular Microscopy, Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY, USA
| | - Edward T Eng
- National Resource for Automated Molecular Microscopy, Simons Electron Microscopy Center, New York Structural Biology Center, New York, NY, USA
| | - Rick K Huang
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Rajesh K Soni
- Proteomics Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Brian Kloss
- Center on Membrane Protein Production and Analysis, New York Structural Biology Center, New York, NY, USA
| | - Zhiheng Yu
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA, USA
| | - Jonathan A Javitch
- Department of Pharmacology, Columbia University, New York, NY, USA
- Department of Psychiatry, Columbia University, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
| | - Wayne A Hendrickson
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA
- Center on Membrane Protein Production and Analysis, New York Structural Biology Center, New York, NY, USA
| | - Paul A Slesinger
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Matthias Quick
- Department of Psychiatry, Columbia University, New York, NY, USA
- Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY, USA
| | - Joseph Graziano
- Department of Environmental Health Sciences, Columbia University, New York, NY, USA
| | - Hongtao Yu
- Howard Hughes Medical Institute, Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Oliver Fiehn
- NIH West Coast Metabolomics Center, University of California Davis, Davis, CA, USA
| | - Oliver B Clarke
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY, USA.
- Department of Anesthesiology and the Irving Institute for Clinical and Translational Research, Columbia University, New York, NY, USA.
| | - Joachim Frank
- Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY, USA.
- Department of Biological Sciences, Columbia University, New York, NY, USA.
| | - Qing R Fan
- Department of Pharmacology, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
107
|
Leach K, Hannan FM, Josephs TM, Keller AN, Møller TC, Ward DT, Kallay E, Mason RS, Thakker RV, Riccardi D, Conigrave AD, Bräuner-Osborne H. International Union of Basic and Clinical Pharmacology. CVIII. Calcium-Sensing Receptor Nomenclature, Pharmacology, and Function. Pharmacol Rev 2020; 72:558-604. [PMID: 32467152 PMCID: PMC7116503 DOI: 10.1124/pr.119.018531] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The calcium-sensing receptor (CaSR) is a class C G protein-coupled receptor that responds to multiple endogenous agonists and allosteric modulators, including divalent and trivalent cations, L-amino acids, γ-glutamyl peptides, polyamines, polycationic peptides, and protons. The CaSR plays a critical role in extracellular calcium (Ca2+ o) homeostasis, as demonstrated by the many naturally occurring mutations in the CaSR or its signaling partners that cause Ca2+ o homeostasis disorders. However, CaSR tissue expression in mammals is broad and includes tissues unrelated to Ca2+ o homeostasis, in which it, for example, regulates the secretion of digestive hormones, airway constriction, cardiovascular effects, cellular differentiation, and proliferation. Thus, although the CaSR is targeted clinically by the positive allosteric modulators (PAMs) cinacalcet, evocalcet, and etelcalcetide in hyperparathyroidism, it is also a putative therapeutic target in diabetes, asthma, cardiovascular disease, and cancer. The CaSR is somewhat unique in possessing multiple ligand binding sites, including at least five putative sites for the "orthosteric" agonist Ca2+ o, an allosteric site for endogenous L-amino acids, two further allosteric sites for small molecules and the peptide PAM, etelcalcetide, and additional sites for other cations and anions. The CaSR is promiscuous in its G protein-coupling preferences, and signals via Gq/11, Gi/o, potentially G12/13, and even Gs in some cell types. Not surprisingly, the CaSR is subject to biased agonism, in which distinct ligands preferentially stimulate a subset of the CaSR's possible signaling responses, to the exclusion of others. The CaSR thus serves as a model receptor to study natural bias and allostery. SIGNIFICANCE STATEMENT: The calcium-sensing receptor (CaSR) is a complex G protein-coupled receptor that possesses multiple orthosteric and allosteric binding sites, is subject to biased signaling via several different G proteins, and has numerous (patho)physiological roles. Understanding the complexities of CaSR structure, function, and biology will aid future drug discovery efforts seeking to target this receptor for a diversity of diseases. This review summarizes what is known to date regarding key structural, pharmacological, and physiological features of the CaSR.
Collapse
Affiliation(s)
- Katie Leach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Fadil M Hannan
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Tracy M Josephs
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Andrew N Keller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Thor C Møller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Donald T Ward
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Enikö Kallay
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Rebecca S Mason
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Rajesh V Thakker
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Daniela Riccardi
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Arthur D Conigrave
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Hans Bräuner-Osborne
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| |
Collapse
|
108
|
Abstract
GABA (γ-aminobutyric acid) stimulation of the metabotropic GABAB receptor results in prolonged inhibition of neurotransmission that is central to brain physiology1. GABAB belongs to the Family C of G protein-coupled receptors (GPCRs), which operate as dimers to relay synaptic neurotransmitter signals into a cellular response through the binding and activation of heterotrimeric G proteins2,3. GABAB, however, is unique in its function as an obligate heterodimer in which agonist binding and G protein activation take place on distinct subunits4,5. Here we show structures of heterodimeric and homodimeric full-length GABAB receptors. Complemented by cellular signaling assays and atomistic simulations, the structures reveal an essential role for the GABAB extracellular loop 2 (ECL2) in relaying structural transitions by ordering the linker connecting the extracellular ligand-binding domain to the transmembrane region. Furthermore, the ECL2 of both GABAB subunits caps and interacts with the hydrophilic head of a phospholipid occupying the extracellular half of the transmembrane domain, thereby providing a potentially crucial link between ligand binding and the receptor core that engages G protein. These results provide a starting framework to decipher mechanistic modes of signal transduction mediated by GABAB dimers and have important implications for rational drug design targeting these receptors.
Collapse
|
109
|
Sutkeviciute I, Vilardaga JP. Structural insights into emergent signaling modes of G protein-coupled receptors. J Biol Chem 2020; 295:11626-11642. [PMID: 32571882 DOI: 10.1074/jbc.rev120.009348] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 06/21/2020] [Indexed: 12/21/2022] Open
Abstract
G protein-coupled receptors (GPCRs) represent the largest family of cell membrane proteins, with >800 GPCRs in humans alone, and recognize highly diverse ligands, ranging from photons to large protein molecules. Very important to human medicine, GPCRs are targeted by about 35% of prescription drugs. GPCRs are characterized by a seven-transmembrane α-helical structure, transmitting extracellular signals into cells to regulate major physiological processes via heterotrimeric G proteins and β-arrestins. Initially viewed as receptors whose signaling via G proteins is delimited to the plasma membrane, it is now recognized that GPCRs signal also at various intracellular locations, and the mechanisms and (patho)physiological relevance of such signaling modes are actively investigated. The propensity of GPCRs to adopt different signaling modes is largely encoded in the structural plasticity of the receptors themselves and of their signaling complexes. Here, we review emerging modes of GPCR signaling via endosomal membranes and the physiological implications of such signaling modes. We further summarize recent structural insights into mechanisms of GPCR activation and signaling. We particularly emphasize the structural mechanisms governing the continued GPCR signaling from endosomes and the structural aspects of the GPCR resensitization mechanism and discuss the recently uncovered and important roles of lipids in these processes.
Collapse
Affiliation(s)
- Ieva Sutkeviciute
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jean-Pierre Vilardaga
- Laboratory for GPCR Biology, Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
110
|
Naveh-Many T, Volovelsky O. Parathyroid Cell Proliferation in Secondary Hyperparathyroidism of Chronic Kidney Disease. Int J Mol Sci 2020; 21:ijms21124332. [PMID: 32570711 PMCID: PMC7352987 DOI: 10.3390/ijms21124332] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Secondary hyperparathyroidism (SHP) is a common complication of chronic kidney disease (CKD) that correlates with morbidity and mortality in uremic patients. It is characterized by high serum parathyroid hormone (PTH) levels and impaired bone and mineral metabolism. The main mechanisms underlying SHP are increased PTH biosynthesis and secretion as well as increased glandular mass. The mechanisms leading to parathyroid cell proliferation in SHP are not fully understood. Reduced expressions of the receptors for calcium and vitamin D contribute to the disinhibition of parathyroid cell proliferation. Activation of transforming growth factor-α-epidermal growth factor receptor (TGF-α-EGFR), nuclear factor kappa B (NF-kB), and cyclooxygenase 2- prostaglandin E2 (Cox2-PGE2) signaling all correlate with parathyroid cell proliferation, underlining their roles in the development of SHP. In addition, the mammalian target of rapamycin (mTOR) pathway is activated in parathyroid glands of experimental SHP rats. Inhibition of mTOR by rapamycin prevents and corrects the increased parathyroid cell proliferation of SHP. Mice with parathyroid-specific deletion of all miRNAs have a muted increase in serum PTH and fail to increase parathyroid cell proliferation when challenged by CKD, suggesting that miRNA is also necessary for the development of SHP. This review summarizes the current knowledge on the mechanisms of parathyroid cell proliferation in SHP.
Collapse
Affiliation(s)
- Tally Naveh-Many
- Minerva Center for Calcium and Bone Metabolism, Nephrology Services, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel;
- The Wohl Institute for Translational Medicine, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel
| | - Oded Volovelsky
- The Wohl Institute for Translational Medicine, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel
- Pediatric Nephrology Unit and Research Lab, Hadassah Hebrew University Medical Center, Jerusalem 91120, Israel
- Correspondence: ; Tel.: +972-26777213
| |
Collapse
|
111
|
Dharmaraj P, Gorvin CM, Soni A, Nelhans ND, Olesen MK, Boon H, Cranston T, Thakker RV, Hannan FM. Neonatal Hypocalcemic Seizures in Offspring of a Mother With Familial Hypocalciuric Hypercalcemia Type 1 (FHH1). J Clin Endocrinol Metab 2020; 105:5801090. [PMID: 32150253 PMCID: PMC7096312 DOI: 10.1210/clinem/dgaa111] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 03/03/2020] [Indexed: 12/28/2022]
Abstract
CONTEXT Familial hypocalciuric hypercalcemia type 1 (FHH1) is caused by loss-of-function mutations of the calcium-sensing receptor (CaSR) and is considered a benign condition associated with mild-to-moderate hypercalcemia. However, the children of parents with FHH1 can develop a variety of disorders of calcium homeostasis in infancy. OBJECTIVE The objective of this work is to characterize the range of calcitropic phenotypes in the children of a mother with FHH1. METHODS A 3-generation FHH kindred was assessed by clinical, biochemical, and mutational analysis following informed consent. RESULTS The FHH kindred comprised a hypercalcemic man and his daughter who had hypercalcemia and hypocalciuria, and her 4 children, 2 of whom had asymptomatic hypercalcemia, 1 was normocalcemic, and 1 suffered from transient neonatal hypocalcemia and seizures. The hypocalcemic infant had a serum calcium of 1.57 mmol/L (6.28 mg/dL); normal, 2.0 to 2.8 mmol/L (8.0-11.2 mg/dL) and parathyroid hormone of 2.2 pmol/L; normal 1.0 to 9.3 pmol/L, and required treatment with intravenous calcium gluconate infusions. A novel heterozygous p.Ser448Pro CaSR variant was identified in the hypercalcemic individuals, but not the children with hypocalcemia or normocalcemia. Three-dimensional modeling predicted the p.Ser448Pro variant to disrupt a hydrogen bond interaction within the CaSR extracellular domain. The variant Pro448 CaSR, when expressed in HEK293 cells, significantly impaired CaSR-mediated intracellular calcium mobilization and mitogen-activated protein kinase responses following stimulation with extracellular calcium, thereby demonstrating it to represent a loss-of-function mutation. CONCLUSIONS Thus, children of a mother with FHH1 can develop hypercalcemia or transient neonatal hypocalcemia, depending on the underlying inherited CaSR mutation, and require investigations for serum calcium and CaSR mutations in early childhood.
Collapse
Affiliation(s)
- Poonam Dharmaraj
- Department of Paediatric Endocrinology, Alder Hey Children’s NHS Foundation Trust, Liverpool, UK
| | - Caroline M Gorvin
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Current Affiliation: The current affiliation of C.M.G. is Institute of Metabolism and Systems Research, University of Birmingham, and Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Astha Soni
- Department of Paediatric Endocrinology, Alder Hey Children’s NHS Foundation Trust, Liverpool, UK
| | - Nick D Nelhans
- Department of Paediatrics, Wrexham Maelor Hospital, Wrexham, UK
| | - Mie K Olesen
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Hannah Boon
- Oxford Molecular Genetics Laboratory, Churchill Hospital, Oxford, UK
| | - Treena Cranston
- Oxford Molecular Genetics Laboratory, Churchill Hospital, Oxford, UK
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Fadil M Hannan
- Nuffield Department of Women’s and Reproductive Health, University of Oxford, Oxford, UK
- Correspondence and Reprint Requests: Fadil Hannan, MBChB, DPhil, Nuffield Department of Women’s and Reproductive Health, Level 3, Women’s Centre, John Radcliffe Hospital, Oxford OX3 9DU, UK. E-mail:
| |
Collapse
|
112
|
Josephs TM, Keller AN, Khajehali E, DeBono A, Langmead CJ, Conigrave AD, Capuano B, Kufareva I, Gregory KJ, Leach K. Negative allosteric modulators of the human calcium-sensing receptor bind to overlapping and distinct sites within the 7-transmembrane domain. Br J Pharmacol 2020; 177:1917-1930. [PMID: 31881094 PMCID: PMC7070164 DOI: 10.1111/bph.14961] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Negative allosteric modulators (NAMs) that target the calcium-sensing receptor (CaS receptor) were originally developed for the treatment of osteoporosis by stimulating the release of endogenous parathyroid hormone, but failed in human clinical trials. Several chemically and structurally distinct NAM scaffolds have been described, but it is not known how these different scaffolds interact with the CaS receptor to inhibit receptor signalling in response to agonists. EXPERIMENTAL APPROACH In the present study, we used a mutagenesis approach combined with analytical pharmacology and computational modelling to probe the binding sites of four distinct NAM scaffolds. KEY RESULTS Although all four scaffolds bind to the 7-transmembrane and/or extracellular or intracellular loops, they occupy distinct regions, as previously shown for positive allosteric modulators of the CaS receptor. Furthermore, different NAM scaffolds mediate negative allosteric modulation via distinct amino acid networks. CONCLUSION AND IMPLICATIONS These findings aid our understanding of how different NAMs bind to and inhibit the CaS receptor. Elucidation of allosteric binding sites in the CaS receptor has implications for the discovery of novel allosteric modulators.
Collapse
Affiliation(s)
- Tracy M. Josephs
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| | - Andrew N. Keller
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| | - Elham Khajehali
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| | - Aaron DeBono
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| | - Christopher J. Langmead
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| | - Arthur D. Conigrave
- School of Life and Environmental SciencesUniversity of SydneySydneyNSWAustralia
| | - Ben Capuano
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| | - Irina Kufareva
- Skaggs School of Pharmacy & Pharmaceutical SciencesUniversity of CaliforniaSan DiegoCAUSA
| | - Karen J. Gregory
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| | - Katie Leach
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| |
Collapse
|
113
|
Marx SJ, Sinaii N. Neonatal Severe Hyperparathyroidism: Novel Insights From Calcium, PTH, and the CASR Gene. J Clin Endocrinol Metab 2020; 105:5645387. [PMID: 31778168 PMCID: PMC7111126 DOI: 10.1210/clinem/dgz233] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 11/26/2019] [Indexed: 12/24/2022]
Abstract
CONTEXT Neonatal severe hyperparathyroidism (NSHPT) is rare and potentially lethal. It is usually from homozygous or heterozygous germline-inactivating CASR variant(s). NSHPT shows a puzzling range of serum calcium and parathyroid hormone (PTH) levels. Optimal therapy is unclear. EVIDENCE ACQUISITION We categorized genotype/phenotype pairings related to CASRs. For the 2 pairings in NSHPT, each of 57 cases of neonatal severe hyperparathyroidism required calcium, PTH, upper normal PTH, and dosage of a germline pathogenic CASR variant. EVIDENCE SYNTHESIS Homozygous and heterozygous NSHPT are 2 among a spectrum of 9 genotype/phenotype pairings relating to CASRs and NSHPT. For the 2 NSHPT pairings, expressions differ in CASR allelic dosage, CASR variant severity, and sufficiency of maternofetal calcium fluxes. Homozygous dosage of CASR variants was generally more aggressive than heterozygous. Among heterozygotes, high-grade CASR variants in vitro were more pathogenic in vivo than low-grade variants. Fetal calcium insufficiency as from maternal hypoparathyroidism caused fetal secondary hyperparathyroidism, which persisted and was reversible in neonates. Among NSHPT pairings, calcium and PTH were higher in CASR homozygotes than in heterozygotes. Extreme hypercalcemia (above 4.5 mM; normal 2.2-2.6 mM) is a robust biomarker, occurring only in homozygotes (83% of that pairing). It could occur during the first week. CONCLUSIONS In NSHPT pairings, the homozygotes for pathogenic CASR variants show higher calcium and PTH levels than heterozygotes. Calcium levels above 4.5 mM among NSHPT are frequent and unique only to most homozygotes. This cutoff supports early and robust diagnosis of CASR dosage. Thereby, it promotes definitive total parathyroidectomy in most homozygotes.
Collapse
MESH Headings
- Biomarkers/analysis
- Calcium/blood
- Female
- Genotype
- Heterozygote
- Homozygote
- Humans
- Hyperparathyroidism, Primary/blood
- Hyperparathyroidism, Primary/diagnosis
- Hyperparathyroidism, Primary/genetics
- Infant, Newborn
- Infant, Newborn, Diseases/blood
- Infant, Newborn, Diseases/diagnosis
- Infant, Newborn, Diseases/genetics
- Male
- Mutation
- Parathyroid Hormone/blood
- Prognosis
- Receptors, Calcium-Sensing/genetics
Collapse
Affiliation(s)
- Stephen J Marx
- Office of the Scientific Director, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), Bethesda, MD
- Correspondence: Stephen Marx MD, N.I.H., Bld 6A, Room 2A-04A, MSC 0614, 6 Center Drive, Bethesda, MD 20892, USA. E-mail:
| | - Ninet Sinaii
- Biostatistics and Clinical Epidemiology Service, National Institutes of Health Clinical Center, Bethesda, MD
| |
Collapse
|
114
|
Jørgensen CV, Bräuner‐Osborne H. Pharmacology and physiological function of the orphan GPRC6A receptor. Basic Clin Pharmacol Toxicol 2020; 126 Suppl 6:77-87. [DOI: 10.1111/bcpt.13397] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 02/11/2020] [Accepted: 02/11/2020] [Indexed: 12/17/2022]
Affiliation(s)
- Christinna V. Jørgensen
- Department of Drug Design and Pharmacology Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| | - Hans Bräuner‐Osborne
- Department of Drug Design and Pharmacology Faculty of Health and Medical Sciences University of Copenhagen Copenhagen Denmark
| |
Collapse
|
115
|
Lazrak A, Yu Z, Doran S, Jian MY, Creighton J, Laube M, Garantziotis S, Prakash YS, Matalon S. Upregulation of airway smooth muscle calcium-sensing receptor by low-molecular-weight hyaluronan. Am J Physiol Lung Cell Mol Physiol 2020; 318:L459-L471. [PMID: 31913654 PMCID: PMC7099432 DOI: 10.1152/ajplung.00429.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 12/29/2019] [Accepted: 12/31/2019] [Indexed: 12/19/2022] Open
Abstract
We investigated the mechanisms involved in the development of airway hyperresponsiveness (AHR) following exposure of mice to halogens. Male mice (C57BL/6; 20-25 g) exposed to either bromine (Br2) or Cl2 (600 or 400 ppm, respectively, for 30 min) developed AHR 24 h after exposure. Nifedipine (5 mg/kg body wt; an L-type calcium channel blocker), administered subcutaneously after Br2 or Cl2 exposure, produced higher AHR compared with Br2 or Cl2 alone. In contrast, diltiazem (5 mg/kg body wt; a nondihydropyridine L-type calcium channel blocker) decreased AHR to control (air) values. Exposure of immortalized human airway smooth muscle cells (hASMC) to Br2 resulted in membrane potential depolarization (Vm Air: 62 ± 3 mV; 3 h post Br2:-45 ± 5 mV; means ± 1 SE; P < 0.001), increased intracellular [Ca2+]i, and increased expression of the calcium-sensing receptor (Ca-SR) protein. Treatment of hASMC with a siRNA against Ca-SR significantly inhibited the Br2 and nifedipine-induced Vm depolarization and [Ca2+]i increase. Intranasal administration of an antagonist to Ca-SR in mice postexposure to Br2 reversed the effects of Br2 and nifedipine on AHR. Incubation of hASMC with low-molecular-weight hyaluronan (LMW-HA), generated by exposing high-molecular-weight hyaluronan (HMW-HA) to Br2, caused Vm depolarization, [Ca2+]i increase, and Ca-SR expression to a similar extent as exposure to Br2 and Cl2. The addition of HMW-HA to cells or mice exposed to Br2, Cl2, or LMW-HA reversed these effects in vitro and improved AHR in vivo. We conclude that detrimental effects of halogen exposure on AHR are mediated via activation of the Ca-SR by LMW-HA.
Collapse
Affiliation(s)
- Ahmed Lazrak
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine & Pulmonary Injury Repair Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Zhihong Yu
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine & Pulmonary Injury Repair Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Stephen Doran
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine & Pulmonary Injury Repair Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Ming-Yuan Jian
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine & Pulmonary Injury Repair Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Judy Creighton
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine & Pulmonary Injury Repair Center, University of Alabama at Birmingham, Birmingham, Alabama
| | - Mandy Laube
- Department of Pediatrics, Division of Neonatology, Leipzig University, Leipzig, Germany
| | - Stavros Garantziotis
- Matrix Biology Group, Immunity, Inflammation, and Disease Laboratory, National Institutes of Health/National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina
| | - Y S Prakash
- Department of Physiology and Biomedical Engineering and Anesthesiology, Mayo Clinic Alix School of Medicine and Science, Rochester, Minnesota
| | - Sadis Matalon
- Department of Anesthesiology and Perioperative Medicine, Division of Molecular and Translational Biomedicine & Pulmonary Injury Repair Center, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
116
|
Wilkens MR, Schnepel N, Muscher-Banse AS. Dietary protein and calcium modulate parathyroid vitamin D receptor expression in young ruminants. J Steroid Biochem Mol Biol 2020; 196:105503. [PMID: 31648052 DOI: 10.1016/j.jsbmb.2019.105503] [Citation(s) in RCA: 2] [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: 07/15/2019] [Revised: 09/20/2019] [Accepted: 10/17/2019] [Indexed: 10/25/2022]
Abstract
For economic reasons and in order to minimize nitrogen excretion and thus pollution, the crude protein content in the diet of livestock animals should be as low as possible without negatively affecting the animals´ health and performance. As ruminants can efficiently use dietary protein because of the ruminohepatic circulation of urea, they are considered to cope more easily with such a feeding regime than monogastric animals. However, despite unaltered daily weight gain, massive changes in mineral homeostasis and vitamin D metabolism were observed with dietary protein reduction (N-) in young, growing goats. Serum concentrations of 1,25-dihydroxyvitamin D3 (1,25-(OH)2D3) were decreased with a low N intake, even if calcium (Ca) was also restricted (Ca-). Interestingly, concentrations of cyclic adenosine monophosphate (cAMP) measured as an indirect assessment for the parathyroid hormone (PTH) activity were not affected by low protein. Therefore, it was hypothesized that the sensitivity of the parathyroid gland is modulated during these dietary interventions. Four groups of male German colored goats received a control (N+/Ca+), a reduced protein (N-/Ca+), a reduced Ca (N+/Ca-) or a reduced protein and Ca (N-/Ca-) diet. After six weeks we determined the expression of PTH, PTH receptor, Ca sensing receptor (CASR), vitamin D receptor (VDR), retinoid X receptor (RXRα), Klotho, fibroblast growth factor receptor 1c-splicing form, and the sodium-dependent Pi transporter (PiT1) in the parathyroid glands. Concentrations of cAMP were not affected, while those of Ca and 1,25-(OH)2D3were diminished and that of 25-hydroxyvitamin D3 was increased with N- feeding. The expression patterns of the described target genes were not altered. In contrast, animals fed the Ca- rations showed enhanced serum 1,25-(OH)2D3 and cAMP levels with no changes in blood Ca concentrations demonstrating an efficient adaptation. The mRNA expression of expression of VDR and CASR in the parathyroid gland was significantly diminished and RXRα, PTHR and PiT1 expression was elevated. Instead of the assumed desensitization of the parathyroid gland with N-, our results indicate elevated responsiveness to decreased blood Ca with feeding Ca-.
Collapse
Affiliation(s)
- Mirja R Wilkens
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173 Hannover, Germany
| | - Nadine Schnepel
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173 Hannover, Germany
| | - Alexandra S Muscher-Banse
- Institute for Physiology and Cell Biology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173 Hannover, Germany.
| |
Collapse
|
117
|
Gregory KJ, Giraldo J, Diao J, Christopoulos A, Leach K. Evaluation of Operational Models of Agonism and Allosterism at Receptors with Multiple Orthosteric Binding Sites. Mol Pharmacol 2020; 97:35-45. [PMID: 31704718 DOI: 10.1124/mol.119.118091] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/04/2019] [Indexed: 02/14/2025] Open
Abstract
Current operational models of agonism and allosterism quantify ligand actions at receptors where agonist concentration-response relationships are nonhyperbolic by introduction of a transducer slope that relates receptor occupancy to response. However, for some receptors nonhyperbolic concentration-response relationships arise from multiple endogenous agonist molecules binding to a receptor in a cooperative manner. Thus, we developed operational models of agonism in systems with cooperative agonist binding and evaluated the models by simulating data describing agonist effects. The models were validated by analyzing experimental data demonstrating the effects of agonists and allosteric modulators at receptors where agonist binding follows hyperbolic (M4 muscarinic acetylcholine receptors) or nonhyperbolic relationships (metabotropic glutamate receptor 5 and calcium-sensing receptor). For hyperbolic agonist concentration-response relationships, no differences in estimates of ligand affinity, efficacy, or cooperativity were observed when the slope was assigned to either a transducer slope or agonist binding slope. In contrast, for receptors with nonhyperbolic agonist concentration-response relationships, estimates of ligand affinity, efficacy, or cooperativity varied depending on the assignment of the slope. The extent of this variation depended on the magnitude of the slope value and agonist efficacy, and for allosteric modulators on the magnitude of cooperativity. The modified operational models described herein are well suited to analyzing agonist and modulator interactions at receptors that bind multiple orthosteric agonists in a cooperative manner. Accounting for cooperative agonist binding is essential to accurately quantify agonist and drug actions. SIGNIFICANCE STATEMENT: Some orthosteric agonists bind to multiple sites on a receptor, but current analytical methods to characterize such interactions are limited. Herein, we develop and validate operational models of agonism and allosterism for receptors with multiple orthosteric binding sites, and demonstrate that such models are essential to accurately quantify agonist and drug actions. These findings have important implications for the discovery and development of drugs targeting receptors such as the calcium-sensing receptor, which binds at least five calcium ions.
Collapse
MESH Headings
- Allosteric Regulation/drug effects
- Binding Sites/drug effects
- Calcium/metabolism
- Calcium Ionophores/pharmacology
- Computer Simulation
- Dose-Response Relationship, Drug
- Drug Agonism
- Drug Synergism
- HEK293 Cells
- Humans
- Ligands
- Models, Biological
- Receptor, Metabotropic Glutamate 5/agonists
- Receptor, Metabotropic Glutamate 5/chemistry
- Receptor, Metabotropic Glutamate 5/metabolism
- Receptor, Muscarinic M4/agonists
- Receptor, Muscarinic M4/chemistry
- Receptor, Muscarinic M4/metabolism
- Receptors, Calcium-Sensing/agonists
- Receptors, Calcium-Sensing/chemistry
- Receptors, Calcium-Sensing/metabolism
Collapse
Affiliation(s)
- Karen J Gregory
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Australia (K.J.G., J.D., A.C., K.L.); Laboratory of Molecular Neuropharmacology and Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain (J.G.); Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, Bellaterra, Spain (J.G.); and Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí and Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain (J.G.)
| | - Jesús Giraldo
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Australia (K.J.G., J.D., A.C., K.L.); Laboratory of Molecular Neuropharmacology and Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain (J.G.); Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, Bellaterra, Spain (J.G.); and Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí and Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain (J.G.)
| | - Jiayin Diao
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Australia (K.J.G., J.D., A.C., K.L.); Laboratory of Molecular Neuropharmacology and Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain (J.G.); Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, Bellaterra, Spain (J.G.); and Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí and Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain (J.G.)
| | - Arthur Christopoulos
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Australia (K.J.G., J.D., A.C., K.L.); Laboratory of Molecular Neuropharmacology and Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain (J.G.); Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, Bellaterra, Spain (J.G.); and Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí and Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain (J.G.)
| | - Katie Leach
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Australia (K.J.G., J.D., A.C., K.L.); Laboratory of Molecular Neuropharmacology and Bioinformatics, Institut de Neurociències and Unitat de Bioestadística, Facultat de Medicina, Universitat Autònoma de Barcelona, Barcelona, Spain (J.G.); Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental, Bellaterra, Spain (J.G.); and Unitat de Neurociència Traslacional, Parc Taulí Hospital Universitari, Institut d'Investigació i Innovació Parc Taulí and Institut de Neurociències, Universitat Autònoma de Barcelona, Bellaterra, Spain (J.G.)
| |
Collapse
|
118
|
Perez-Aguilar JM, Kang SG, Zhang L, Zhou R. Modeling and Structural Characterization of the Sweet Taste Receptor Heterodimer. ACS Chem Neurosci 2019; 10:4579-4592. [PMID: 31553164 DOI: 10.1021/acschemneuro.9b00438] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Sweet taste receptor, a heterodimer belonging to the class C G-protein coupled receptor (GPCR) family and composed of the T1R2 and T1R3 subunits, is responsible for the perception of natural sugars, sweet proteins, various d-amino acids, as well as artificial sweeteners. Despite the critical importance of the sweet receptor not only in mediating gustation but also in its role in the food industry, the architecture of the T1R2-T1R3 complex and the mechanism by which extracellular stimuli induce conformational changes that are propagated to the intracellular milieu, i.e., the signal transduction pathway, remain largely unknown. Here, we constructed and characterized a full-length structural model of the T1R2-T1R3 receptor, including both the transmembrane (TM) and extracellular (EC) domains of the heterodimer, using comparative modeling and extensive all-atom molecular dynamics simulations. Several heterodimer interfaces were first examined for the TM domain, and conformational changes occurring at the intracellular side and associated with the receptor's activation were characterized. From the analysis on the simulated data, putative allosteric binding sites for ligands, ions, and cholesterol were proposed. Also, insights into the protein interface of the TM domain upon activation are provided. The EC domain of the heterodimer, including both the Venus flytrap and cysteine-rich domains, was also investigated. Several important intersubunit interactions located at regions responsible for the receptor's proper function were observed, which resemble those recently identified in other class C GPCR members. Integration of the results from the TM and EC domains facilitates the generation of a full-length T1R2-T1R3 receptor. These findings along with the full-length structural model of the T1R2-T1R3 receptor provide a structural framework that may assist in understanding the mechanistic details associated with the receptor activation process for the sweet T1R2-T1R3 receptor as well as other members of the same family.
Collapse
Affiliation(s)
- Jose Manuel Perez-Aguilar
- IBM Thomas J. Watson Research Center, Yorktown Heights, New York 10598, United States
- School of Chemical Sciences, Meritorious Autonomous University of Puebla (BUAP), University City, Puebla 72570, Mexico
| | - Seung-gu Kang
- IBM Thomas J. Watson Research Center, Yorktown Heights, New York 10598, United States
| | - Leili Zhang
- IBM Thomas J. Watson Research Center, Yorktown Heights, New York 10598, United States
| | - Ruhong Zhou
- IBM Thomas J. Watson Research Center, Yorktown Heights, New York 10598, United States
| |
Collapse
|
119
|
Fonin AV, Darling AL, Kuznetsova IM, Turoverov KK, Uversky VN. Multi-functionality of proteins involved in GPCR and G protein signaling: making sense of structure-function continuum with intrinsic disorder-based proteoforms. Cell Mol Life Sci 2019; 76:4461-4492. [PMID: 31428838 PMCID: PMC11105632 DOI: 10.1007/s00018-019-03276-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 08/05/2019] [Accepted: 08/12/2019] [Indexed: 12/21/2022]
Abstract
GPCR-G protein signaling system recognizes a multitude of extracellular ligands and triggers a variety of intracellular signaling cascades in response. In humans, this system includes more than 800 various GPCRs and a large set of heterotrimeric G proteins. Complexity of this system goes far beyond a multitude of pair-wise ligand-GPCR and GPCR-G protein interactions. In fact, one GPCR can recognize more than one extracellular signal and interact with more than one G protein. Furthermore, one ligand can activate more than one GPCR, and multiple GPCRs can couple to the same G protein. This defines an intricate multifunctionality of this important signaling system. Here, we show that the multifunctionality of GPCR-G protein system represents an illustrative example of the protein structure-function continuum, where structures of the involved proteins represent a complex mosaic of differently folded regions (foldons, non-foldons, unfoldons, semi-foldons, and inducible foldons). The functionality of resulting highly dynamic conformational ensembles is fine-tuned by various post-translational modifications and alternative splicing, and such ensembles can undergo dramatic changes at interaction with their specific partners. In other words, GPCRs and G proteins exist as sets of conformational/basic, inducible/modified, and functioning proteoforms characterized by a broad spectrum of structural features and possessing various functional potentials.
Collapse
Affiliation(s)
- Alexander V Fonin
- Laboratory of structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russian Federation
| | - April L Darling
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Irina M Kuznetsova
- Laboratory of structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russian Federation
| | - Konstantin K Turoverov
- Laboratory of structural Dynamics, Stability and Folding of Proteins, Institute of Cytology, Russian Academy of Sciences, St. Petersburg, 194064, Russian Federation
- Department of Biophysics, Peter the Great St. Petersburg Polytechnic University, Polytechnicheskaya av. 29, St. Petersburg, 195251, Russian Federation
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
- Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino, Moscow, Russian Federation.
| |
Collapse
|
120
|
Dal Prà I, Armato U, Chiarini A. Family C G-Protein-Coupled Receptors in Alzheimer's Disease and Therapeutic Implications. Front Pharmacol 2019; 10:1282. [PMID: 31719824 PMCID: PMC6826475 DOI: 10.3389/fphar.2019.01282] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 10/07/2019] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD), particularly its sporadic or late-onset form (SAD/LOAD), is the most prevalent (96–98% of cases) neurodegenerative dementia in aged people. AD’s neuropathology hallmarks are intrabrain accumulation of amyloid-β peptides (Aβs) and of hyperphosphorylated Tau (p-Tau) proteins, diffuse neuroinflammation, and progressive death of neurons and oligodendrocytes. Mounting evidences suggest that family C G-protein-coupled receptors (GPCRs), which include γ-aminobutyric acid B receptors (GABABRs), metabotropic glutamate receptors (mGluR1-8), and the calcium-sensing receptor (CaSR), are involved in many neurotransmitter systems that dysfunction in AD. This review updates the available knowledge about the roles of GPCRs, particularly but not exclusively those expressed by brain astrocytes, in SAD/LOAD onset and progression, taking stock of their respective mechanisms of action and of their potential as anti-AD therapeutic targets. In particular, GABABRs prevent Aβs synthesis and neuronal hyperexcitability and group I mGluRs play important pathogenetic roles in transgenic AD-model animals. Moreover, the specific binding of Aβs to the CaSRs of human cortical astrocytes and neurons cultured in vitro engenders a pathological signaling that crucially promotes the surplus synthesis and release of Aβs and hyperphosphorylated Tau proteins, and also of nitric oxide, vascular endothelial growth factor-A, and proinflammatory agents. Concurrently, Aβs•CaSR signaling hinders the release of soluble (s)APP-α peptide, a neurotrophic agent and GABABR1a agonist. Altogether these effects progressively kill human cortical neurons in vitro and likely also in vivo. Several CaSR’s negative allosteric modulators suppress all the noxious effects elicited by Aβs•CaSR signaling in human cortical astrocytes and neurons thus safeguarding neurons’ viability in vitro and raising hopes about their potential therapeutic benefits in AD patients. Further basic and clinical investigations on these hot topics are needed taking always heed that activation of the several brain family C GPCRs may elicit divergent upshots according to the models studied.
Collapse
Affiliation(s)
- Ilaria Dal Prà
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| | - Ubaldo Armato
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| | - Anna Chiarini
- Human Histology and Embryology Unit, University of Verona Medical School, Verona, Italy
| |
Collapse
|
121
|
Centeno PP, Herberger A, Mun HC, Tu C, Nemeth EF, Chang W, Conigrave AD, Ward DT. Phosphate acts directly on the calcium-sensing receptor to stimulate parathyroid hormone secretion. Nat Commun 2019; 10:4693. [PMID: 31619668 PMCID: PMC6795806 DOI: 10.1038/s41467-019-12399-9] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 09/05/2019] [Indexed: 02/06/2023] Open
Abstract
Extracellular phosphate regulates its own renal excretion by eliciting concentration-dependent secretion of parathyroid hormone (PTH). However, the phosphate-sensing mechanism remains unknown and requires elucidation for understanding the aetiology of secondary hyperparathyroidism in chronic kidney disease (CKD). The calcium-sensing receptor (CaSR) is the main controller of PTH secretion and here we show that raising phosphate concentration within the pathophysiologic range for CKD significantly inhibits CaSR activity via non-competitive antagonism. Mutation of residue R62 in anion binding site-1 abolishes phosphate-induced inhibition of CaSR. Further, pathophysiologic phosphate concentrations elicit rapid and reversible increases in PTH secretion from freshly-isolated human parathyroid cells consistent with a receptor-mediated action. The same effect is seen in wild-type murine parathyroid glands, but not in CaSR knockout glands. By sensing moderate changes in extracellular phosphate concentration, the CaSR represents a phosphate sensor in the parathyroid gland, explaining the stimulatory effect of phosphate on PTH secretion. Elevated inorganic phosphate levels promote excessive parathyroid hormone secretion, which contributes to the aetiology of secondary hyperparathyroidism. Here, the authors show that phosphate directly inhibits the calcium-sensing receptor, the main regulator of parathyroid hormone secretion.
Collapse
Affiliation(s)
- Patricia P Centeno
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Amanda Herberger
- UCSF Department of Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Hee-Chang Mun
- Charles Perkins Centre, University of Sydney, School of Life and Environmental Sciences, Sydney, NSW, Australia
| | - Chialing Tu
- UCSF Department of Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Edward F Nemeth
- MetisMedica, 13 Poplar Plains Road, Toronto, ON, M4V 2M7, Canada
| | - Wenhan Chang
- UCSF Department of Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Arthur D Conigrave
- Charles Perkins Centre, University of Sydney, School of Life and Environmental Sciences, Sydney, NSW, Australia
| | - Donald T Ward
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
| |
Collapse
|
122
|
Mos I, Jacobsen SE, Foster SR, Bräuner-Osborne H. Calcium-Sensing Receptor Internalization Is β-Arrestin-Dependent and Modulated by Allosteric Ligands. Mol Pharmacol 2019; 96:463-474. [PMID: 31399503 DOI: 10.1124/mol.119.116772] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 07/30/2019] [Indexed: 02/14/2025] Open
Abstract
G protein-coupled receptor (GPCR) internalization is crucial for the termination of GPCR activity, and in some cases is associated with G protein-independent signaling and endosomal receptor signaling. To date, internalization has been studied in great detail for class A GPCRs; whereas it is not well established to what extent the observations can be generalized to class C GPCRs, including the extracellular calcium-sensing receptor (CaSR). The CaSR is a prototypical class C GPCR that maintains stable blood calcium (Ca2+) levels by sensing minute changes in extracellular free Ca2+ It is thus necessary that the activity of the CaSR is tightly regulated, even while continuously being exposed to its endogenous agonist. Previous studies have used overexpression of intracellular proteins involved in GPCR trafficking, pathway inhibitors, and cell-surface expression or functional desensitization as indirect measures to investigate CaSR internalization. However, there is no general consensus on the processes involved, and the mechanism of CaSR internalization remains poorly understood. The current study provides new insights into the internalization mechanism of the CaSR. We have used a state-of-the-art time-resolved fluorescence resonance energy transfer-based internalization assay to directly measure CaSR internalization in real-time. We demonstrate that the CaSR displays both constitutive and concentration-dependent Ca2+-mediated internalization. For the first time, we conclusively show that CaSR internalization is sensitive to immediate positive and negative modulation by the CaSR-specific allosteric modulators N-(3-[2-chlorophenyl]propyl)-(R)-α-methyl-3-methoxybenzylamine (NPS R-568) and 2-chloro-6-[(2R)-2-hydroxy-3-[(2-methyl-1-naphthalen-2-ylpropan-2-yl)amino]propoxy]benzonitrile (NPS 2143), respectively. In addition, we provide compelling evidence that CaSR internalization is β-arrestin-dependent while interestingly being largely independent of Gq/11 and Gi/o protein signaling. SIGNIFICANCE STATEMENT: A novel highly efficient cell-based real-time internalization assay to show that calcium-sensing receptor (CaSR) internalization is β-arrestin-dependent and sensitive to modulation by allosteric ligands.
Collapse
Affiliation(s)
- Iris Mos
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stine E Jacobsen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simon R Foster
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans Bräuner-Osborne
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
123
|
Binmahfouz LS, Centeno PP, Conigrave AD, Ward DT. Identification of Serine-875 as an Inhibitory Phosphorylation Site in the Calcium-Sensing Receptor. Mol Pharmacol 2019; 96:204-211. [PMID: 31189667 DOI: 10.1124/mol.119.116178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/20/2019] [Indexed: 02/14/2025] Open
Abstract
The calcium-sensing receptor (CaS) is the principal controller of extracellular calcium (Ca2+ o) homeostasis and is inhibited in vitro and in vivo by protein kinase C (PKC)-mediated phosphorylation at CaST888 However, PKC inhibition enhances signaling even in CaSs lacking Thr-888, suggesting that an additional inhibitory site exists. An apparently equivalent PKC regulatory site in metabotropic glutamate receptor 5 (Ser-839) aligns not with CaST888 but instead with CaSS875, which was not previously considered to be a PKC site. CaSS875A (nonphosphorylatable) exhibited significantly enhanced Ca2+ o sensitivity of both intracellular Ca2+ mobilization and extracellular signal-regulated kinase 1/2 activation, whereas the phosphomimetic CaSS875D mutant exhibited a loss of function. The CaSS875A/T888A double mutant exhibited even greater Ca2+ o sensitivity than CaST888A alone, a response no longer enhanced by PKC inhibition. Finally, when expressed in CaS lacking its extracellular domain, the CaSS875A/T888A double mutation elicited maximal activation even under control conditions, but remained sensitive to negative allosteric modulation [N-(2-hydroxy-3-(2-cyano-3-chlorophenoxy)propyl)-1,1-dimethyl-2-(2-nephthyl)ethylamine] or Ca2+ o removal. Therefore, we have now identified CaSS875 as the missing PKC phosphorylation site that, together with CaST888, shapes the CaS signaling that underpins Ca2+ o homeostasis. Together with the inactive form of the CaS extracellular domain, these sites attenuate Ca2+ o sensitivity to attain appropriate physiologic Ca2+ o sensing. SIGNIFICANCE STATEMENT: Serine-875 represents the missing inhibitory PKC phosphorlyation site in CaS that in tandem with Thr-888 controls receptor activity.
Collapse
Affiliation(s)
- Lenah S Binmahfouz
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (L.S.B., P.P.C., D.T.W.); King Abdulaziz University, Jeddah, Saudi Arabia (L.S.B.); and Charles Perkins Center, School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia (A.D.C.)
| | - Patricia P Centeno
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (L.S.B., P.P.C., D.T.W.); King Abdulaziz University, Jeddah, Saudi Arabia (L.S.B.); and Charles Perkins Center, School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia (A.D.C.)
| | - Arthur D Conigrave
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (L.S.B., P.P.C., D.T.W.); King Abdulaziz University, Jeddah, Saudi Arabia (L.S.B.); and Charles Perkins Center, School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia (A.D.C.)
| | - Donald T Ward
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (L.S.B., P.P.C., D.T.W.); King Abdulaziz University, Jeddah, Saudi Arabia (L.S.B.); and Charles Perkins Center, School of Life and Environmental Sciences, University of Sydney, Camperdown, New South Wales, Australia (A.D.C.)
| |
Collapse
|
124
|
The Nervous System Relevance of the Calcium Sensing Receptor in Health and Disease. Molecules 2019; 24:molecules24142546. [PMID: 31336912 PMCID: PMC6680999 DOI: 10.3390/molecules24142546] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 07/05/2019] [Accepted: 07/10/2019] [Indexed: 02/08/2023] Open
Abstract
The calcium sensing receptor (CaSR) was first identified in parathyroid glands, and its primary role in controlling systemic calcium homeostasis by the regulation of parathyroid hormone (PTH) secretion has been extensively described in literature. Additionally, the receptor has also been investigated in cells and tissues not directly involved in calcium homeostasis, e.g., the nervous system (NS), where it plays crucial roles in early neural development for the differentiation of neurons and glial cells, as well as in the adult nervous system for synaptic transmission and plasticity. Advances in the knowledge of the CaSR's function in such physiological processes have encouraged researchers to further broaden the receptor's investigation in the neuro-pathological conditions of the NS. Interestingly, pre-clinical data suggest that receptor inhibition by calcilytics might be effective in counteracting the pathomechanism underlying Alzheimer's disease and ischemia, while a CaSR positive modulation with calcimimetics has been proposed as a potential approach for treating neuroblastoma. Importantly, such promising findings led to the repurposing of CaSR modulators as novel pharmacological alternatives for these disorders. Therefore, the aim of this review article is to critically appraise evidence which, so far, has been yielded from the investigation of the role of the CaSR in physiology of the nervous system and to focus on the most recent emerging concepts which have reported the receptor as a therapeutic target for neurodegeneration and neuroblastic tumors.
Collapse
|
125
|
Mun HC, Leach KM, Conigrave AD. L-Amino Acids Promote Calcitonin Release via a Calcium-Sensing Receptor: Gq/11-Mediated Pathway in Human C-Cells. Endocrinology 2019; 160:1590-1599. [PMID: 31127815 DOI: 10.1210/en.2018-00860] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 04/19/2019] [Indexed: 11/19/2022]
Abstract
Human calcitonin release is promoted by elevated extracellular Ca2+ (Ca2+o) concentration acting, at least in part, via the calcium-sensing receptor (CaSR). The CaSR is positively modulated by L-amino acids, including the aromatic amino acids L-phenylalanine (Phe) and L-tryptophan (Trp). To investigate the effect of L-amino acids on human calcitonin secretion, we selected thyroid TT cells and exposed them to various Ca2+o concentrations in the absence or presence of L-Phe, plasma-like mixtures of L-amino acids, or the clinically effective positive modulator (calcimimetic) cinacalcet. In the presence of L-Phe or plasma-like mixtures of amino acids, TT cells exhibited enhanced Ca2+o sensitivity in assays of calcitonin release and intracellular Ca2+ mobilization. Furthermore, the effect of elevated Ca2+o and L-Phe on calcitonin release was markedly suppressed by the calcilytic NPS-2143. These effects were dependent on CaSR-mediated activation of Gq/11 as revealed by the specific inhibitor YM-254890. The findings support the hypothesis that calcitonin release is stimulated by increases in plasma L-amino acid levels as well as elevated Ca2+o concentration. They also demonstrate that stimulated calcitonin release as well as basal levels of calcitonin secretion are mediated by a CaSR:Gq/11 signaling mechanism.
Collapse
Affiliation(s)
- Hee-Chang Mun
- School of Life and Environmental Sciences, Charles Perkins Centre (D17), University of Sydney, Sydney, New South Wales, Australia
| | - Katie M Leach
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Parkville, Victoria, Australia
| | - Arthur D Conigrave
- School of Life and Environmental Sciences, Charles Perkins Centre (D17), University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
126
|
Nordholm A, Egstrand S, Gravesen E, Mace ML, Morevati M, Olgaard K, Lewin E. Circadian rhythm of activin A and related parameters of mineral metabolism in normal and uremic rats. Pflugers Arch 2019; 471:1079-1094. [PMID: 31236663 PMCID: PMC6614158 DOI: 10.1007/s00424-019-02291-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022]
Abstract
Activin A is a new fascinating player in chronic kidney disease-mineral and bone disorder (CKD-MBD), which is implicated in progressive renal disease, vascular calcification, and osteodystrophy. Plasma activin A rises early in the progression of renal disease. Disruption of circadian rhythms is related to increased risk of several diseases and circadian rhythms are observed in mineral homeostasis, bone parameters, and plasma levels of phosphate and PTH. Therefore, we examined the circadian rhythm of activin A and CKD-MBD-related parameters (phosphate, PTH, FGF23, and klotho) in healthy controls and CKD rats (5/6 nephrectomy) on high-, standard- and low-dietary phosphate contents as well as during fasting conditions. Plasma activin A exhibited circadian rhythmicity in healthy control rats with fourfold higher values at acrophase compared with nadir. The rhythm was obliterated in CKD. Activin A was higher in CKD rats compared with controls when measured at daytime but not significantly when measured at evening/nighttime, stressing the importance of time-specific reference intervals when interpreting plasma values. Plasma phosphate, PTH, and FGF23 all showed circadian rhythms in control rats, which were abolished or disrupted in CKD. Plasma klotho did not show circadian rhythm. Thus, the present investigation shows, for the first time, circadian rhythm of plasma activin A. The rhythmicity is severely disturbed by CKD and is associated with disturbed rhythms of phosphate and phosphate-regulating hormones PTH and FGF23, indicating that disturbed circadian rhythmicity is an important feature of CKD-MBD.
Collapse
Affiliation(s)
- Anders Nordholm
- Nephrological Department, Herlev Hospital, University of Copenhagen, 2730, Herlev, Denmark.,Nephrological Department, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Søren Egstrand
- Nephrological Department, Herlev Hospital, University of Copenhagen, 2730, Herlev, Denmark.,Nephrological Department, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Eva Gravesen
- Nephrological Department, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Maria L Mace
- Nephrological Department, Herlev Hospital, University of Copenhagen, 2730, Herlev, Denmark.,Nephrological Department, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Marya Morevati
- Nephrological Department, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Klaus Olgaard
- Nephrological Department, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Ewa Lewin
- Nephrological Department, Herlev Hospital, University of Copenhagen, 2730, Herlev, Denmark. .,Nephrological Department, Rigshospitalet, University of Copenhagen, 2100, Copenhagen, Denmark.
| |
Collapse
|
127
|
Zhao X, Schindell B, Li W, Ni L, Liu S, Wijerathne CUB, Gong J, Nyachoti CM, O K, Yang C. Distribution and localization of porcine calcium sensing receptor in different tissues of weaned piglets1. J Anim Sci 2019; 97:2402-2413. [PMID: 30887022 PMCID: PMC6541828 DOI: 10.1093/jas/skz096] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 03/18/2019] [Indexed: 02/06/2023] Open
Abstract
Taste receptors including calcium sensing receptor (CaSR) are expressed in various animal tissues, and CaSR plays important roles in nutrient sensing and the physiology, growth, and development of animals. However, molecular distribution of porcine CaSR (pCaSR) in different tissues, especially along the longitudinal axis of the digestive tract in weaned piglets, is still unknown. In the present study, we investigated the distribution and localization of pCaSR in the different tissues including intestinal segments of weaned piglets. Six male pigs were anesthetized and euthanized. Different tissues such as intestinal segments were collected. The pCaSR mRNA abundance, protein abundance, and localization were measured by real-time PCR, Western blotting, and immunohistochemistry, respectively. The mRNA and protein of pCaSR were detected in the kidney, lung, liver, stomach, duodenum, jejunum, ileum, and colon. The pCaSR mRNA was much higher (five to 180 times) in the kidney when compared with other tissues (P < 0.05). The ileum had higher pCaSR mRNA and protein abundances than the stomach, duodenum, jejunum, and colon (P < 0.05). Immunohistochemical staining results indicated that the pCaSR protein was mostly located in the epithelia of the stomach, duodenum, jejunum, ileum, and colon. These results demonstrate that pCaSR is widely expressed in different tissues including intestinal segments in weaned piglets and the ileum has a higher expression level of pCaSR. Further research is needed to confirm the expression of CaSR in the different types of epithelial cells isolated from weaned piglets and characterize the functions of pCaSR, its potential ligands and cell signaling pathways related to CaSR activation in enteroendocrine cells and potentially in enterocytes.
Collapse
Affiliation(s)
- Xiaoya Zhao
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| | - Brayden Schindell
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| | - Weiqi Li
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| | - Liju Ni
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
- Shanghai Lab-Animal Research Center, Shanghai, China
| | - Shangxi Liu
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| | - Charith U B Wijerathne
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
- CCARM, St. Boniface Hospital Research Centre, Winnipeg, MB, Canada
| | - Joshua Gong
- Guelph Research and Development Centre, Agriculture Agri-Food Canada, Guelph, ON, Canada
| | - C Martin Nyachoti
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| | - Karmin O
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
- CCARM, St. Boniface Hospital Research Centre, Winnipeg, MB, Canada
| | - Chengbo Yang
- Department of Animal Science, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
128
|
Seyedabadi M, Ghahremani MH, Albert PR. Biased signaling of G protein coupled receptors (GPCRs): Molecular determinants of GPCR/transducer selectivity and therapeutic potential. Pharmacol Ther 2019; 200:148-178. [PMID: 31075355 DOI: 10.1016/j.pharmthera.2019.05.006] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023]
Abstract
G protein coupled receptors (GPCRs) convey signals across membranes via interaction with G proteins. Originally, an individual GPCR was thought to signal through one G protein family, comprising cognate G proteins that mediate canonical receptor signaling. However, several deviations from canonical signaling pathways for GPCRs have been described. It is now clear that GPCRs can engage with multiple G proteins and the line between cognate and non-cognate signaling is increasingly blurred. Furthermore, GPCRs couple to non-G protein transducers, including β-arrestins or other scaffold proteins, to initiate additional signaling cascades. Receptor/transducer selectivity is dictated by agonist-induced receptor conformations as well as by collateral factors. In particular, ligands stabilize distinct receptor conformations to preferentially activate certain pathways, designated 'biased signaling'. In this regard, receptor sequence alignment and mutagenesis have helped to identify key receptor domains for receptor/transducer specificity. Furthermore, molecular structures of GPCRs bound to different ligands or transducers have provided detailed insights into mechanisms of coupling selectivity. However, receptor dimerization, compartmentalization, and trafficking, receptor-transducer-effector stoichiometry, and ligand residence and exposure times can each affect GPCR coupling. Extrinsic factors including cell type or assay conditions can also influence receptor signaling. Understanding these factors may lead to the development of improved biased ligands with the potential to enhance therapeutic benefit, while minimizing adverse effects. In this review, evidence for ligand-specific GPCR signaling toward different transducers or pathways is elaborated. Furthermore, molecular determinants of biased signaling toward these pathways and relevant examples of the potential clinical benefits and pitfalls of biased ligands are discussed.
Collapse
Affiliation(s)
- Mohammad Seyedabadi
- Department of Pharmacology, School of Medicine, Bushehr University of Medical Sciences, Iran; Education Development Center, Bushehr University of Medical Sciences, Iran
| | | | - Paul R Albert
- Ottawa Hospital Research Institute, Neuroscience, University of Ottawa, Canada.
| |
Collapse
|
129
|
Kashani-Amin E, Sakhteman A, Larijani B, Ebrahim-Habibi A. Introducing a New Model of Sweet Taste Receptor, a Class C G-protein Coupled Receptor (C GPCR). Cell Biochem Biophys 2019; 77:227-243. [PMID: 31069640 DOI: 10.1007/s12013-019-00872-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Accepted: 04/27/2019] [Indexed: 12/31/2022]
Abstract
The structure of sweet taste receptor (STR), a heterodimer of class C G-protein coupled receptors comprising T1R2 and T1R3 molecules, is still undetermined. In this study, a new enhanced model of the receptor is introduced based on the most recent templates. The improvement, stability, and reliability of the model are discussed in details. Each domain of the protein, i.e., VFTM, CR, and TMD, were separately constructed by hybrid-model construction methods and then assembled to build whole monomers. Overall, 680 ns molecular dynamics simulation was performed for the individual domains, the whole monomers and the heterodimer form of the VFTM orthosteric binding site. The latter's structure obtained from 200 ns simulation was docked with aspartame; among various binding sites suggested by FTMAP server, the experimentally suggested binding domain in T1R2 was retrieved. Local three-dimensional structures and helices spans were evaluated and showed acceptable accordance with the template structures and secondary structure predictions. Individual domains and whole monomer structures were found stable and reliable to be used. In conclusion, several validations have shown reliability of the new and enhanced models for further molecular modeling studies on structure and function of STR and C GPCRs.
Collapse
Affiliation(s)
- Elaheh Kashani-Amin
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sakhteman
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.,Medicinal Chemistry and Natural Products Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadeh Ebrahim-Habibi
- Biosensor Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
130
|
Lei Q, Lin D, Huang WX, Wu D, Chen J. [Effects of calcium ion on the migration and osteogenic differentiation of human osteoblasts]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2019; 36:602-608. [PMID: 30593103 DOI: 10.7518/hxkq.2018.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
OBJECTIVE This study aimed to investigate the effect of calcium ion (Ca²⁺) on the migration and osteogenic differentiation of human osteoblasts and explore the proper concentration and correlation mechanism. METHODS A series of Ca²⁺ solutions with different concentrations was prepared. Osteoblast migration was assessed by Transwell assay, and proliferation was studied via the CCK-8 colorimetric assay. The mRNA expression of osteogenic genes was examined via reverse transcription-polymerase chain reaction (RT-PCR), and the mineralized nodule was examined by alizarin red-S method. After calcium sensitive receptor (CaSR) antagonism, Ca²⁺-induced migration and osteogenic differentiation were analyzed. RESULTS In the migration experiment, 2, 4, and 6 mmol·L⁻¹ Ca²⁺ could promoted osteoblast migration at three timepoints (8, 16, and 24 h), whereas 10 mmol·L⁻¹ Ca²⁺ considerably inhibited migration at 8 h. The Ca²⁺ concentration range of 2-10 mmol·L⁻¹ could promote proliferation, osteogenic differentiation, and mineralization of human osteoblasts. Moreover, mineralization was predominantly induced by 8 and 10 mmol·L⁻¹ Ca²⁺. CaSR antagonism could reduce Ca²⁺-induced migration and osteogenic differentiation of human osteoblasts. CONCLUSIONS Low Ca²⁺ concentration favored osteoblast migration, whereas high Ca²⁺ concentration favored osteogenic differentiation. The Ca²⁺ concentrations of 4 and 6 mmol·L⁻¹ could substantially induce osteoblast migration and osteogenic differentiation, and the Ca²⁺-CaSR pathway participated in signal transduction.
Collapse
Affiliation(s)
- Qun Lei
- Stomatological Hospital of Fujian Medical University, Fuzhou 350000, China
| | - Dong Lin
- Stomatological Hospital of Fujian Medical University, Fuzhou 350000, China
| | - Wen-Xiu Huang
- Stomatological Hospital of Fujian Medical University, Fuzhou 350000, China
| | - Dong Wu
- Stomatological Hospital of Fujian Medical University, Fuzhou 350000, China
| | - Jiang Chen
- Stomatological Hospital of Fujian Medical University, Fuzhou 350000, China
| |
Collapse
|
131
|
Makita N, Ando T, Sato J, Manaka K, Mitani K, Kikuchi Y, Niwa T, Ootaki M, Takeba Y, Matsumoto N, Kawakami A, Ogawa T, Nangaku M, Iiri T. Cinacalcet corrects biased allosteric modulation of CaSR by AHH autoantibody. JCI Insight 2019; 4:126449. [PMID: 30996138 DOI: 10.1172/jci.insight.126449] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 03/12/2019] [Indexed: 12/13/2022] Open
Abstract
Biased agonism is a paradigm that may explain the selective activation of a signaling pathway via a GPCR that activates multiple signals. The autoantibody-induced inactivation of the calcium-sensing receptor (CaSR) causes acquired hypocalciuric hypercalcemia (AHH). Here, we describe an instructive case of AHH in which severe hypercalcemia was accompanied by an increased CaSR antibody titer. These autoantibodies operated as biased allosteric modulators of CaSR by targeting its Venus flytrap domain near the Ca2+-binding site. A positive allosteric modulator of CaSR, cinacalcet, which targets its transmembrane domain, overcame this autoantibody effect and successfully corrected the hypercalcemia in this patient. Hence, this is the first study to our knowledge that identifies the interaction site of a disease-causing GPCR autoantibody working as its biased allosteric modulator and demonstrates that cinacalcet can correct the AHH autoantibody effects both in vitro and in our AHH patient. Our observations provide potentially new insights into how biased agonism works and how to design a biased allosteric modulator of a GPCR. Our observations also indicate that the diagnosis of AHH is important because the severity of hypercalcemia may become fatal if the autoantibody titer increases. Calcimimetics may serve as good treatment options for some patients with severe AHH.
Collapse
Affiliation(s)
- Noriko Makita
- Department of Endocrinology and Nephrology, The University of Tokyo School of Medicine, Tokyo, Japan
| | - Takao Ando
- Division of Endocrinology and Metabolism, Nagasaki Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Junichiro Sato
- Department of Endocrinology and Nephrology, The University of Tokyo School of Medicine, Tokyo, Japan
| | - Katsunori Manaka
- Department of Endocrinology and Nephrology, The University of Tokyo School of Medicine, Tokyo, Japan
| | - Koji Mitani
- Department of Endocrinology and Nephrology, The University of Tokyo School of Medicine, Tokyo, Japan
| | - Yasuko Kikuchi
- Department of Breast and Endocrine Surgery, The University of Tokyo School of Medicine, Tokyo, Japan
| | - Takayoshi Niwa
- Department of Breast and Endocrine Surgery, The University of Tokyo School of Medicine, Tokyo, Japan
| | - Masanori Ootaki
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yuko Takeba
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Naoki Matsumoto
- Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Atsushi Kawakami
- Division of Endocrinology and Metabolism, Nagasaki Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Toshihisa Ogawa
- Breast Center, Dokkyo Medical University Koshigaya Hospital, Saitama, Japan
| | - Masaomi Nangaku
- Department of Endocrinology and Nephrology, The University of Tokyo School of Medicine, Tokyo, Japan
| | - Taroh Iiri
- Department of Endocrinology and Nephrology, The University of Tokyo School of Medicine, Tokyo, Japan.,Department of Pharmacology, St. Marianna University School of Medicine, Kawasaki, Japan
| |
Collapse
|
132
|
A Novel Mutation of the Calcium-Sensing Receptor Gene Causing Familial Hypocalciuric Hypercalcemia Complicates Medical Followup after Roux-en-Y Gastric Bypass: A Case Report and a Summary of Mutations Found in the Same Hospital Laboratory. Case Rep Endocrinol 2019; 2019:9468252. [PMID: 30895164 PMCID: PMC6393866 DOI: 10.1155/2019/9468252] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/05/2019] [Indexed: 12/22/2022] Open
Abstract
Heterozygous inactivating mutations in the calcium-sensing receptor (CaSR) gene are known to cause familial hypocalciuric hypercalcemia (FHH), usually a benign form of hypercalcemia without symptoms of a disrupted calcium homeostasis. FHH can be mistaken for the more common primary hyperparathyroidism (PHPT), for which surgical treatment may be needed. We describe a case of a 36-year-old woman with hypercalcemia and elevated PTH, initially suspected of having PHPT. Sequencing of the CaSR-gene revealed a mutation in nucleotide 437, changing the amino acid in position 146 from Glycine to Aspartate. The mutation was previously undescribed in the literature, but a very low calcium:creatinine clearance ratio supported the diagnosis FHH. A few years later, the patient's two daughters were tested and the association between mutation and hypercalcemia could be confirmed. The patient was gastric bypass-operated and therefore, due to malabsorption and increased risk of fracture, was in need of adequate calcium supplementation. The chronically elevated calcium levels challenged medical followup, as calcium sufficiency could not be monitored in a traditional manner. Eventually the patient developed elevated alkaline phosphatase, a further increased PTH and a decreased DXA T-score indicating calcium deficiency and bone resorption. As a supplement, all CaSR-mutations found at our hospital, 2005-2018.
Collapse
|
133
|
Feng R, Ding F, Mi XH, Liu SF, Jiang AL, Liu BH, Lian Y, Shi Q, Wang YJ, Zhang Y. Protective Effects of Ligustroflavone, an Active Compound from Ligustrum lucidum, on Diabetes-Induced Osteoporosis in Mice: A Potential Candidate as Calcium-Sensing Receptor Antagonist. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:457-476. [PMID: 30834778 DOI: 10.1142/s0192415x1950023x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Ligustroflavone is one major compound contained in active fraction from Fructus Ligustri Lucidi (the fruit of Ligustrum lucidum), which could regulate parathyroid hormone (PTH) levels and improve calcium balance by acting on calcium-sensing receptors (CaSR). This study aimed to explore the potency of ligustroflavone as a CaSR antagonist and its protective effects against diabetic osteoporosis in mice. LF interacted well with the allosteric site of CaSR shown by molecular docking analysis, increased PTH release of primary parathyroid gland cells and suppressed extracellular calcium influx in HEK-293 cells. The serum level of PTH attained peak value at 2 h and maintained high during the period of 1 h and 3 h than that before treatment in mice after a single dose of LF. Treatment of diabetic mice with LF inhibited the decrease in calcium level of serum and bone and the enhancement in urinary calcium excretion as well as elevated circulating PTH levels. Trabecular bone mineral density and micro-architecture were markedly improved in diabetic mice upon to LF treatment for 8 weeks. LF reduced CaSR mRNA and protein expression in the kidneys of diabetic mice. Taken together, ligustroflavone could transiently increase PTH level and regulate calcium metabolism as well as prevent osteoporosis in diabetic mice, suggesting that ligustroflavone might be an effective antagonist on CaSR.
Collapse
Affiliation(s)
- Rui Feng
- * Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P. R. China
| | - Fan Ding
- * Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P. R. China
| | - Xiu-Hua Mi
- † Section of Nephrology, Yangpu Traditional Chinese Medicine Hospital, Shanghai 200090, P. R. China
| | - Shu-Fen Liu
- * Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P. R. China.,‡ Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai 200032, P. R. China
| | - Ai-Ling Jiang
- § School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China
| | - Bi-Hui Liu
- ¶ Department of Orthopaedic, Shenzhen Pingle Orthopaedic Hospital, Shenzhen 518000, P. R. China
| | - Yin Lian
- ¶ Department of Orthopaedic, Shenzhen Pingle Orthopaedic Hospital, Shenzhen 518000, P. R. China
| | - Qi Shi
- * Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P. R. China.,‡ Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai 200032, P. R. China
| | - Yong-Jun Wang
- * Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P. R. China.,‡ Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai 200032, P. R. China
| | - Yan Zhang
- * Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P. R. China.,‡ Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai 200032, P. R. China
| |
Collapse
|
134
|
Behrens M, Briand L, de March CA, Matsunami H, Yamashita A, Meyerhof W, Weyand S. Structure-Function Relationships of Olfactory and Taste Receptors. Chem Senses 2019; 43:81-87. [PMID: 29342245 DOI: 10.1093/chemse/bjx083] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The field of chemical senses has made major progress in understanding the cellular mechanisms of olfaction and taste in the past 2 decades. However, the molecular understanding of odor and taste recognition is still lagging far behind and will require solving multiple structures of the relevant full-length receptors in complex with native ligands to achieve this goal. However, the development of multiple complimentary strategies for the structure determination of G protein-coupled receptors (GPCRs) makes this goal realistic. The common conundrum of how multi-specific receptors that recognize a large number of different ligands results in a sensory perception in the brain will only be fully understood by a combination of high-resolution receptor structures and functional studies. This review discusses the first steps on this pathway, including biochemical and physiological assays, forward genetics approaches, molecular modeling, and the first steps towards the structural biology of olfactory and taste receptors.
Collapse
Affiliation(s)
- Maik Behrens
- Department of Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke, Germany
| | - Loïc Briand
- Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRA, Univ. de Bourgogne- Franche-Comté, France
| | - Claire A de March
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, USA
| | - Hiroaki Matsunami
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, USA
| | - Atsuko Yamashita
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Japan
| | - Wolfgang Meyerhof
- Department of Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke, Germany
| | - Simone Weyand
- Department of Biochemistry, University of Cambridge, UK
| |
Collapse
|
135
|
Park J, Selvam B, Sanematsu K, Shigemura N, Shukla D, Procko E. Structural architecture of a dimeric class C GPCR based on co-trafficking of sweet taste receptor subunits. J Biol Chem 2019; 294:4759-4774. [PMID: 30723160 DOI: 10.1074/jbc.ra118.006173] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 01/24/2019] [Indexed: 01/06/2023] Open
Abstract
Class C G protein-coupled receptors (GPCRs) are obligatory dimers that are particularly important for neuronal responses to endogenous and environmental stimuli. Ligand recognition through large extracellular domains leads to the reorganization of transmembrane regions to activate G protein signaling. Although structures of individual domains are known, the complete architecture of a class C GPCR and the mechanism of interdomain coupling during receptor activation are unclear. By screening a mutagenesis library of the human class C sweet taste receptor subunit T1R2, we enhanced surface expression and identified a dibasic intracellular retention motif that modulates surface expression and co-trafficking with its heterodimeric partner T1R3. Using a highly expressed T1R2 variant, dimerization sites along the entire subunit within all the structural domains were identified by a comprehensive mutational scan for co-trafficking with T1R3 in human cells. The data further reveal that the C terminus of the extracellular cysteine-rich domain needs to be properly folded for T1R3 dimerization and co-trafficking, but not for surface expression of T1R2 alone. These results guided the modeling of the T1R2-T1R3 dimer in living cells, which predicts a twisted arrangement of domains around the central axis, and a continuous folded structure between transmembrane domain loops and the cysteine-rich domains. These insights have implications for how conformational changes between domains are coupled within class C GPCRs.
Collapse
Affiliation(s)
- Jihye Park
- From the Departments of Biochemistry and
| | - Balaji Selvam
- Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801 and
| | - Keisuke Sanematsu
- the Section of Oral Neuroscience, Graduate School of Dental Science, and.,the Division of Sensory Physiology-Medical Application Sensing, Research and Development Center for Five-Sense Devices, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Noriatsu Shigemura
- the Section of Oral Neuroscience, Graduate School of Dental Science, and.,the Division of Sensory Physiology-Medical Application Sensing, Research and Development Center for Five-Sense Devices, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Diwakar Shukla
- Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801 and
| | | |
Collapse
|
136
|
Koehl A, Hu H, Feng D, Sun B, Zhang Y, Robertson MJ, Chu M, Kobilka TS, Laeremans T, Steyaert J, Tarrasch J, Dutta S, Fonseca R, Weis WI, Mathiesen JM, Skiniotis G, Kobilka BK. Structural insights into the activation of metabotropic glutamate receptors. Nature 2019; 566:79-84. [PMID: 30675062 PMCID: PMC6709600 DOI: 10.1038/s41586-019-0881-4] [Citation(s) in RCA: 235] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/12/2018] [Indexed: 12/11/2022]
Abstract
Metabotropic glutamate receptors are family C G-protein-coupled receptors. They form obligate dimers and possess extracellular ligand-binding Venus flytrap domains, which are linked by cysteine-rich domains to their 7-transmembrane domains. Spectroscopic studies show that signalling is a dynamic process, in which large-scale conformational changes underlie the transmission of signals from the extracellular Venus flytraps to the G protein-coupling domains-the 7-transmembrane domains-in the membrane. Here, using a combination of X-ray crystallography, cryo-electron microscopy and signalling studies, we present a structural framework for the activation mechanism of metabotropic glutamate receptor subtype 5. Our results show that agonist binding at the Venus flytraps leads to a compaction of the intersubunit dimer interface, thereby bringing the cysteine-rich domains into close proximity. Interactions between the cysteine-rich domains and the second extracellular loops of the receptor enable the rigid-body repositioning of the 7-transmembrane domains, which come into contact with each other to initiate signalling.
Collapse
Affiliation(s)
- Antoine Koehl
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongli Hu
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Dan Feng
- ConfometRx, Santa Clara, CA, USA
| | | | - Yan Zhang
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael J Robertson
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Tong Sun Kobilka
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.,ConfometRx, Santa Clara, CA, USA
| | - Toon Laeremans
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium.,VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
| | - Jan Steyaert
- Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Brussels, Belgium.,VIB-VUB Center for Structural Biology, VIB, Brussels, Belgium
| | - Jeffrey Tarrasch
- Life Sciences Institute and Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Somnath Dutta
- Life Sciences Institute and Department of Biological Chemistry, University of Michigan Medical School, Ann Arbor, MI, USA.,Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India
| | - Rasmus Fonseca
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.,Biosciences Division, SLAC National Accelerator Laboratory, Stanford University, Stanford, CA, USA
| | - William I Weis
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.,Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jesper M Mathiesen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Georgios Skiniotis
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA. .,Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Brian K Kobilka
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA. .,ConfometRx, Santa Clara, CA, USA.
| |
Collapse
|
137
|
Marx SJ, Goltzman D. Evolution of Our Understanding of the Hyperparathyroid Syndromes: A Historical Perspective. J Bone Miner Res 2019; 34:22-37. [PMID: 30536424 PMCID: PMC6396287 DOI: 10.1002/jbmr.3650] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/14/2018] [Accepted: 11/20/2018] [Indexed: 12/19/2022]
Abstract
We review advancing and overlapping stages for our understanding of the expressions of six hyperparathyroid (HPT) syndromes: multiple endocrine neoplasia type 1 (MEN1) or type 4, multiple endocrine neoplasia type 2A (MEN2A), hyperparathyroidism-jaw tumor syndrome, familial hypocalciuric hypercalcemia, neonatal severe primary hyperparathyroidism, and familial isolated hyperparathyroidism. During stage 1 (1903 to 1967), the introduction of robust measurement of serum calcium was a milestone that uncovered hypercalcemia as the first sign of dysfunction in many HPT subjects, and inheritability was reported in each syndrome. The earliest reports of HPT syndromes were biased toward severe or striking manifestations. During stage 2 (1959 to 1985), the early formulations of a syndrome were improved. Radioimmunoassays (parathyroid hormone [PTH], gastrin, insulin, prolactin, calcitonin) were breakthroughs. They could identify a syndrome carrier, indicate an emerging tumor, characterize a tumor, or monitor a tumor. During stage 3 (1981 to 2006), the assembly of many cases enabled recognition of further details. For example, hormone non-secreting skin lesions were discovered in MEN1 and MEN2A. During stage 4 (1985 to the present), new genomic tools were a revolution for gene identification. Four principal genes ("principal" implies mutated or deleted in 50% or more probands for its syndrome) (MEN1, RET, CASR, CDC73) were identified for five syndromes. During stage 5 (1993 to the present), seven syndromal genes other than a principal gene were identified (CDKN1B, CDKN2B, CDKN2C, CDKN1A, GNA11, AP2S1, GCM2). Identification of AP2S1 and GCM2 became possible because of whole-exome sequencing. During stages 4 and 5, the newly identified genes enabled many studies, including robust assignment of the carriers and non-carriers of a mutation. Furthermore, molecular pathways of RET and the calcium-sensing receptor were elaborated, thereby facilitating developments in pharmacotherapy. Current findings hold the promise that more genes for HPT syndromes will be identified and studied in the near future. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Stephen J Marx
- Office of the Scientific Director, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - David Goltzman
- Calcium Research Laboratory, Metabolic Disorders and Complications Program, Research Institute of the McGill University Health Centre, Montreal, Canada
| |
Collapse
|
138
|
Hannan FM, Kallay E, Chang W, Brandi ML, Thakker RV. The calcium-sensing receptor in physiology and in calcitropic and noncalcitropic diseases. Nat Rev Endocrinol 2018; 15:33-51. [PMID: 30443043 PMCID: PMC6535143 DOI: 10.1038/s41574-018-0115-0] [Citation(s) in RCA: 236] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Ca2+-sensing receptor (CaSR) is a dimeric family C G protein-coupled receptor that is expressed in calcitropic tissues such as the parathyroid glands and the kidneys and signals via G proteins and β-arrestin. The CaSR has a pivotal role in bone and mineral metabolism, as it regulates parathyroid hormone secretion, urinary Ca2+ excretion, skeletal development and lactation. The importance of the CaSR for these calcitropic processes is highlighted by loss-of-function and gain-of-function CaSR mutations that cause familial hypocalciuric hypercalcaemia and autosomal dominant hypocalcaemia, respectively, and also by the fact that alterations in parathyroid CaSR expression contribute to the pathogenesis of primary and secondary hyperparathyroidism. Moreover, the CaSR is an established therapeutic target for hyperparathyroid disorders. The CaSR is also expressed in organs not involved in Ca2+ homeostasis: it has noncalcitropic roles in lung and neuronal development, vascular tone, gastrointestinal nutrient sensing, wound healing and secretion of insulin and enteroendocrine hormones. Furthermore, the abnormal expression or function of the CaSR is implicated in cardiovascular and neurological diseases, as well as in asthma, and the CaSR is reported to protect against colorectal cancer and neuroblastoma but increase the malignant potential of prostate and breast cancers.
Collapse
Affiliation(s)
- Fadil M Hannan
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Enikö Kallay
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Wenhan Chang
- Endocrine Research Unit, Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, CA, USA
| | - Maria Luisa Brandi
- Metabolic Bone Diseases Unit, Department of Surgery and Translational Medicine, University of Florence, Florence, Italy.
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
139
|
Gregory K, Kufareva I, Keller AN, Khajehali E, Mun HC, Goolam MA, Mason RS, Capuano B, Conigrave AD, Christopoulos A, Leach K. Dual Action Calcium-Sensing Receptor Modulator Unmasks Novel Mode-Switching Mechanism. ACS Pharmacol Transl Sci 2018; 1:96-109. [PMID: 32219206 PMCID: PMC7089027 DOI: 10.1021/acsptsci.8b00021] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Indexed: 12/17/2022]
Abstract
Negative allosteric modulators (NAMs) of the human calcium-sensing receptor (CaSR) have previously failed to show efficacy in human osteoporosis clinical trials, but there is now significant interest in repurposing these drugs for hypocalcemic disorders and inflammatory lung diseases. However, little is known about how CaSR NAMs inhibit the response to endogenous activators. An improved understanding of CaSR negative allosteric modulation may afford the opportunity to develop therapeutically superior CaSR-targeting drugs. In an attempt to elucidate the mechanistic and structural basis of allosteric modulation mediated by the previously reported NAM, calhex231, we herein demonstrate that calhex231 actually potentiates or inhibits the activity of multiple CaSR agonists depending on whether it occupies one or both protomers in a CaSR dimer. These findings reveal a novel mechanism of mode-switching at a Class C G protein-coupled receptor that has implications for drug discovery and potential clinical utility.
Collapse
Affiliation(s)
- Karen
J. Gregory
- Drug
Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Monash University, Parkville, Victoria 3052, Australia
| | - Irina Kufareva
- Skaggs
School of Pharmacy & Pharmaceutical Sciences, University of California, 9500 Gilman Drive, La Jolla, San Diego, California MC 0747, United States
| | - Andrew N. Keller
- Drug
Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Monash University, Parkville, Victoria 3052, Australia
| | - Elham Khajehali
- Drug
Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Monash University, Parkville, Victoria 3052, Australia
| | - Hee-Chang Mun
- School of Life and Environmental
Sciences, Charles Perkins Centre, and Physiology and
Bosch Institute, Building F13, University
of Sydney, Sidney, New South Wales 2006, Australia
| | - Mahvash A. Goolam
- School of Life and Environmental
Sciences, Charles Perkins Centre, and Physiology and
Bosch Institute, Building F13, University
of Sydney, Sidney, New South Wales 2006, Australia
| | - Rebecca S. Mason
- School of Life and Environmental
Sciences, Charles Perkins Centre, and Physiology and
Bosch Institute, Building F13, University
of Sydney, Sidney, New South Wales 2006, Australia
| | - Ben Capuano
- Drug
Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Monash University, Parkville, Victoria 3052, Australia
| | - Arthur D. Conigrave
- School of Life and Environmental
Sciences, Charles Perkins Centre, and Physiology and
Bosch Institute, Building F13, University
of Sydney, Sidney, New South Wales 2006, Australia
| | - Arthur Christopoulos
- Drug
Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Monash University, Parkville, Victoria 3052, Australia
| | - Katie Leach
- Drug
Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Monash University, Parkville, Victoria 3052, Australia
| |
Collapse
|
140
|
Sun X, Huang L, Wu J, Tao Y, Yang F. Novel homozygous inactivating mutation of the calcium-sensing receptor gene in neonatal severe hyperparathyroidism responding to cinacalcet therapy: A case report and literature review. Medicine (Baltimore) 2018; 97:e13128. [PMID: 30407334 PMCID: PMC6250440 DOI: 10.1097/md.0000000000013128] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
RATIONALE Calcium-sensing receptor (CaSR) mutations can cause life-threatening neonatal severe hyperparathyroidism (NSHPT). The medical management of NSHPT is often challenging and complex. Here, we present a case of NSHPT caused by a novel homozygous CaSR mutation. PATIENT CONCERNS A Chinese female infant presented with poor feeding, constipation, severe hypotonia, and periodic bradycardia. Biochemistry tests revealed markedly elevated serum levels of Ca and parathyroid hormone (PTH). DIAGNOSES Genetic sequencing revealed a previously undescribed CaSR mutation in exon 3 (c.242T>A; p.I81K). A diagnosis of NSHPT secondary to homozygously inherited familial hypocalciuric hypercalcemia syndrome was established. INTERVENTIONS Cinacalcet was administered after the common treatments (low-calcium intake, hydration, and furosemide), calcitonin, and pamidronate therapy all failed. OUTCOMES Serum Ca decreased and stabilized with cinacalcet therapy. During a 10-month follow-up, total calcium was maintained within the high-normal range and PTH was normalized. LESSONS A trial of cinacalcet therapy might be undertaken in cases of NSHPT while definitive results of the genetic analysis are awaited.
Collapse
MESH Headings
- Calcimimetic Agents/therapeutic use
- Calcium/blood
- Cinacalcet/therapeutic use
- Female
- Genetic Testing
- Homozygote
- Humans
- Hyperparathyroidism, Primary/diagnosis
- Hyperparathyroidism, Primary/drug therapy
- Hyperparathyroidism, Primary/genetics
- Infant
- Infant, Newborn
- Infant, Newborn, Diseases/diagnosis
- Infant, Newborn, Diseases/drug therapy
- Infant, Newborn, Diseases/genetics
- Mutation
- Parathyroid Hormone/blood
- Receptors, Calcium-Sensing/genetics
Collapse
Affiliation(s)
- Xiaomei Sun
- Department of Pediatrics
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education
| | - Liang Huang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education
- Department of Pharmacy/Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jin Wu
- Department of Pediatrics
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education
| | - Yuhong Tao
- Department of Pediatrics
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education
| | - Fan Yang
- Department of Pediatrics
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education
| |
Collapse
|
141
|
Gorvin CM, Frost M, Malinauskas T, Cranston T, Boon H, Siebold C, Jones EY, Hannan FM, Thakker RV. Calcium-sensing receptor residues with loss- and gain-of-function mutations are located in regions of conformational change and cause signalling bias. Hum Mol Genet 2018; 27:3720-3733. [PMID: 30052933 PMCID: PMC6196656 DOI: 10.1093/hmg/ddy263] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 12/20/2022] Open
Abstract
The calcium-sensing receptor (CaSR) is a homodimeric G-protein-coupled receptor that signals via intracellular calcium (Ca2+i) mobilisation and phosphorylation of extracellular signal-regulated kinase 1/2 (ERK) to regulate extracellular calcium (Ca2+e) homeostasis. The central importance of the CaSR in Ca2+e homeostasis has been demonstrated by the identification of loss- or gain-of-function CaSR mutations that lead to familial hypocalciuric hypercalcaemia (FHH) or autosomal dominant hypocalcaemia (ADH), respectively. However, the mechanisms determining whether the CaSR signals via Ca2+i or ERK have not been established, and we hypothesised that some CaSR residues, which are the site of both loss- and gain-of-function mutations, may act as molecular switches to direct signalling through these pathways. An analysis of CaSR mutations identified in >300 hypercalcaemic and hypocalcaemic probands revealed five 'disease-switch' residues (Gln27, Asn178, Ser657, Ser820 and Thr828) that are affected by FHH and ADH mutations. Functional expression studies using HEK293 cells showed disease-switch residue mutations to commonly display signalling bias. For example, two FHH-associated mutations (p.Asn178Asp and p.Ser820Ala) impaired Ca2+i signalling without altering ERK phosphorylation. In contrast, an ADH-associated p.Ser657Cys mutation uncoupled signalling by leading to increased Ca2+i mobilization while decreasing ERK phosphorylation. Structural analysis of these five CaSR disease-switch residues together with four reported disease-switch residues revealed these residues to be located at conformationally active regions of the CaSR such as the extracellular dimer interface and transmembrane domain. Thus, our findings indicate that disease-switch residues are located at sites critical for CaSR activation and play a role in mediating signalling bias.
Collapse
Affiliation(s)
- Caroline M Gorvin
- Academic Endocrine Unit, Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Oxford OX3 7LJ, UK
| | - Morten Frost
- Academic Endocrine Unit, Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Oxford OX3 7LJ, UK
- University of Southern Denmark, Odense C, Denmark
| | - Tomas Malinauskas
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Treena Cranston
- Oxford Molecular Genetics Laboratory, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Hannah Boon
- Oxford Molecular Genetics Laboratory, Churchill Hospital, Oxford OX3 7LJ, UK
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - E Yvonne Jones
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Fadil M Hannan
- Academic Endocrine Unit, Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Oxford OX3 7LJ, UK
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford, Oxford OX3 7LJ, UK
| |
Collapse
|
142
|
Michigami T, Kawai M, Yamazaki M, Ozono K. Phosphate as a Signaling Molecule and Its Sensing Mechanism. Physiol Rev 2018; 98:2317-2348. [DOI: 10.1152/physrev.00022.2017] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In mammals, phosphate balance is maintained by influx and efflux via the intestines, kidneys, bone, and soft tissue, which involves multiple sodium/phosphate (Na+/Pi) cotransporters, as well as regulation by several hormones. Alterations in the levels of extracellular phosphate exert effects on both skeletal and extra-skeletal tissues, and accumulating evidence has suggested that phosphate itself evokes signal transduction to regulate gene expression and cell behavior. Several in vitro studies have demonstrated that an elevation in extracellular Piactivates fibroblast growth factor receptor, Raf/MEK (mitogen-activated protein kinase/ERK kinase)/ERK (extracellular signal-regulated kinase) pathway and Akt pathway, which might involve the type III Na+/Picotransporter PiT-1. Excessive phosphate loading can lead to various harmful effects by accelerating ectopic calcification, enhancing oxidative stress, and dysregulating signal transduction. The responsiveness of mammalian cells to altered extracellular phosphate levels suggests that they may sense and adapt to phosphate availability, although the precise mechanism for phosphate sensing in mammals remains unclear. Unicellular organisms, such as bacteria and yeast, use some types of Pitransporters and other molecules, such as kinases, to sense the environmental Piavailability. Multicellular animals may need to integrate signals from various organs to sense the phosphate levels as a whole organism, similarly to higher plants. Clarification of the phosphate-sensing mechanism in humans may lead to the development of new therapeutic strategies to prevent and treat diseases caused by phosphate imbalance.
Collapse
Affiliation(s)
- Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masanobu Kawai
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Miwa Yamazaki
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Keiichi Ozono
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
143
|
Abstract
Calcium is vital for life, and extracellular calcium concentrations must constantly be maintained within a precise concentration range. Low serum calcium (hypocalcemia) occurs in conjunction with multiple disorders and can be life-threatening if severe. Symptoms of acute hypocalcemia include neuromuscular irritability, tetany, and seizures, which are rapidly resolved with intravenous administration of calcium gluconate. However, disorders that lead to chronic hypocalcemia often have more subtle manifestations. Hypoparathyroidism, characterized by impaired secretion of parathyroid hormone (PTH), a key regulatory hormone for maintaining calcium homeostasis, is a classic cause of chronic hypocalcemia. Disorders that disrupt the metabolism of vitamin D can also lead to chronic hypocalcemia, as vitamin D is responsible for increasing the gut absorption of dietary calcium. Treatment and management options for chronic hypocalcemia vary depending on the underlying disorder. For example, in patients with hypoparathyroidism, calcium and vitamin D supplementation must be carefully titrated to avoid symptoms of hypocalcemia while keeping serum calcium in the low-normal range to minimize hypercalciuria, which can lead to renal dysfunction. Management of chronic hypocalcemia requires knowledge of the factors that influence the complex regulatory axes of calcium homeostasis in a given disorder. This chapter discusses common and rare disorders of hypocalcemia, symptoms and workup, and management options including replacement of PTH in hypoparathyroidism.
Collapse
Affiliation(s)
- Erin Bove-Fenderson
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Thier 1101, 50 Blossom St, Boston, MA, 02114, USA
| | - Michael Mannstadt
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Thier 1101, 50 Blossom St, Boston, MA, 02114, USA.
| |
Collapse
|
144
|
Acar I, Cetinkaya A, Lay I, Ileri-Gurel E. The role of calcium sensing receptors in GLP-1 and PYY secretion after acute intraduodenal administration of L-Tryptophan in rats. Nutr Neurosci 2018; 23:481-489. [PMID: 30222528 DOI: 10.1080/1028415x.2018.1521906] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objectives: The calcium-sensing receptor (CaSR), the major sensor of extracellular Ca2+, is expressed in various tissues, including the gastrointestinal tract. Although the essential ligand of CaSR is calcium, its activity can be regulated by aromatic L-amino acids. The expression of CaSR on enteroendocrine cells suggests that CaSR functions as a physiological amino acid sensor for gut hormone release. Here, we investigated the effects of L-tryptophan (L-Trp) on rat glucagon-like peptide-1 (GLP-1), peptide YY (PYY), and insulin secretion, and the role of CaSR in this mechanism in vivo.Methods: The effects of intraduodenal L-Trp on GLP-1, PYY, and insulin secretion were investigated. A CaSR antagonist, NPS 2143, was administered to determine whether CaSR plays a role in L-Trp-mediated gut hormone release. Male Wistar rats were divided into L-Trp, L-Trp+NPS 2143, and L-Trp+vehicle groups. Blood samples were collected, before and after the intraduodenal infusions, for determining plasma glucose, L-Trp, insulin, GLP-1, and PYY levels.Results: Our study showed a significant increase in plasma GLP-1 and insulin levels, but not plasma PYY and glucose levels, following the acute intraduodenal administration of L-Trp. We demonstrated that CaSR plays a role in L-Trp-mediated GLP-1 secretion due to attenuation of GLP-1 release with the CaSR antagonist NPS 2143.Discussion: We demonstrated that GLP-1, but not PYY, secretion following intraduodenal L-Trp administration was mediated through calcium-sensing receptors. This mechanism underlying protein sensing in the gastrointestinal system may be important for the development of new therapeutic strategies without side effects for obesity and diabetes.
Collapse
Affiliation(s)
- Ipek Acar
- Physiology Department, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Alper Cetinkaya
- Laboratory Animals Research and Application Center, Hacettepe University, Ankara, Turkey
| | - Incilay Lay
- Medical Biochemistry Department, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Esin Ileri-Gurel
- Physiology Department, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| |
Collapse
|
145
|
Mechanisms of signalling and biased agonism in G protein-coupled receptors. Nat Rev Mol Cell Biol 2018; 19:638-653. [DOI: 10.1038/s41580-018-0049-3] [Citation(s) in RCA: 323] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
146
|
Cavaco BM, Canaff L, Nolin-Lapalme A, Vieira M, Silva TN, Saramago A, Domingues R, Rutter MM, Hudon J, Gleason JL, Leite V, Hendy GN. Homozygous Calcium-Sensing Receptor Polymorphism R544Q Presents as Hypocalcemic Hypoparathyroidism. J Clin Endocrinol Metab 2018; 103:2879-2888. [PMID: 29846619 DOI: 10.1210/jc.2017-02407] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 05/22/2018] [Indexed: 02/11/2023]
Abstract
CONTEXT Autosomal dominant hypocalcemia type 1 (ADH1) is caused by heterozygous activating mutations in the calcium-sensing receptor gene (CASR). Whether polymorphisms that are benign in the heterozygous state pathologically alter receptor function in the homozygous state is unknown. OBJECTIVE To identify the genetic defect in an adolescent female with a history of surgery for bilateral cataracts and seizures. The patient has hypocalcemia, hyperphosphatemia, and low serum PTH level. The parents of the proband are healthy. METHODS Mutation testing of PTH, GNA11, GCM2, and CASR was done on leukocyte DNA of the proband. Functional analysis in transfected cells was conducted on the gene variant identified. Public single nucleotide polymorphism (SNP) databases were searched for the presence of the variant allele. RESULTS No mutations were identified in PTH, GNA11, and GCM2 in the proband. However, a germline homozygous variant (c.1631G>A; p.R544Q) in exon 6 of the CASR was identified. Both parents are heterozygous for the variant. The variant allele frequency was near 0.1% in SNP databases. By in vitro functional analysis, the variant was significantly more potent in stimulating both the Ca2+i and MAPK signaling pathways than wild type when transfected alone (P < 0.05) but not when transfected together with wild type. The overactivity of the mutant CaSR is due to loss of a critical structural cation-π interaction. CONCLUSIONS The patient's hypoparathyroidism is due to homozygosity of a variant in the CASR that normally has weak or no phenotypic expression in heterozygosity. Although rare, this has important implications for genetic counseling and clinical management.
Collapse
Affiliation(s)
- Branca M Cavaco
- Unidade de Investigação em Patobiologia Molecular, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal
| | - Lucie Canaff
- Departments of Medicine, Physiology, and Human Genetics, McGill University Health Centre Research Institute, McGill University, Montréal, Quebec, Canada
| | - Alexis Nolin-Lapalme
- Departments of Medicine, Physiology, and Human Genetics, McGill University Health Centre Research Institute, McGill University, Montréal, Quebec, Canada
| | - Margarida Vieira
- Serviço de Endocrinologia, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal
| | - Tiago N Silva
- Serviço de Endocrinologia, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal
| | - Ana Saramago
- Unidade de Investigação em Patobiologia Molecular, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal
| | - Rita Domingues
- Unidade de Investigação em Patobiologia Molecular, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal
| | - Meilan M Rutter
- Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jonathan Hudon
- Department of Chemistry, McGill University, Montreal, Quebec, Canada
| | - James L Gleason
- Department of Chemistry, McGill University, Montreal, Quebec, Canada
| | - Valeriano Leite
- Unidade de Investigação em Patobiologia Molecular, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal
- Serviço de Endocrinologia, Instituto Português de Oncologia de Lisboa Francisco Gentil, Lisboa, Portugal
| | - Geoffrey N Hendy
- Departments of Medicine, Physiology, and Human Genetics, McGill University Health Centre Research Institute, McGill University, Montréal, Quebec, Canada
| |
Collapse
|
147
|
Ferraretto A, Bottani M, Villa I, Giusto L, Signo M, Senesi P, Montesano A, Vacante F, Luzi L, Rubinacci A, Terruzzi I. L-Carnitine activates calcium signaling in human osteoblasts. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.05.068] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
148
|
Structural insights into G-protein-coupled receptor allostery. Nature 2018; 559:45-53. [DOI: 10.1038/s41586-018-0259-z] [Citation(s) in RCA: 194] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 05/17/2018] [Indexed: 01/14/2023]
|
149
|
Gorvin CM. Insights into calcium-sensing receptor trafficking and biased signalling by studies of calcium homeostasis. J Mol Endocrinol 2018; 61:R1-R12. [PMID: 29599414 DOI: 10.1530/jme-18-0049] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 03/29/2018] [Indexed: 12/18/2022]
Abstract
The calcium-sensing receptor (CASR) is a class C G-protein-coupled receptor (GPCR) that detects extracellular calcium concentrations, and modulates parathyroid hormone secretion and urinary calcium excretion to maintain calcium homeostasis. The CASR utilises multiple heterotrimeric G-proteins to mediate signalling effects including activation of intracellular calcium release; mitogen-activated protein kinase (MAPK) pathways; membrane ruffling; and inhibition of cAMP production. By studying germline mutations in the CASR and proteins within its signalling pathway that cause hyper- and hypocalcaemic disorders, novel mechanisms governing GPCR signalling and trafficking have been elucidated. This review focusses on two recently described pathways that provide novel insights into CASR signalling and trafficking mechanisms. The first, identified by studying a CASR gain-of-function mutation that causes autosomal dominant hypocalcaemia (ADH), demonstrated a structural motif located between the third transmembrane domain and the second extracellular loop of the CASR that mediates biased signalling by activating a novel β-arrestin-mediated G-protein-independent pathway. The second, in which the mechanism by which adaptor protein-2 σ-subunit (AP2σ) mutations cause familial hypocalciuric hypercalcaemia (FHH) was investigated, demonstrated that AP2σ mutations impair CASR internalisation and reduce multiple CASR-mediated signalling pathways. Furthermore, these studies showed that the CASR can signal from the cell surface using multiple G-protein pathways, whilst sustained signalling is mediated only by the Gq/11 pathway. Thus, studies of FHH- and ADH-associated mutations have revealed novel steps by which CASR mediates signalling and compartmental bias, and these pathways could provide new targets for therapies for patients with calcaemic disorders.
Collapse
Affiliation(s)
- Caroline M Gorvin
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, UK
- Centre for Endocrinology, Diabetes and Metabolism (CEDAM), Birmingham Health Partners, Birmingham, UK
| |
Collapse
|
150
|
Frangaj A, Fan QR. Structural biology of GABA B receptor. Neuropharmacology 2018; 136:68-79. [PMID: 29031577 PMCID: PMC5897222 DOI: 10.1016/j.neuropharm.2017.10.011] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 10/09/2017] [Accepted: 10/11/2017] [Indexed: 11/17/2022]
Abstract
Metabotropic GABAB receptor is a G protein-coupled receptor (GPCR) that mediates slow and prolonged inhibitory neurotransmission in the brain. It functions as a constitutive heterodimer composed of the GABAB1 and GABAB2 subunits. Each subunit contains three domains; the extracellular Venus flytrap module, seven-helix transmembrane region and cytoplasmic tail. In recent years, the three-dimensional structures of GABAB receptor extracellular and intracellular domains have been elucidated. These structures reveal the molecular basis of ligand recognition, receptor heterodimerization and receptor activation. Here we provide a brief review of the GABAB receptor structures, with an emphasis on describing the different ligand-bound states of the receptor. We will also compare these with the known structures of related GPCRs to shed light on the molecular mechanisms of activation and regulation in the GABAB system, as well as GPCR dimers in general. This article is part of the "Special Issue Dedicated to Norman G. Bowery".
Collapse
Affiliation(s)
- Aurel Frangaj
- Department of Pharmacology, Columbia University, New York, NY 10032, USA
| | - Qing R Fan
- Department of Pharmacology, Columbia University, New York, NY 10032, USA; Department of Pathology & Cell Biology, Columbia University, New York, NY 10032, USA.
| |
Collapse
|