1
|
Tian L, Andrews C, Yan Q, Yang JJ. Molecular regulation of calcium-sensing receptor (CaSR)-mediated signaling. Chronic Dis Transl Med 2024; 10:167-194. [PMID: 39027195 PMCID: PMC11252437 DOI: 10.1002/cdt3.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 03/29/2024] [Accepted: 04/09/2024] [Indexed: 07/20/2024] Open
Abstract
Calcium-sensing receptor (CaSR), a family C G-protein-coupled receptor, plays a crucial role in regulating calcium homeostasis by sensing small concentration changes of extracellular Ca2+, Mg2+, amino acids (e.g., L-Trp and L-Phe), small peptides, anions (e.g., HCO3 - and PO4 3-), and pH. CaSR-mediated intracellular Ca2+ signaling regulates a diverse set of cellular processes including gene transcription, cell proliferation, differentiation, apoptosis, muscle contraction, and neuronal transmission. Dysfunction of CaSR with mutations results in diseases such as autosomal dominant hypocalcemia, familial hypocalciuric hypercalcemia, and neonatal severe hyperparathyroidism. CaSR also influences calciotropic disorders, such as osteoporosis, and noncalciotropic disorders, such as cancer, Alzheimer's disease, and pulmonary arterial hypertension. This study first reviews recent advances in biochemical and structural determination of the framework of CaSR and its interaction sites with natural ligands, as well as exogenous positive allosteric modulators and negative allosteric modulators. The establishment of the first CaSR protein-protein interactome network revealed 94 novel players involved in protein processing in endoplasmic reticulum, trafficking, cell surface expression, endocytosis, degradation, and signaling pathways. The roles of these proteins in Ca2+-dependent cellular physiological processes and in CaSR-dependent cellular signaling provide new insights into the molecular basis of diseases caused by CaSR mutations and dysregulated CaSR activity caused by its protein interactors and facilitate the design of therapeutic agents that target CaSR and other family C G-protein-coupled receptors.
Collapse
Affiliation(s)
- Li Tian
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging FacilityGeorgia State UniversityAtlantaGeorgiaUSA
| | - Corey Andrews
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging FacilityGeorgia State UniversityAtlantaGeorgiaUSA
| | - Qiuyun Yan
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging FacilityGeorgia State UniversityAtlantaGeorgiaUSA
| | - Jenny J. Yang
- Department of Chemistry, Center for Diagnostics and Therapeutics, Advanced Translational Imaging FacilityGeorgia State UniversityAtlantaGeorgiaUSA
| |
Collapse
|
2
|
Saglia C, Arruga F, Scolari C, Kalantari S, Albanese S, Bracciamà V, Corso Faini A, Brach Del Prever G, Luca M, Romeo C, Mioli F, Migliorero M, Tessaris D, Carli D, Amoroso A, Vaisitti T, De Sanctis L, Deaglio S. Functional evaluation of a novel nonsense variant of the calcium-sensing receptor gene leading to hypocalcemia. Eur J Endocrinol 2024; 190:296-306. [PMID: 38561929 DOI: 10.1093/ejendo/lvae035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVE The calcium-sensing receptor (CASR) gene encodes a G protein-coupled receptor crucial for calcium homeostasis. Gain-of-function CASR variants result in hypocalcemia, while loss-of-function variants lead to hypercalcemia. This study aims to assess the functional consequences of the novel nonsense CASR variant [c.2897_2898insCTGA, p.(Gln967*) (Q967*)] identified in adolescent patient with chronic hypocalcemia, a phenotype expected for a gain-of-function variants. DESIGN AND METHODS To functionally characterize the Q967* mutant receptor, both wild-type (WT) and mutant CASR were transiently transfected into HEK293T cells and calcium-sensing receptor (CaSR) protein expression and functions were comparatively evaluated using multiple read-outs. RESULTS Western blot analysis revealed that the CaSR mutant protein displayed a lower molecular weight compared with the WT, consistent with the loss of the last 122 amino acids in the intracellular domain. Mitogen-activated protein kinase activation and serum responsive element luciferase assays demonstrated that the mutant receptor had higher baseline activity than the WT. Extracellular-signal-regulated kinase/c-Jun N-terminal kinase phosphorylation, however, remained consistently high in the mutant, without significant modulations following exposure to increasing extracellular calcium (Ca2+o) levels, suggesting that the mutant receptor is more sensitive to Ca2+o compared with the WT. CONCLUSIONS This study provides functional validation of the pathogenicity of a novel nonsense CASR variant, resulting in an abnormally hyperfunctioning protein consistent with the patient's phenotype. Functional analyses indicate that mutant receptor is constitutively active and poorly sensitive to increasing concentrations of extracellular calcium, suggesting that the cytoplasmic tail may contain elements regulating signal transduction.
Collapse
Affiliation(s)
- Claudia Saglia
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin 10126, Italy
| | - Francesca Arruga
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin 10126, Italy
| | - Caterina Scolari
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin 10126, Italy
| | - Silvia Kalantari
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin 10126, Italy
| | - Serena Albanese
- Department of Public Health and Pediatric Sciences, University of Torino, Torino 10126, Italy
- Pediatric Endocrinology, Regina Margherita Childrens' Hospital, Torino 10126, Italy
| | - Valeria Bracciamà
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin 10126, Italy
| | - Angelo Corso Faini
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin 10126, Italy
| | - Giulia Brach Del Prever
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin 10126, Italy
| | - Maria Luca
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin 10126, Italy
| | - Carmelo Romeo
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin 10126, Italy
| | - Fiorenza Mioli
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin 10126, Italy
| | | | - Daniele Tessaris
- Department of Public Health and Pediatric Sciences, University of Torino, Torino 10126, Italy
- Pediatric Endocrinology, Regina Margherita Childrens' Hospital, Torino 10126, Italy
| | - Diana Carli
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin 10126, Italy
| | - Antonio Amoroso
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin 10126, Italy
| | - Tiziana Vaisitti
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin 10126, Italy
| | - Luisa De Sanctis
- Department of Public Health and Pediatric Sciences, University of Torino, Torino 10126, Italy
- Pediatric Endocrinology, Regina Margherita Childrens' Hospital, Torino 10126, Italy
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Turin 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin 10126, Italy
| |
Collapse
|
3
|
Hakimi S, Dutta P, Layton AT. Coupling of renal sodium and calcium transport: a modeling analysis of transporter inhibition and sex differences. Am J Physiol Renal Physiol 2023; 325:F536-F551. [PMID: 37615047 DOI: 10.1152/ajprenal.00145.2023] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/17/2023] [Accepted: 08/17/2023] [Indexed: 08/25/2023] Open
Abstract
Ca2+ transport along the nephron occurs via specific transcellular and paracellular pathways and is coupled to the transport of other electrolytes. Notably, Na+ transport establishes an electrochemical gradient to drive Ca2+ reabsorption. Hence, alterations in renal Na+ handling, under pathophysiological conditions or pharmacological manipulations, can have major effects on Ca2+ transport. An important class of pharmacological agent is diuretics, which are commonly prescribed for the management of blood pressure and fluid balance. The pharmacological targets of diuretics generally directly facilitate Na+ transport but also indirectly affect renal Ca2+ handling. To better understand the underlying mechanisms, we developed a computational model of electrolyte transport along the superficial nephron in the kidney of a male and female rat. Sex differences in renal Ca2+ handling are represented. Model simulations predicted in the female rat nephron lower Ca2+ reabsorption in the proximal tubule and thick ascending limb, but higher reabsorption in the late distal convoluted tubule and connecting tubule, compared with the male nephron. The male rat kidney model yielded a higher urinary Ca2+ excretion than the female model, consistent with animal experiments. Model results indicated that along the proximal tubule and thick ascending limb, Ca2+ and Na+ transport occurred in parallel, but those processes were dissociated in the distal convoluted tubule. Additionally, we conducted simulations of inhibition of channels and transporters that play a major role in Na+ and Ca2+ transport. Simulation results revealed alterations in transepithelial Ca2+ transport, with differential effects among nephron segments and between the sexes.NEW & NOTEWORTHY The kidney plays an important role in the maintenance of whole body Ca2+ balance by regulating Ca2+ reabsorption and excretion. This computational modeling study provides insights into how Ca2+ transport along the nephron is coupled to Na+. Model results indicated that along the proximal tubule and thick ascending limb, Ca2+ and Na+ transport occur in parallel, but those processes were dissociated in the distal convoluted tubule. Simulations also revealed sex-specific responses to different pharmacological manipulations.
Collapse
Affiliation(s)
- Shervin Hakimi
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
| | - Pritha Dutta
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
| | - Anita T Layton
- Department of Applied Mathematics, University of Waterloo, Waterloo, Ontario, Canada
- Department of Biology, Cheriton School of Computer Science, and School of Pharmacology, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
4
|
Meng KL, Jiao MZ, Shi XG, Xu R, Cheng PX, Lv HT, Zheng XH, Xiao CN. A rapid approach to capture the potential bioactive compounds from Rhizoma Drynariae, utilizing disease-associated mutation in calcium sensing receptor to alter the binding affinity for agonists. J Pharm Biomed Anal 2023; 226:115253. [PMID: 36657349 DOI: 10.1016/j.jpba.2023.115253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/15/2023]
Abstract
Rhizoma Drynariae (RD) was used clinically to treat osteoporosis in China due to stimulating bone formation and inhibiting bone resorption, however, the bioactive constituents with the dual effect on bone are still unknown exactly. Disease-causing mutations in calcium sensing receptor (CaSR) can alter parathyroid hormone secretion and affect Ca2+ release from bone and Ca2+ reabsorption from kidney, which gives an indication that CaSR is a potential target for developing therapeutics to manage osteoporosis. Herein, a chromatographic approach was established, by immobilizing the mutant CaSR onto the surface of silica gels as stationary phase in a one-step procedure and then adding the different amino acids into mobile phase as competitors, for exploring the binding features of the known agonists and further screening ligands from RD. The mutant CaSR-coated column was prepared rapidly without the complicated purification and separation of the receptor, which had the large capacity of 13.1 mg CaSR /g silica gels and kept a good stability and specificity for at least 35 days. The CaSR mutation can weaken the binding affinities for three agonists, and the largest decreases occurred on the mutational site Thr151Met for neomycin, on the two sites of Asn118Lys and Glu191Lys for gentamicin-C, and on the site Phe612Ser for kanamycin, which gained new insights into their structure-function relationship. The potential bioactive compounds from RD were screened using the mutant CaSR-coated column and were recognized as coumaric acid 4-O-β-D-glucopyranoside, caffeic acid, and naringin using UPLC-MS. Among them, naringin targeting CaSR gives a possible explanation that RD could manage osteoporosis. These results indicated that, such a rapid and simple method, utilizing disease-associated mutation in CaSR to alter the binding affinity for agonists, can be applied in capturing the potential bioactive compounds efficiently from complex matrices like herb medicines.
Collapse
Affiliation(s)
- Kai-Li Meng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Shaanxi Provincial Key Laboratory of Biotechnology; College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Mei-Zhi Jiao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Shaanxi Provincial Key Laboratory of Biotechnology; College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Xian-Gang Shi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Shaanxi Provincial Key Laboratory of Biotechnology; College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Ru Xu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Shaanxi Provincial Key Laboratory of Biotechnology; College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Pei-Xuan Cheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Shaanxi Provincial Key Laboratory of Biotechnology; College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Hui-Ting Lv
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Shaanxi Provincial Key Laboratory of Biotechnology; College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Xiao-Hui Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Shaanxi Provincial Key Laboratory of Biotechnology; College of Life Sciences, Northwest University, Xi'an 710069, PR China
| | - Chao-Ni Xiao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education; Shaanxi Provincial Key Laboratory of Biotechnology; College of Life Sciences, Northwest University, Xi'an 710069, PR China.
| |
Collapse
|
5
|
Goolam MA, Brown AP, Edwards KT, Gregory KJ, Leach K, Conigrave AD. Cell Surface Calcium-Sensing Receptor Heterodimers: Mutant Gene Dosage Affects Ca 2+ Sensing but Not G Protein Interaction. J Bone Miner Res 2022; 37:1787-1807. [PMID: 35848051 PMCID: PMC9545990 DOI: 10.1002/jbmr.4651] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 06/20/2022] [Accepted: 07/14/2022] [Indexed: 11/20/2022]
Abstract
The calcium-sensing receptor is a homodimeric class C G protein-coupled receptor (GPCR) that senses extracellular Ca2+ (Ca2+ o ) via a dimeric extracellular Venus flytrap (VFT) unit that activates G protein-dependent signaling via twin Cysteine-rich domains linked to transmembrane heptahelical (HH) bundles. It plays a key role in the regulation of human calcium and thus mineral metabolism. However, the nature of interactions between VFT units and HH bundles, and the impacts of heterozygous or homozygous inactivating mutations, which have implications for disorders of calcium metabolism are not yet clearly defined. Herein we generated CaSR-GABAB1 and CaSR-GABAB2 chimeras subject to GABAB -dependent endoplasmic reticulum sorting to traffic mutant heterodimers to the cell surface. Transfected HEK-293 cells were assessed for Ca2+ o -stimulated Ca2+ i mobilization using mutations in either the VFT domains and/or HH bundle intraloop-2 or intraloop-3. When the same mutation was present in both VFT domains of receptor dimers, analogous to homozygous neonatal severe hyperparathyroidism (NSHPT), receptor function was markedly impaired. Mutant heterodimers containing one wild-type (WT) and one mutant VFT domain, however, corresponding to heterozygous familial hypocalciuric hypercalcemia type-1 (FHH-1), supported maximal signaling with reduced Ca2+ o potency. Thus two WT VFT domains were required for normal Ca2+ o potency and there was a pronounced gene-dosage effect. In contrast, a single WT HH bundle was insufficient for maximal signaling and there was no functional difference between heterodimers in which the mutation was present in one or both intraloops; ie, no gene-dosage effect. Finally, we observed that the Ca2+ o -stimulated CaSR operated exclusively via signaling in-trans and not via combined in-trans and in-cis signaling. We consider how receptor asymmetry may support the underlying mechanisms. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Mahvash A Goolam
- School of Life and Environmental Sciences, Charles Perkins Centre (D17), University of Sydney, NSW, Australia
| | - Alice P Brown
- School of Life and Environmental Sciences, Charles Perkins Centre (D17), University of Sydney, NSW, Australia
| | - Kimberly T Edwards
- School of Life and Environmental Sciences, Charles Perkins Centre (D17), University of Sydney, NSW, Australia
| | - Karen J Gregory
- Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Katie Leach
- Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria, Australia
| | - Arthur D Conigrave
- School of Life and Environmental Sciences, Charles Perkins Centre (D17), University of Sydney, NSW, Australia
| |
Collapse
|
6
|
Wang X, Lundblad J, Smith SM. Reduced affinity of calcium sensing-receptor heterodimers and reduced mutant homodimer trafficking combine to impair function in a model of familial hypocalciuric hypercalcemia type 1. PLoS One 2022; 17:e0266993. [PMID: 35857775 PMCID: PMC9299317 DOI: 10.1371/journal.pone.0266993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/30/2022] [Indexed: 11/18/2022] Open
Abstract
Heterozygous loss-of-function mutation of the calcium sensing-receptor (CaSR), causes familial hypocalciuric hypercalcemia type 1 (FHH1), a typically benign condition characterized by mild hypercalcemia. In contrast, homozygous mutation of this dimer-forming G-protein coupled receptor manifests as the lethal neonatal severe hyperparathyroidism (NSHPT). To investigate the mechanisms by which CaSR mutations lead to these distinct disease states, we engineered wild-type (WT) and an exon 5-deficient disease-causing mutation, and transfected expression constructs into human embryonic kidney (HEK) cells. WT protein was mainly membrane-expressed whereas the mutant CaSR protein (mCaSR) was confined to the cytoplasm. Co-expression of WT CaSR directed mCaSR to the cell membrane. In assays of CaSR function, increases in extracellular [Ca2+] ([Ca2+]o) increased intracellular [Ca2+] ([Ca2+]i) in cells expressing WT CaSR while the response was reduced in cells co-expressing mutant and WT receptor. Untransfected cells or those expressing mCaSR alone, showed minimal, equivalent responses to increased [Ca2+]o. Immunoprecipitation experiments confirmed an association between mutant and wild-type CaSR. The affinity of the WT CaSR for calcium was three times greater than that of the heterodimer. The maximal functional response to [Ca]o was dependent on localization of CaSR to the membrane level and independent of homo- or heterodimerizations. In summary, these results suggest that heterodimerization of WT and mCaSR receptors, rescues the trafficking defect of the mutant receptors and also reduces the affinity of the WT-mutant heterodimer for [Ca]o. In contrast, the homozygous mutants do not produce functional receptors on cell membrane. These data indicate how substantial differences between signaling of hetero- and homodimeric mutants may lead to profound differences in the severity of disease in heterozygous and homozygous carriers of these mutations.
Collapse
Affiliation(s)
- Xiaohua Wang
- Division of Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, Oregon, United States of America
| | - James Lundblad
- Division of Endocrinology and Diabetes, Oregon Health and Science University, Portland, Oregon, United States of America
- Section of Endocrinology and Diabetes, VA Portland Health Care System, Portland, Oregon, United States of America
| | - Stephen M. Smith
- Division of Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, Oregon, United States of America
- Sections of Pulmonary and Critical Care Medicine and Research & Development, VA Portland Health Care System, Portland, Oregon, United States of America
- * E-mail:
| |
Collapse
|
7
|
Bridges LE, Williams CL, Awumey EM. High Salt Upregulates Ca 2+-Sensing Receptor Expression and Ca 2+-Induced Relaxation of Contracted Mesenteric Arteries from Dahl Salt-Sensitive Rats. J Pharmacol Exp Ther 2022; 381:120-128. [PMID: 35306475 PMCID: PMC9048267 DOI: 10.1124/jpet.121.001034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 03/01/2022] [Indexed: 11/22/2022] Open
Abstract
High Ca2+ lowers blood pressure in hypertension, but the mechanism is not clear. The missing link may be the perivascular sensory nerve Ca2+-sensing receptor (CaSR) that mediates a vasodilator system after activation by interstitial Ca2+ Our results show that high salt increased CaSR expression in mesenteric arteries as well as Ca2+ relaxation of contracted mesenteric arteries from salt-sensitive (SS) rats. The CaSR was expressed as a doublet (≈120-150 kDa) in arteries from animals fed a high-salt diet for 1-4 weeks. The higher molecular weight glycosylated protein increased in arteries from SS animals; however, expression of the low molecular mass high-mannose protein decreased over 4 weeks of feeding the diet. In tissues from salt-resistant (SR) rats, the diet decreased CaSR expression after 4 weeks. Ca2+ relaxation of mesenteric arteries under phenylephrine tone increased in SS rats but decreased in arteries from SR rats fed the high-salt diet. Ca2+-activated K+ channels have a larger role in Ca2+ relaxation of arteries in SR than SS rats. The data suggest that high salt epigenetically regulates the receptor at the translational level in vivo and that the in vitro effect of Ca2+ is on receptor trafficking and signaling. In conclusion, upregulated expression of the CaSR in salt sensitivity increased receptor-mediated vascular relaxation. These findings show that CaSR signaling may compensate for changes in the vasculature in salt-sensitive hypertension. SIGNIFICANCE STATEMENT: The perivascular sensory nerve Ca2+-sensing receptor (CaSR) mediates Ca2+ relaxation of isolated mesenteric arteries under tension. This receptor may therefore play a significant role in relaxation of resistance arteries in vivo, thus explaining the blood pressure-lowering effect of dietary Ca2+. The present studies describe the effect of high salt-induced upregulation of the CaSR in salt-sensitive rats and the roles played by Ca2+-activated K+ channels and nitric oxide in Ca2+ responses.
Collapse
Affiliation(s)
- Lakeesha E Bridges
- Julius L. Chambers Biomedical and Biotechnology Research Institute (L.E.B., C.L.W., E.M.A.) and Department of Biological and Biomedical Sciences (E.M.A.), North Carolina Central University, Durham, North Carolina; and Department of Physiology and Pharmacology and Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston Salem, North Carolina (E.M.A.)
| | - Cicely L Williams
- Julius L. Chambers Biomedical and Biotechnology Research Institute (L.E.B., C.L.W., E.M.A.) and Department of Biological and Biomedical Sciences (E.M.A.), North Carolina Central University, Durham, North Carolina; and Department of Physiology and Pharmacology and Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston Salem, North Carolina (E.M.A.)
| | - Emmanuel M Awumey
- Julius L. Chambers Biomedical and Biotechnology Research Institute (L.E.B., C.L.W., E.M.A.) and Department of Biological and Biomedical Sciences (E.M.A.), North Carolina Central University, Durham, North Carolina; and Department of Physiology and Pharmacology and Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston Salem, North Carolina (E.M.A.)
| |
Collapse
|
8
|
Mullin BH, Pavlos NJ, Brown SJ, Walsh JP, McKellar RA, Wilson SG, Ward BK. Functional Assessment of Calcium-Sensing Receptor Variants Confirms Familial Hypocalciuric Hypercalcaemia. J Endocr Soc 2022; 6:bvac025. [PMID: 35356007 PMCID: PMC8962451 DOI: 10.1210/jendso/bvac025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Indexed: 11/26/2022] Open
Abstract
Context In the clinic it is important to differentiate primary hyperparathyroidism (PHPT) from the more benign, inherited disorder, familial hypocalciuric hypercalcemia (FHH). Since the conditions may sometimes overlap biochemically, identification of calcium-sensing receptor (CASR) gene variants causative of FHH (but not PHPT) is the most decisive diagnostic aid. When novel variants are identified, bioinformatics and functional assessment are required to establish pathogenicity. Objective We identified 3 novel CASR transmembrane domain missense variants, Thr699Asn, Arg701Gly, and Thr808Pro, in 3 probands provisionally diagnosed with FHH and examined the variants using bioinformatics and functional analysis. Methods Bioinformatics assessment utilized wANNOVAR software. For functional characterization, each variant was cloned into a mammalian expression vector; wild-type and variant receptors were transfected into HEK293 cells, and their expression and cellular localization were assessed by Western blotting and confocal immunofluorescence, respectively. Receptor activation in HEK293 cells was determined using an IP-One ELISA assay following stimulation with Ca++ ions. Results Bioinformatics analysis of the variants was unable to definitively assign pathogenicity. Compared with wild-type receptor, all variants demonstrated impaired expression of mature receptor reaching the cell surface and diminished activation at physiologically relevant Ca++ concentrations. Conclusion Three CASR missense variants identified in probands provisionally diagnosed with FHH result in receptor inactivation and are therefore likely causative of FHH. Inactivation may be due to inadequate processing/trafficking of mature receptor and/or conformational changes induced by the variants affecting receptor signaling. This study demonstrates the value of functional studies in assessing genetic variants identified in hypercalcemic patients.
Collapse
Affiliation(s)
- Benjamin H Mullin
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA, Australia
| | - Nathan J Pavlos
- School of Biomedical Sciences, University of Western Australia, Nedlands, WA, Australia
| | - Suzanne J Brown
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - John P Walsh
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
- Medical School, University of Western Australia, Nedlands, WA, Australia
| | - Ross A McKellar
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Scott G Wilson
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, 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
| | - Bryan K Ward
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
- Harry Perkins Institute of Medical Research, Centre for Medical Research, QEII Medical Centre, University of Western Australia, Nedlands, WA, Australia
| |
Collapse
|
9
|
Beasley HK, Widatalla SE, Whalen DS, Williams SD, Korolkova OY, Namba C, Pratap S, Ochieng J, Sakwe AM. Identification of MAGEC2/CT10 as a High Calcium-Inducible Gene in Triple-Negative Breast Cancer. Front Endocrinol (Lausanne) 2022; 13:816598. [PMID: 35355564 PMCID: PMC8959981 DOI: 10.3389/fendo.2022.816598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/17/2022] [Indexed: 11/29/2022] Open
Abstract
The expression of the melanoma/cancer-testis antigen MAGEC2/CT10 is restricted to germline cells, but like most cancer-testis antigens, it is frequently upregulated in advanced breast tumors and other malignant tumors. However, the physiological cues that trigger the expression of this gene during malignancy remain unknown. Given that malignant breast cancer is often associated with skeletal metastasis and co-morbidities such as cancer-induced hypercalcemia, we evaluated the effect of high Ca2+ on the calcium-sensing receptor (CaSR) and potential mechanisms underlying the survival of triple-negative breast cancer (TNBC) cells at high Ca2+. We show that chronic exposure of TNBC cells to high Ca2+ decreased the sensitivity of CaSR to Ca2+ but stimulated tumor cell growth and migration. Furthermore, high extracellular Ca2+ also stimulated the expression of early response genes such as FOS/FOSB and a unique set of genes associated with malignant tumors, including MAGEC2. We further show that the MAGEC2 proximal promoter is Ca2+ inducible and that FOS/FOSB binds to this promoter in a Ca2+- dependent manner. Finally, downregulation of MAGEC2 strongly inhibited the growth of TNBC cells in vitro. These data suggest for the first time that MAGEC2 is a high Ca2+ inducible gene and that aberrant expression of MAGEC2 in malignant TNBC tissues is at least in part mediated by an increase in circulating Ca2+via the AP-1 transcription factor.
Collapse
Affiliation(s)
- Heather K. Beasley
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Sarrah E. Widatalla
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Diva S. Whalen
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Stephen D. Williams
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Olga Y. Korolkova
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Clementine Namba
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Siddharth Pratap
- Bioinformatics Core, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Josiah Ochieng
- Bioinformatics Core, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
| | - Amos M. Sakwe
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN, United States
- *Correspondence: Amos M. Sakwe,
| |
Collapse
|
10
|
Orduña-Castillo LB, Del-Río-Robles JE, García-Jiménez I, Zavala-Barrera C, Beltrán-Navarro YM, Hidalgo-Moyle JJ, Ramírez-Rangel I, Hernández-Bedolla MA, Reyes-Ibarra AP, Valadez-Sánchez M, Vázquez-Prado J, Reyes-Cruz G. Calcium sensing receptor stimulates breast cancer cell migration via the Gβγ-AKT-mTORC2 signaling pathway. J Cell Commun Signal 2021; 16:239-252. [PMID: 34854057 DOI: 10.1007/s12079-021-00662-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 11/21/2021] [Indexed: 10/19/2022] Open
Abstract
Calcium sensing receptor, a pleiotropic G protein coupled receptor, activates secretory pathways in cancer cells and putatively exacerbates their metastatic behavior. Here, we show that various CaSR mutants, identified in breast cancer patients, differ in their ability to stimulate Rac, a small Rho GTPase linked to cytoskeletal reorganization and cell protrusion, but are similarly active on the mitogenic ERK pathway. To investigate how CaSR activates Rac and drives cell migration, we used invasive MDA-MB-231 breast cancer cells. We revealed, by pharmacological and knockdown strategies, that CaSR activates Rac and cell migration via the Gβγ-PI3K-mTORC2 pathway. These findings further support current efforts to validate CaSR as a relevant therapeutic target in metastatic cancer.
Collapse
Affiliation(s)
- Lennis Beatriz Orduña-Castillo
- Department of Cell Biology, CINVESTAV, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, Mexico City, Mexico
| | - Jorge Eduardo Del-Río-Robles
- Department of Cell Biology, CINVESTAV, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, Mexico City, Mexico
| | - Irving García-Jiménez
- Department of Cell Biology, CINVESTAV, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, Mexico City, Mexico
| | - César Zavala-Barrera
- Department of Cell Biology, CINVESTAV, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, Mexico City, Mexico
| | | | - Joseline Janai Hidalgo-Moyle
- Department of Cell Biology, CINVESTAV, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, Mexico City, Mexico
| | | | - Marco A Hernández-Bedolla
- Department of Cell Biology, CINVESTAV, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, Mexico City, Mexico.,Licenciatura en Enfermería, Escuela Superior de Huejutla, Universidad Autónoma del Estado de Hidalgo, Huejutla de Reyes, Hidalgo, México
| | - Alma P Reyes-Ibarra
- Department of Cell Biology, CINVESTAV, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, Mexico City, Mexico
| | - Margarita Valadez-Sánchez
- Department of Cell Biology, CINVESTAV, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, Mexico City, Mexico
| | | | - Guadalupe Reyes-Cruz
- Department of Cell Biology, CINVESTAV, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, CP 07360, Mexico City, Mexico.
| |
Collapse
|
11
|
Salinity-dependent expression of calcium-sensing receptors in Atlantic salmon (Salmo salar) tissues. J Comp Physiol A Neuroethol Sens Neural Behav Physiol 2021; 207:505-522. [PMID: 34114081 DOI: 10.1007/s00359-021-01493-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 05/23/2021] [Accepted: 05/26/2021] [Indexed: 10/21/2022]
Abstract
Multiple reports suggest that calcium-sensing receptors (CaSRs) are involved in calcium homeostasis, osmoregulation, and/or salinity sensing in fish (Loretz 2008, Herberger and Loretz 2013). We have isolated three unique full-length CaSR cDNAs from Atlantic salmon (Salmo salar) kidney that share many features with other reported CaSRs. Using anti-CaSR antibodies and PCR primers specific for individual salmon CaSR transcripts we show expression in osmoregulatory, neuroendocrine and sensory tissues. Furthermore, CaSRs are expressed in different patterns in salmon tissues where mRNA and protein expression are modified by freshwater or seawater acclimation. For example, in seawater, CaSR mRNA and protein expression is increased significantly in kidney as compared to freshwater. Electrophysiological recordings of olfactory responses produced upon exposure of salmon olfactory epithelium to CaSR agonists suggest a role for CaSRs in chemoreception in this species consistent with other freshwater, anadromous, and marine species where similar olfactory responses to divalent and polyvalent cations have been reported. These data provide further support for a role of CaSR proteins in osmoregulatory and sensory functions in Atlantic salmon, an anadromous species that experiences a broad range of environmental salinities in its life history.
Collapse
|
12
|
Huang A, Binmahfouz L, Hancock DP, Anderson PH, Ward DT, Conigrave AD. Calcium-Sensing Receptors Control CYP27B1-Luciferase Expression: Transcriptional and Posttranscriptional Mechanisms. J Endocr Soc 2021; 5:bvab057. [PMID: 34337274 PMCID: PMC8317635 DOI: 10.1210/jendso/bvab057] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Indexed: 12/19/2022] Open
Abstract
25-hydroxyvitamin D 1α-hydroxylase (encoded by CYP27B1), which catalyzes the synthesis of 1,25-dihydroxyvitamin D3, is subject to negative or positive modulation by extracellular Ca2+ (Ca2+o) depending on the tissue. However, the Ca2+ sensors and underlying mechanisms are unidentified. We tested whether calcium-sensing receptors (CaSRs) mediate Ca2+o-dependent control of 1α-hydroxylase using HEK-293 cells stably expressing the CaSR (HEK-CaSR cells). In HEK-CaSR cells, but not control HEK-293 cells, cotransfected with reporter genes for CYP27B1-Photinus pyralis (firefly) luciferase and control Renilla luciferase, an increase in Ca2+o from 0.5mM to 3.0mM induced a 2- to 3-fold increase in firefly luciferase activity as well as mRNA and protein levels. Surprisingly, firefly luciferase was specifically suppressed at Ca2+o ≥ 5.0mM, demonstrating biphasic Ca2+o control. Both phases were mediated by CaSRs as revealed by positive and negative modulators. However, Ca2+o induced simple monotonic increases in firefly luciferase and endogenous CYP27B1 mRNA levels, indicating that the inhibitory effect of high Ca2+o was posttranscriptional. Studies with inhibitors and the CaSR C-terminal mutant T888A identified roles for protein kinase C (PKC), phosphorylation of T888, and extracellular regulated protein kinase (ERK)1/2 in high Ca2+o-dependent suppression of firefly luciferase. Blockade of both PKC and ERK1/2 abolished Ca2+o-stimulated firefly luciferase, demonstrating that either PKC or ERK1/2 is sufficient to stimulate the CYP27B1 promoter. A key CCAAT box (−74 bp to −68 bp), which is regulated downstream of PKC and ERK1/2, was required for both basal transcription and Ca2+o-mediated transcriptional upregulation. The CaSR mediates Ca2+o-dependent transcriptional upregulation of 1α-hydroxylase and an additional CaSR-mediated mechanism is identified by which Ca2+o can promote luciferase and possibly 1α-hydroxylase breakdown.
Collapse
Affiliation(s)
- Alice Huang
- School of Life and Environmental Sciences, Charles Perkins Centre (D17), University of Sydney, NSW 2006Australia
| | - Lenah Binmahfouz
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Dale P Hancock
- School of Life and Environmental Sciences, Charles Perkins Centre (D17), University of Sydney, NSW 2006Australia
| | - Paul H Anderson
- Clinical and Health Sciences, Health and Biomedical Innovation, University of South Australia, Adelaide, SA, 5001, Australia
| | - Donald T Ward
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK
| | - Arthur D Conigrave
- School of Life and Environmental Sciences, Charles Perkins Centre (D17), University of Sydney, NSW 2006Australia
| |
Collapse
|
13
|
Wen T, Wang Z, Chen X, Ren Y, Lu X, Xing Y, Lu J, Chang S, Zhang X, Shen Y, Yang X. Structural basis for activation and allosteric modulation of full-length calcium-sensing receptor. SCIENCE ADVANCES 2021; 7:7/23/eabg1483. [PMID: 34088669 PMCID: PMC8177707 DOI: 10.1126/sciadv.abg1483] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/19/2021] [Indexed: 06/12/2023]
Abstract
Calcium-sensing receptor (CaSR) is a class C G protein-coupled receptor (GPCR) that plays an important role in calcium homeostasis and parathyroid hormone secretion. Here, we present multiple cryo-electron microscopy structures of full-length CaSR in distinct ligand-bound states. Ligands (Ca2+ and l-tryptophan) bind to the extracellular domain of CaSR and induce large-scale conformational changes, leading to the closure of two heptahelical transmembrane domains (7TMDs) for activation. The positive modulator (evocalcet) and the negative allosteric modulator (NPS-2143) occupy the similar binding pocket in 7TMD. The binding of NPS-2143 causes a considerable rearrangement of two 7TMDs, forming an inactivated TM6/TM6 interface. Moreover, a total of 305 disease-causing missense mutations of CaSR have been mapped to the structure in the active state, creating hotspot maps of five clinical endocrine disorders. Our results provide a structural framework for understanding the activation, allosteric modulation mechanism, and disease therapy for class C GPCRs.
Collapse
Affiliation(s)
- Tianlei Wen
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, China
| | - Ziyu Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, China
| | - Xiaozhe Chen
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, China
| | - Yue Ren
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, China
| | - Xuhang Lu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, China
| | - Yangfei Xing
- State Key Laboratory of Medical Genomics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Jing Lu
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, China
| | - Shenghai Chang
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Center of Cryo-Electron Microscopy, Zhejiang University School of Medicine, Hangzhou, China
| | - Xing Zhang
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Center of Cryo-Electron Microscopy, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuequan Shen
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, China.
- Synergetic Innovation Center of Chemical Science and Engineering, Tianjin 300071, China
| | - Xue Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Life Sciences, Nankai University, Tianjin 300350, China.
| |
Collapse
|
14
|
Liu H, Zhou D, Liu C, Zhuan Q, Luo Y, Mo X, Fu X, Hou Y. The Calcium-Sensing Receptor Is Involved in Follicle-Stimulating Hormone-Induced Cumulus Expansion in in vitro Cultured Porcine Cumulus-Oocyte Complexes. Front Cell Dev Biol 2021; 9:625036. [PMID: 34095106 PMCID: PMC8173154 DOI: 10.3389/fcell.2021.625036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 04/07/2021] [Indexed: 12/02/2022] Open
Abstract
The Calcium-Sensing Receptor (CASR) is a G protein-coupled receptor of the C family that reportedly promotes maturation of porcine oocytes. However, its role in cumulus expansion of cumulus-oocyte complexes (COCs) is not well known. This study was conducted to determine the role of CASR and potential mechanisms involved during in vitro maturation (IVM) of porcine COCs. After culture of COCs in follicle-stimulating hormone (FSH)-supplement maturation medium for 24 h, the time of breakdown of the germinal vesicle (GVBD), indicative of initiation of meiotic maturation, resulted in an increased (p < 0.05) CASR mRNA expression level in cumulus cells. Moreover, IVM of COCs in 10 μM of the CASR agonist NPS R-568 promoted (p < 0.05) cumulus expansion but only in FSH-containing medium. Conversely, 20 μM of the CASR inhibitor NPS2390 precluded cumulus expansion. We next tested the effect of the CASR agonist/inhibitor on the expression of cumulus expansion-related genes. The CASR agonist significantly upregulated the expression of hyaluronan acid synthase 2 (HAS2), whereas the CASR inhibitor downregulated the expression of all HAS2, prostaglandin-endoperoxide synthase 2 (PTGS2), and tumor necrosis factor a-induced protein 6 (TNFAIP6). Altogether, these results suggest that CASR activity is involved in FSH-stimulated porcine cumulus expansion.
Collapse
Affiliation(s)
- Huage Liu
- Institute of Reproductive Medicine, Nantong University, Nantong, China.,State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Dan Zhou
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Cong Liu
- School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Qingrui Zhuan
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yan Luo
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Xianhong Mo
- College of Life Sciences, Chifeng University, Chifeng, China
| | - Xiangwei Fu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yunpeng Hou
- State Key Laboratory of Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
15
|
Tan RSG, Lee CHL, Dimke H, Todd Alexander R. The role of calcium-sensing receptor signaling in regulating transepithelial calcium transport. Exp Biol Med (Maywood) 2021; 246:2407-2419. [PMID: 33926258 DOI: 10.1177/15353702211010415] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The calcium-sensing receptor (CaSR) plays a critical role in sensing extracellular calcium (Ca2+) and signaling to maintain Ca2+ homeostasis. In the parathyroid, the CaSR regulates secretion of parathyroid hormone, which functions to increase extracellular Ca2+ levels. The CaSR is also located in other organs imperative to Ca2+ homeostasis including the kidney and intestine, where it modulates Ca2+ reabsorption and absorption, respectively. In this review, we describe CaSR expression and its function in transepithelial Ca2+ transport in the kidney and intestine. Activation of the CaSR leads to G protein dependent and independent signaling cascades. The known CaSR signal transduction pathways involved in modulating paracellular and transcellular epithelial Ca2+ transport are discussed. Mutations in the CaSR cause a range of diseases that manifest in altered serum Ca2+ levels. Gain-of-function mutations in the CaSR result in autosomal dominant hypocalcemia type 1, while loss-of-function mutations cause familial hypocalciuric hypercalcemia. Additionally, the putative serine protease, FAM111A, is discussed as a potential regulator of the CaSR because mutations in FAM111A cause Kenny Caffey syndrome type 2, gracile bone dysplasia, and osteocraniostenosis, diseases that are characterized by hypocalcemia, hypoparathyroidism, and bony abnormalities, i.e. share phenotypic features of autosomal dominant hypocalcemia. Recent work has helped to elucidate the effect of CaSR signaling cascades on downstream proteins involved in Ca2+ transport across renal and intestinal epithelia; however, much remains to be discovered.
Collapse
Affiliation(s)
- Rebecca Siu Ga Tan
- Department of Physiology, University of Alberta, Edmonton T6G 1C9, Canada.,Membrane Protein Disease Research Group, University of Alberta, Edmonton T6G 1C9, Canada
| | | | - Henrik Dimke
- Department of Cardiovascular and Renal Research, Institute of Molecular Medicine, University of Southern Denmark, Odense 5000, Denmark.,Department of Nephrology, Odense University Hospital, Odense 5000, Denmark
| | - R Todd Alexander
- Department of Physiology, University of Alberta, Edmonton T6G 1C9, Canada.,Membrane Protein Disease Research Group, University of Alberta, Edmonton T6G 1C9, Canada.,Department of Pediatrics, University of Alberta, Edmonton T6G 1C9, Canada
| |
Collapse
|
16
|
Moon JE, Yang HY, Wee G, ParK SH, Ko CW. A cell function study on calcium regulation of a novel calcium-sensing receptor mutation (p.Tyr825Phe). Ann Pediatr Endocrinol Metab 2021; 26:24-30. [PMID: 32871647 PMCID: PMC8026336 DOI: 10.6065/apem.2040022.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 03/30/2020] [Indexed: 11/21/2022] Open
Abstract
PURPOSE Autosomal dominant hypocalcemia with hypercalciuria is a genetic disease characterized by hypoparathyroidism with hypercalciuria. We discovered a novel variant (p.Tyr825Phe[Y825F]) of the CASR gene in a neonate with congenital hypoparathyroidism and hypercalciuria and conducted a cell function study to determine whether the CASR-Y825F variant was pathogenic. METHODS To perform a functional study on CaSR-Y825F, we constructed expression vectors expressing wild-type (WT) CASR and CASR-Y825F. After transfection of each expression vector into HEK293 cells, we examined alterations in intracellular signaling. Mitogen-activated protein kinase (MAPK) signaling activity of HEK293 cells expressing CASR-WT or CASR-Y825F was determined. Changes in intracellular calcium ions ([Ca2+]i) by extracellular calcium ion ([Ca2+]e) stimulation were quantitatively compared and analyzed. RESULTS Cells expressing CASR-Y825F showed elevated of MAPK signaling (phospho-ERK [pERK], phospho-JNK [pJNK], phospho-p38 [pp38]) and increased [Ca2+]i levels at low [Ca2+]e stimulation compared with cells expressing CASR-WT. Additionally, [Ca2+]i levels in HEK293 cells expression CASR-WT and CASR-Y825F were determined at 340 nm/380 nm wavelength ratios using Fura-2 AM. At [Ca2+]e concentrations of 2.5 mM and 3 mM, the ratios of CASR-Y825F cells were higher (2.6 and 3.5, respectively) than those of CASR-WT cells (1.04 and 1.40, respectively). CONCLUSION This cell function study proved that the CASR-Y825F expressed in HEK293 cells elevated MAPK signaling (pERK, pJNK, pp38) and increased [Ca2+]i to induce hypocalcemia.
Collapse
Affiliation(s)
- Jung Eun Moon
- Department of Pediatrics, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Hee-Young Yang
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, Korea
| | - Gabbine Wee
- Laboratory Animal Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu, Korea
| | - Suk-Hyun ParK
- Department of Pediatrics, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Cheol Woo Ko
- Department of Pediatrics, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Korea,Address for correspondence: Cheol Woo Ko Department of Pediatric Endocrinology, Kyungpook National University Children's Hospital, 807, Hoguk-ro, Buk-gu, Daegu 41404, Korea
| |
Collapse
|
17
|
Höppner J, Lais S, Roll C, Wegener-Panzer A, Wieczorek D, Högler W, Grasemann C. Case Report: Severe Neonatal Course in Paternally Derived Familial Hypocalciuric Hypercalcemia. Front Endocrinol (Lausanne) 2021; 12:700612. [PMID: 34659108 PMCID: PMC8518617 DOI: 10.3389/fendo.2021.700612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 09/06/2021] [Indexed: 12/23/2022] Open
Abstract
Familial hypocalciuric hypercalcemia (FHH, [OMIM #145980]) is recognized as a benign endocrine condition affecting PTH and calcium levels due to heterozygous inactivating mutations in the calcium sensing receptor (CaSR). The condition is often un- or misdiagnosed but may have a prevalence as high as 74 in 100.000. Here, the neonatal courses of two brothers with paternally inherited FHH (CaSR c.554G>A; p.(Arg185Gln)) are described. The older brother was born preterm at 25 weeks gestation with hypercalcemia and hyperparathyroidism. The younger brother, born full-term, had severe hyperparathyroidism, muscular hypotonia, thrombocytopenia, failure to thrive and multiple metaphyseal fractures. Treatment with cinacalcet was initiated, which resulted in subsequent reduction of PTH levels and prompt clinical improvement. While it is known that homozygous mutations in CaSR may lead to life-threatening forms of neonatal severe hyperparathyroidism (NSHPT), few reports have described a severe clinical course in neonates with FHH due to heterozygous mutations. However, based on the pathophysiological framework, in de novo or paternally transmitted FHH the differing calcium needs of mother and fetus can be expected to induce fetal hyperparathyroidism and may result in severe perinatal complications as described in this report. In summary, FHH is a mostly benign condition, but transient neonatal hyperparathyroidism may occur in affected neonates if the mutation is paternally inherited. If severe, the condition can be treated successfully with cinacalcet. Patients with FHH should be informed about the risk of neonatal disease manifestation in order to monitor pregnancies and neonates.
Collapse
MESH Headings
- Calcium/metabolism
- Heat-Shock Proteins/genetics
- Humans
- Hypercalcemia/complications
- Hypercalcemia/congenital
- Hyperparathyroidism, Primary/etiology
- Hyperparathyroidism, Primary/metabolism
- Hyperparathyroidism, Primary/pathology
- Infant, Newborn
- Infant, Newborn, Diseases/etiology
- Infant, Newborn, Diseases/metabolism
- Infant, Newborn, Diseases/pathology
- Male
- Mutation
- Paternal Inheritance
- Prognosis
- Scavenger Receptors, Class A/genetics
Collapse
Affiliation(s)
- Jakob Höppner
- Department of Pediatrics, St Josef-Hospital Bochum, Ruhr-University Bochum, Bochum, Germany
| | - Sabrina Lais
- Department of Neonatology, Pediatric Intensive Care and Sleep Medicine, Vestische Kinder- und Jugendklinik Datteln, University Witten/Herdecke, Datteln, Germany
| | - Claudia Roll
- Department of Neonatology, Pediatric Intensive Care and Sleep Medicine, Vestische Kinder- und Jugendklinik Datteln, University Witten/Herdecke, Datteln, Germany
| | - Andreas Wegener-Panzer
- Department of Radiology, Vestische Kinder- und Jugendklinik Datteln, University Witten/Herdecke, Datteln, Germany
| | - Dagmar Wieczorek
- Institute of Human Genetics, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Wolfgang Högler
- Department of Paediatrics and Adolescent Medicine, Johannes Kepler University Linz, Linz, Austria
| | - Corinna Grasemann
- Department of Pediatrics, St Josef-Hospital Bochum, Ruhr-University Bochum, Bochum, Germany
- *Correspondence: Corinna Grasemann,
| |
Collapse
|
18
|
Ramezani A, Rasaee MJ, Jalaeefar A, Salmanian AH. Efficient detection of eukaryotic calcium-sensing receptor (CaSR) by polyclonal antibody against prokaryotic expressed truncated CaSR. Mol Biol Rep 2020; 47:7723-7734. [PMID: 33001312 DOI: 10.1007/s11033-020-05847-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Accepted: 09/15/2020] [Indexed: 11/26/2022]
Abstract
Calcium-sensing receptor (CaSR), which is better known for its action as regulating calcium homeostasis, can bind various ligands. To facilitate research on CaSR and understand the receptor's function further, an in silico designed truncated protein was developed. The resulting protein folding indicated that 99% of predicted three dimensional (3D) structure residues are located in favored and allowed Ramachandran plots. However, it was found that such protein does not fold properly when expressed in prokaryotic host cells. Thioredoxin (Trx) tag was conjugated to increase the final protein's solubility, which could help obtain the soluble antigen with better immunogenic properties. The truncated recombinant proteins were expressed and purified in two forms (Trx-CaSR: RR19 and CaSR: RRJ19). The polyclonal antibody was induced by the rabbit immunization with the form of RR19. Western blot on mouse kidney lysates evidenced the proper immune recognition of the receptor by the produced antibody. The specificity and sensitivity of antibodies were also assayed by immunohistofluorescence. These experiments affirmed antibody's ability to indicate the receptor on the cell surface in native form and the possibility of applying such antibodies in further cellular and tissue assays.
Collapse
Affiliation(s)
- Aghdas Ramezani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, PO Box 14115-331, Tehran, Iran
| | - Mohammad Javad Rasaee
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, PO Box 14115-331, Tehran, Iran.
| | - Amirmohsen Jalaeefar
- Department of Surgical Oncology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Hatef Salmanian
- Departments of Plant Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| |
Collapse
|
19
|
Lee JH, Davaatseren M, Lee S. Rare PTH Gene Mutations Causing Parathyroid Disorders: A Review. Endocrinol Metab (Seoul) 2020; 35:64-70. [PMID: 32207265 PMCID: PMC7090289 DOI: 10.3803/enm.2020.35.1.64] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/27/2020] [Accepted: 03/05/2020] [Indexed: 11/11/2022] Open
Abstract
Since parathyroid hormone (PTH) was first isolated and its gene (PTH) was sequenced, only eight PTH mutations have been discovered. The C18R mutation in PTH, discovered in 1990, was the first to be reported. This autosomal dominant mutation induces endoplasmic reticulum stress and subsequent apoptosis in parathyroid cells. The next mutation, which was reported in 1992, is associated with exon skipping. The substitution of G with C in the first nucleotide of the second intron results in the exclusion of the second exon; since this exon includes the initiation codon, translation initiation is prevented. An S23P mutation and an S23X mutation at the same residue were reported in 1999 and 2012, respectively. Both mutations resulted in hypoparathyroidism. In 2008, a somatic R83X mutation was detected in a parathyroid adenoma tissue sample collected from a patient with hyperparathyroidism. In 2013, a heterozygous p.Met1_Asp6del mutation was incidentally discovered in a case-control study. Two years later, the R56C mutation was reported; this is the only reported hypoparathyroidism-causing mutation in the mature bioactive part of PTH. In 2017, another heterozygous mutation, M14K, was detected. The discovery of these eight mutations in the PTH gene has provided insights into its function and broadened our understanding of the molecular mechanisms underlying mutation progression. Further attempts to detect other such mutations will help elucidate the functions of PTH in a more sophisticated manner.
Collapse
Affiliation(s)
- Joon Hyop Lee
- Laboratory of Genomics and Translational Medicine, Gachon University College of Medicine, Incheon, Korea
- Department of Surgery, Gachon University College of Medicine, Incheon, Korea
| | - Munkhtugs Davaatseren
- Laboratory of Genomics and Translational Medicine, Gachon University College of Medicine, Incheon, Korea
| | - Sihoon Lee
- Laboratory of Genomics and Translational Medicine, Gachon University College of Medicine, Incheon, Korea
- Department of Internal Medicine, Gachon University College of Medicine, Incheon, Korea.
| |
Collapse
|
20
|
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
|
21
|
Chen Y, Gao Y, Tao Y, Lin D, An S. Identification of a Calcium-sensing Receptor in Human Dental Pulp Cells That Regulates Mineral Trioxide Aggregate–induced Mineralization. J Endod 2019; 45:907-916. [DOI: 10.1016/j.joen.2019.03.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 02/28/2019] [Accepted: 03/28/2019] [Indexed: 12/23/2022]
|
22
|
Sensory Axon Growth Requires Spatiotemporal Integration of CaSR and TrkB Signaling. J Neurosci 2019; 39:5842-5860. [PMID: 31123102 DOI: 10.1523/jneurosci.0027-19.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 05/12/2019] [Accepted: 05/15/2019] [Indexed: 12/18/2022] Open
Abstract
Neural circuit development involves the coordinated growth and guidance of axons. During this process, axons encounter many different cues, but how these cues are integrated and translated into growth is poorly understood. In this study, we report that receptor signaling does not follow a linear path but changes dependent on developmental stage and coreceptors involved. Using developing chicken embryos of both sexes, our data show that calcium-sensing receptor (CaSR), a G-protein-coupled receptor important for regulating calcium homeostasis, regulates neurite growth in two distinct ways. First, when signaling in isolation, CaSR promotes growth through the PI3-kinase-Akt pathway. At later developmental stages, CaSR enhances tropomyosin receptor kinase B (TrkB)/BDNF-mediated neurite growth. This enhancement is facilitated through a switch in the signaling cascade downstream of CaSR (i.e., from the PI3-kinase-Akt pathway to activation of GSK3α Tyr279). TrkB and CaSR colocalize within late endosomes, cotraffic and coactivate GSK3, which serves as a shared signaling node for both receptors. Our study provides evidence that two unrelated receptors can integrate their individual signaling cascades toward a nonadditive effect and thus control neurite growth during development.SIGNIFICANCE STATEMENT This work highlights the effect of receptor coactivation and signal integration in a developmental setting. During embryonic development, neurites grow toward their targets guided by cues in the extracellular environment. These cues are sensed by receptors at the surface that trigger intracellular signaling events modulating the cytoskeleton. Emerging evidence suggests that the effects of guidance cues are diversified, therefore expanding the number of responses. Here, we show that two unrelated receptors can change the downstream signaling cascade and regulate neuronal growth through a shared signaling node. In addition to unraveling a novel signaling pathway in neurite growth, this research stresses the importance of receptor coactivation and signal integration during development of the nervous system.
Collapse
|
23
|
Issa H, Hénaut L, Abdallah JB, Boudot C, Lenglet G, Avondo C, Ibrik A, Caus T, Brazier M, Mentaverri R, Zibara K, Kamel S. Activation of the calcium-sensing receptor in human valvular interstitial cells promotes calcification. J Mol Cell Cardiol 2019; 129:2-12. [DOI: 10.1016/j.yjmcc.2019.01.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 12/17/2018] [Accepted: 01/24/2019] [Indexed: 01/10/2023]
|
24
|
Decmann A, Patócs A, Igaz P. Overview of Genetically Determined Diseases/Multiple Endocrine Neoplasia Syndromes Predisposing to Endocrine Tumors. EXPERIENTIA SUPPLEMENTUM (2012) 2019; 111:105-127. [PMID: 31588530 DOI: 10.1007/978-3-030-25905-1_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In this chapter, we present an overview of multiple endocrine neoplasia syndromes including their most important clinical and molecular features. Multiple endocrine neoplasia type 1 and 2 syndromes (MEN1 and MEN2) are discussed in detail. Syndromes that are presented in other chapters are only briefly mentioned. We discuss the relevance of germline gene alterations in apparently sporadic endocrine tumors, e.g., medullary thyroid cancer, primary hyperparathyroidism, and neuroendocrine tumors. McCune-Albright syndrome that only exists in non-hereditary, sporadic forms is also discussed in detail, as tumors of several endocrine organs can develop in the same individual.
Collapse
Affiliation(s)
- Abel Decmann
- 2nd Department of Internal Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Attila Patócs
- Department of Laboratory Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary
- "Lendület" Hereditary Endocrine Tumors Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
- Department of Molecular Genetics, National Institute of Oncology, Budapest, Hungary
| | - Peter Igaz
- 2nd Department of Internal Medicine, Faculty of Medicine, Semmelweis University, Budapest, Hungary.
- MTA-SE Molecular Medicine Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary.
| |
Collapse
|
25
|
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: 14] [Impact Index Per Article: 2.0] [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
|
26
|
Calcium-sensing receptor in nutrient sensing: an insight into the modulation of intestinal homoeostasis. Br J Nutr 2018; 120:881-890. [DOI: 10.1017/s0007114518002088] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AbstractThe animal gut effectively prevents the entry of hazardous substances and microbes while permitting the transfer of nutrients, such as water, electrolytes, vitamins, proteins, lipids, carbohydrates, minerals and microbial metabolites, which are intimately associated with intestinal homoeostasis. The gut maintains biological functions through its nutrient-sensing receptors, including the Ca-sensing receptor (CaSR), which activates a variety of signalling pathways, depending on cellular context. CaSR coordinates food digestion and nutrient absorption, promotes cell proliferation and differentiation, regulates energy metabolism and immune response, stimulates hormone secretion, mitigates secretory diarrhoea and enhances intestinal barrier function. Thus, CaSR is crucial to the maintenance of gut homoeostasis and protection of intestinal health. In this review, we focused on the emerging roles of CaSR in the modulation of intestinal homoeostasis including related underlying mechanisms. By elucidating the relationship between CaSR and animal gut homoeostasis, effective and inexpensive methods for treating intestinal health imbalance through nutritional manipulation can be developed. This article is expected to provide experimental data of the effects of CaSR on animal or human health.
Collapse
|
27
|
Ittisoponpisan S, David A. Structural Biology Helps Interpret Variants of Uncertain Significance in Genes Causing Endocrine and Metabolic Disorders. J Endocr Soc 2018; 2:842-854. [PMID: 30019023 PMCID: PMC6041779 DOI: 10.1210/js.2018-00077] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/08/2018] [Indexed: 12/22/2022] Open
Abstract
CONTEXT Variants of uncertain significance (VUSs) lack sufficient evidence, in terms of statistical power or experimental studies, to allow unequivocal determination of their damaging effect. VUSs are a major burden in performing genetic analysis. Although in silico prediction tools are widely used, their specificity is low, thus urgently calling for methods for prioritizing and characterizing variants. OBJECTIVE To assess the frequency of VUSs in genes causing endocrine and metabolic disorders, the concordance rate of predictions from different in silico methods, and the added value of three-dimensional protein structure analysis in discerning and prioritizing damaging variants. RESULTS A total of 12,266 missense variants reported in 641 genes causing endocrine and metabolic disorders were analyzed. Among these, 4123 (33.7%) were VUSs, of which 2010 (48.8%) were predicted to be damaging and 1452 (35.2%) were predicted to be tolerated according to in silico tools. A total of 5383 (87.7%) of 6133 disease-causing variants and 823 (55.8%) of 1474 benign variants were correctly predicted. In silico predictions were noninformative in 5.7%, 14.4%, and 16% of damaging, benign, and VUSs, respectively. A damaging effect on 3D protein structure was present in 240 (30.9%) of predicted damaging and 40 (9.7%) of predicted tolerated VUSs (P < 0.001). An in-depth analysis of nine VUSs occurring in TSHR, LDLR, CASR, and APOE showed that they greatly affect protein stability and are therefore strong candidates for disease. CONCLUSIONS In our dataset, we confirmed the high sensitivity but low specificity of in silico predictions tools. 3D protein structural analysis is a compelling tool for characterizing and prioritizing VUSs and should be a part of genetic variant analysis.
Collapse
Affiliation(s)
- Sirawit Ittisoponpisan
- Structural Bioinformatics Group, Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Alessia David
- Structural Bioinformatics Group, Department of Life Sciences, Imperial College London, London, United Kingdom
| |
Collapse
|
28
|
|
29
|
Bazúa-Valenti S, Rojas-Vega L, Castañeda-Bueno M, Barrera-Chimal J, Bautista R, Cervantes-Pérez LG, Vázquez N, Plata C, Murillo-de-Ozores AR, González-Mariscal L, Ellison DH, Riccardi D, Bobadilla NA, Gamba G. The Calcium-Sensing Receptor Increases Activity of the Renal NCC through the WNK4-SPAK Pathway. J Am Soc Nephrol 2018; 29:1838-1848. [PMID: 29848507 DOI: 10.1681/asn.2017111155] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 04/10/2018] [Indexed: 01/07/2023] Open
Abstract
Background Hypercalciuria can result from activation of the basolateral calcium-sensing receptor (CaSR), which in the thick ascending limb of Henle's loop controls Ca2+ excretion and NaCl reabsorption in response to extracellular Ca2+ However, the function of CaSR in the regulation of NaCl reabsorption in the distal convoluted tubule (DCT) is unknown. We hypothesized that CaSR in this location is involved in activating the thiazide-sensitive NaCl cotransporter (NCC) to prevent NaCl loss.Methods We used a combination of in vitro and in vivo models to examine the effects of CaSR on NCC activity. Because the KLHL3-WNK4-SPAK pathway is involved in regulating NaCl reabsorption in the DCT, we assessed the involvement of this pathway as well.Results Thiazide-sensitive 22Na+ uptake assays in Xenopus laevis oocytes revealed that NCC activity increased in a WNK4-dependent manner upon activation of CaSR with Gd3+ In HEK293 cells, treatment with the calcimimetic R-568 stimulated SPAK phosphorylation only in the presence of WNK4. The WNK4 inhibitor WNK463 also prevented this effect. Furthermore, CaSR activation in HEK293 cells led to phosphorylation of KLHL3 and WNK4 and increased WNK4 abundance and activity. Finally, acute oral administration of R-568 in mice led to the phosphorylation of NCC.Conclusions Activation of CaSR can increase NCC activity via the WNK4-SPAK pathway. It is possible that activation of CaSR by Ca2+ in the apical membrane of the DCT increases NaCl reabsorption by NCC, with the consequent, well known decrease of Ca2+ reabsorption, further promoting hypercalciuria.
Collapse
Affiliation(s)
- Silvana Bazúa-Valenti
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Lorena Rojas-Vega
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - María Castañeda-Bueno
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jonatan Barrera-Chimal
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | | | | | - Norma Vázquez
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Consuelo Plata
- Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Adrián R Murillo-de-Ozores
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Lorenza González-Mariscal
- Department of Physiology, Biophysics and Neuroscience, Center for Research and Advanced Studies (Cinvestav), Mexico City, Mexico
| | - David H Ellison
- Department of Medicine, Oregon Health and Science University, Portland, Oregon.,Renal Section, Veterans Administration Portland Health Care System, Portland, Oregon
| | - Daniela Riccardi
- School of Biosciences, Cardiff University, Cardiff, United Kingdom; and
| | - Norma A Bobadilla
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Gerardo Gamba
- Molecular Physiology Unit, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico; .,Department of Nephrology and Mineral Metabolism, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey, Nuevo León, Mexico
| |
Collapse
|
30
|
Gerbino A, Colella M. The Different Facets of Extracellular Calcium Sensors: Old and New Concepts in Calcium-Sensing Receptor Signalling and Pharmacology. Int J Mol Sci 2018; 19:E999. [PMID: 29584660 PMCID: PMC5979557 DOI: 10.3390/ijms19040999] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 03/23/2018] [Accepted: 03/25/2018] [Indexed: 12/14/2022] Open
Abstract
The current interest of the scientific community for research in the field of calcium sensing in general and on the calcium-sensing Receptor (CaR) in particular is demonstrated by the still increasing number of papers published on this topic. The extracellular calcium-sensing receptor is the best-known G-protein-coupled receptor (GPCR) able to sense external Ca2+ changes. Widely recognized as a fundamental player in systemic Ca2+ homeostasis, the CaR is ubiquitously expressed in the human body where it activates multiple signalling pathways. In this review, old and new notions regarding the mechanisms by which extracellular Ca2+ microdomains are created and the tools available to measure them are analyzed. After a survey of the main signalling pathways triggered by the CaR, a special attention is reserved for the emerging concepts regarding CaR function in the heart, CaR trafficking and pharmacology. Finally, an overview on other Ca2+ sensors is provided.
Collapse
Affiliation(s)
- Andrea Gerbino
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70121 Bari, Italy.
| | - Matilde Colella
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70121 Bari, Italy.
| |
Collapse
|
31
|
Rasmussen AQ, Jørgensen NR, Schwarz P. Identification and Functional Characterization of a Novel Mutation in the Human Calcium-Sensing Receptor That Co-Segregates With Autosomal-Dominant Hypocalcemia. Front Endocrinol (Lausanne) 2018; 9:200. [PMID: 29743878 PMCID: PMC5930847 DOI: 10.3389/fendo.2018.00200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 04/10/2018] [Indexed: 12/31/2022] Open
Abstract
The human calcium-sensing receptor (CASR) is the key controller of extracellular Cao2+ homeostasis, and different mutations in the CASR gene have been linked to different calcium diseases, such as familial hypocalciuric hypercalcemia, severe hyperparathyroidism, autosomal-dominant hypocalcemia (ADH), and Bartter's syndrome type V. In this study, two generations of a family with biochemically and clinically confirmed ADH who suffered severe muscle pain, arthralgia, tetany, abdominal pain, and fatigue were evaluated for mutations in the CASR gene. The study comprises genotyping of all family members, functional characterization of a potential mutant receptor by in vitro analysis related to the wild-type receptor to reveal an association between the genotype and phenotype in the affected family members. The in vitro analysis of functional characteristics includes measurements of inositol trisphosphate accumulation, Ca2+ mobilization in response to [Ca2+]o-stimulation and receptor expression. The results reveal a significant leftward shift of inositol trisphosphate accumulation as a result of the "gain-of-function" mutant receptor and surprisingly a normalization of the response in (Ca2+)i release in the downstream pathway and additionally the maximal response of (Ca2+)i release was significantly decreased compared to the wild type. However, no gross differences were seen in D126V and the D126V/WT CASR dimeric >250 kDa band expression compared to the WT receptor, however, the D126V and D126V/WT CASR immature ~140 kDa species appear to have reduced expression compared to the WT receptor. In conclusion, in this study, a family with a clinical diagnosis of ADH in two generations was evaluated to identify a mutation in the CASR gene and reveal an association between genotype and phenotype in the affected family members. The clinical condition was caused by a novel, activating, missense mutation (D126V) in the CASR gene and the in vitro functional characteristics of the mutation co-segregated with their individual phenotype.
Collapse
Affiliation(s)
- Anne Qvist Rasmussen
- Research Centre of Ageing and Osteoporosis, Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
- *Correspondence: Anne Qvist Rasmussen,
| | - Niklas Rye Jørgensen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Institute of Clinical Research, University of Southern, Odense, Denmark
| | - Peter Schwarz
- Research Centre of Ageing and Osteoporosis, Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health Sciences, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
32
|
Ward BK, Rea SL, Magno AL, Pedersen B, Brown SJ, Mullin S, Arulpragasam A, Ingley E, Conigrave AD, Ratajczak T. The endoplasmic reticulum-associated protein, OS-9, behaves as a lectin in targeting the immature calcium-sensing receptor. J Cell Physiol 2017; 233:38-56. [PMID: 28419469 DOI: 10.1002/jcp.25957] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 04/13/2017] [Indexed: 11/07/2022]
Abstract
The mechanisms responsible for the processing and quality control of the calcium-sensing receptor (CaSR) in the endoplasmic reticulum (ER) are largely unknown. In a yeast two-hybrid screen of the CaSR C-terminal tail (residues 865-1078), we identified osteosarcoma-9 (OS-9) protein as a binding partner. OS-9 is an ER-resident lectin that targets misfolded glycoproteins to the ER-associated degradation (ERAD) pathway through recognition of specific N-glycans by its mannose-6-phosphate receptor homology (MRH) domain. We show by confocal microscopy that the CaSR and OS-9 co-localize in the ER in COS-1 cells. In immunoprecipitation studies with co-expressed OS-9 and CaSR, OS-9 specifically bound the immature form of wild-type CaSR in the ER. OS-9 also bound the immature forms of a CaSR C-terminal deletion mutant and a C677A mutant that remains trapped in the ER, although binding to neither mutant was favored over wild-type receptor. OS-9 binding to immature CaSR required the MRH domain of OS-9 indicating that OS-9 acts as a lectin most likely to target misfolded CaSR to ERAD. Our results also identify two distinct binding interactions between OS-9 and the CaSR, one involving both C-terminal domains of the two proteins and the other involving both N-terminal domains. This suggests the possibility of more than one functional interaction between OS-9 and the CaSR. When we investigated the functional consequences of altered OS-9 expression, neither knockdown nor overexpression of OS-9 was found to have a significant effect on CaSR cell surface expression or CaSR-mediated ERK1/2 phosphorylation.
Collapse
Affiliation(s)
- Bryan K Ward
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- Laboratory for Molecular Endocrinology, Harry Perkins Institute of Medical Research and the Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Sarah L Rea
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- Laboratory for Molecular Endocrinology, Harry Perkins Institute of Medical Research and the Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Aaron L Magno
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- Laboratory for Molecular Endocrinology, Harry Perkins Institute of Medical Research and the Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Bernadette Pedersen
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- Laboratory for Molecular Endocrinology, Harry Perkins Institute of Medical Research and the Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Suzanne J Brown
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Shelby Mullin
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Ajanthy Arulpragasam
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- Laboratory for Molecular Endocrinology, Harry Perkins Institute of Medical Research and the Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Evan Ingley
- Cell Signalling Group, Harry Perkins Institute of Medical Research and the Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Arthur D Conigrave
- School of Life and Environmental Sciences, Charles Perkins Centre, University of Sydney, New South Wales, Australia
| | - Thomas Ratajczak
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
- Laboratory for Molecular Endocrinology, Harry Perkins Institute of Medical Research and the Centre for Medical Research, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
33
|
Tunç E, Demirhan O, Sağliker Y, Yildiz İ, Paylar N, Güzel Aİ. Chromosomal findings and sequence analysis of target exons of calcium-sensingreceptor (CaSR) gene in patients with Sagliker syndrome. Turk J Med Sci 2017; 47:13-21. [PMID: 28263480 DOI: 10.3906/sag-1507-102] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 01/28/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND/AIM Sagliker syndrome (SS) develops as a continuation of chronic kidney disease and secondary hyperparathyroidism conditions. It was thought that there are some genetic predisposition factors leading to SS. The calcium-sensing receptor (CaSR) is essential for calcium homeostasis in the body. We aimed to examine SS patients for chromosome aberrations (CAs) and CaSR gene abnormalities in exons 2 and 3. MATERIALS AND METHODS Twenty-three patients and 23 control subjects were admitted to Balcalı Hospital of the Medical Faculty of Çukurova University in Turkey between 2009 and 2011. Chromosomal analysis was performed according to standard cytogenetic methods. Full sequencing of exons 2 and 3 of the CaSR gene was done. RESULTS We found base alterations and deletions in exons 2 and 3 of the CaSR gene. We also found a statistically significant increase in the rate of CAs in patients compared to controls. In total we evaluated 639 metaphase plaques in 23 patients and found 241 CAs, of which 88% were structural and 12% were numerical abnormalities. CONCLUSION There is no relation between the etiology of SS and nucleotide alterations that we could find in exons 2 and 3 of the CaSR gene. Our data suggest that there may be a correlation between CAs and the progression of SS.
Collapse
Affiliation(s)
- Erdal Tunç
- Department of Medical Biology and Genetics, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Osman Demirhan
- Department of Medical Biology and Genetics, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Yahya Sağliker
- Department of Internal Medicine, Faculty of Medicine, Çukurova University, Adana, Turkey
| | | | - Nuray Paylar
- Department of Internal Medicine, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Ali İrfan Güzel
- Department of Medical Biology and Genetics, Faculty of Medicine, Recep Tayyip Erdoğan University, Rize, Turkey
| |
Collapse
|
34
|
Chéron JB, Golebiowski J, Antonczak S, Fiorucci S. The anatomy of mammalian sweet taste receptors. Proteins 2017; 85:332-341. [PMID: 27936499 DOI: 10.1002/prot.25228] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/09/2016] [Accepted: 11/30/2016] [Indexed: 01/02/2023]
Abstract
All sweet-tasting compounds are detected by a single G-protein coupled receptor (GPCR), the heterodimer T1R2-T1R3, for which no experimental structure is available. The sweet taste receptor is a class C GPCR, and the recently published crystallographic structures of metabotropic glutamate receptor (mGluR) 1 and 5 provide a significant step forward for understanding structure-function relationships within this family. In this article, we recapitulate more than 600 single point site-directed mutations and available structural data to obtain a critical alignment of the sweet taste receptor sequences with respect to other class C GPCRs. Using this alignment, a homology 3D-model of the human sweet taste receptor is built and analyzed to dissect out the role of key residues involved in ligand binding and those responsible for receptor activation. Proteins 2017; 85:332-341. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jean-Baptiste Chéron
- Université Côte d'azur, CNRS, Institut de Chimie de Nice UMR7272, 06108 Nice, France
| | - Jérôme Golebiowski
- Université Côte d'azur, CNRS, Institut de Chimie de Nice UMR7272, 06108 Nice, France
- Department of Brain and Cognitive Science, DGIST (Daegu Gyeongbuk Institute of Science & Technology), Daegu, Korea
| | - Serge Antonczak
- Université Côte d'azur, CNRS, Institut de Chimie de Nice UMR7272, 06108 Nice, France
| | - Sébastien Fiorucci
- Université Côte d'azur, CNRS, Institut de Chimie de Nice UMR7272, 06108 Nice, France
| |
Collapse
|
35
|
Colella M, Gerbino A, Hofer AM, Curci S. Recent advances in understanding the extracellular calcium-sensing receptor. F1000Res 2016; 5. [PMID: 27803801 PMCID: PMC5074356 DOI: 10.12688/f1000research.8963.1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/13/2016] [Indexed: 12/11/2022] Open
Abstract
The extracellular calcium-sensing receptor (CaR), a ubiquitous class C G-protein-coupled receptor (GPCR), is responsible for the control of calcium homeostasis in body fluids. It integrates information about external Ca
2+ and a surfeit of other endogenous ligands into multiple intracellular signals, but how is this achieved? This review will focus on some of the exciting concepts in CaR signaling and pharmacology that have emerged in the last few years.
Collapse
Affiliation(s)
- Matilde Colella
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari , Bari, Italy
| | - Andrea Gerbino
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari , Bari, Italy
| | - Aldebaran M Hofer
- Department of Surgery, Brigham & Women's Hospital, Harvard Medical School and VA Boston Healthcare System, West Roxbury, MA, USA
| | - Silvana Curci
- Department of Surgery, Brigham & Women's Hospital, Harvard Medical School and VA Boston Healthcare System, West Roxbury, MA, USA
| |
Collapse
|
36
|
Hannan FM, Babinsky VN, Thakker RV. Disorders of the calcium-sensing receptor and partner proteins: insights into the molecular basis of calcium homeostasis. J Mol Endocrinol 2016; 57:R127-42. [PMID: 27647839 PMCID: PMC5064759 DOI: 10.1530/jme-16-0124] [Citation(s) in RCA: 114] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 08/08/2016] [Indexed: 12/20/2022]
Abstract
The extracellular calcium (Ca(2+) o)-sensing receptor (CaSR) is a family C G protein-coupled receptor, which detects alterations in Ca(2+) o concentrations and modulates parathyroid hormone secretion and urinary calcium excretion. The central role of the CaSR in Ca(2+) o homeostasis has been highlighted by the identification of mutations affecting the CASR gene on chromosome 3q21.1. Loss-of-function CASR mutations cause familial hypocalciuric hypercalcaemia (FHH), whereas gain-of-function mutations lead to autosomal dominant hypocalcaemia (ADH). However, CASR mutations are only detected in ≤70% of FHH and ADH cases, referred to as FHH type 1 and ADH type 1, respectively, and studies in other FHH and ADH kindreds have revealed these disorders to be genetically heterogeneous. Thus, loss- and gain-of-function mutations of the GNA11 gene on chromosome 19p13.3, which encodes the G-protein α-11 (Gα11) subunit, lead to FHH type 2 and ADH type 2, respectively; whilst loss-of-function mutations of AP2S1 on chromosome 19q13.3, which encodes the adaptor-related protein complex 2 sigma (AP2σ) subunit, cause FHH type 3. These studies have demonstrated Gα11 to be a key mediator of downstream CaSR signal transduction, and also revealed a role for AP2σ, which is involved in clathrin-mediated endocytosis, in CaSR signalling and trafficking. Moreover, FHH type 3 has been demonstrated to represent a more severe FHH variant that may lead to symptomatic hypercalcaemia, low bone mineral density and cognitive dysfunction. In addition, calcimimetic and calcilytic drugs, which are positive and negative CaSR allosteric modulators, respectively, have been shown to be of potential benefit for these FHH and ADH disorders.
Collapse
Affiliation(s)
- Fadil M Hannan
- Academic Endocrine UnitRadcliffe Department of Medicine, University of Oxford, Oxford, UK Department of Musculoskeletal BiologyInstitute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Valerie N Babinsky
- Academic Endocrine UnitRadcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Rajesh V Thakker
- Academic Endocrine UnitRadcliffe Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
37
|
Zhang C, Miller CL, Gorkhali R, Zou J, Huang K, Brown EM, Yang JJ. Molecular Basis of the Extracellular Ligands Mediated Signaling by the Calcium Sensing Receptor. Front Physiol 2016; 7:441. [PMID: 27746744 PMCID: PMC5043022 DOI: 10.3389/fphys.2016.00441] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 09/16/2016] [Indexed: 12/20/2022] Open
Abstract
Ca2+-sensing receptors (CaSRs) play a central role in regulating extracellular calcium concentration ([Ca2+]o) homeostasis and many (patho)physiological processes in multiple organs. This regulation is orchestrated by a cooperative response to extracellular stimuli such as small changes in Ca2+, Mg2+, amino acids, and other ligands. In addition, CaSR is a pleiotropic receptor regulating several intracellular signaling pathways, including calcium mobilization and intracellular calcium oscillation. Nearly 200 mutations and polymorphisms have been found in CaSR in relation to a variety of human disorders associated with abnormal Ca2+ homeostasis. In this review, we summarize efforts directed at identifying binding sites for calcium and amino acids. Both homotropic cooperativity among multiple calcium binding sites and heterotropic cooperativity between calcium and amino acid were revealed using computational modeling, predictions, and site-directed mutagenesis coupled with functional assays. The hinge region of the bilobed Venus flytrap (VFT) domain of CaSR plays a pivotal role in coordinating multiple extracellular stimuli, leading to cooperative responses from the receptor. We further highlight the extensive number of disease-associated mutations that have also been shown to affect CaSR's cooperative action via several types of mechanisms. These results provide insights into the molecular bases of the structure and functional cooperativity of this receptor and other members of family C of the G protein-coupled receptors (cGPCRs) in health and disease states, and may assist in the prospective development of novel receptor-based therapeutics.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Chemistry, Georgia State University Atlanta, GA, USA
| | | | - Rakshya Gorkhali
- Department of Chemistry, Georgia State University Atlanta, GA, USA
| | - Juan Zou
- Department of Chemistry, Georgia State University Atlanta, GA, USA
| | - Kenneth Huang
- Department of Chemistry, Georgia State University Atlanta, GA, USA
| | - Edward M Brown
- Center for Diagnostics and Therapeutics, Georgia State UniversityAtlanta, GA, USA; Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's HospitalBoston, MA, USA
| | - Jenny J Yang
- Department of Chemistry, Georgia State University Atlanta, GA, USA
| |
Collapse
|
38
|
Mayr B, Glaudo M, Schöfl C. Activating Calcium-Sensing Receptor Mutations: Prospects for Future Treatment with Calcilytics. Trends Endocrinol Metab 2016; 27:643-652. [PMID: 27339034 DOI: 10.1016/j.tem.2016.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/17/2016] [Accepted: 05/20/2016] [Indexed: 12/20/2022]
Abstract
Activating mutations of the G protein-coupled receptor, calcium-sensing receptor (CaSR), cause autosomal dominant hypocalcemia and Bartter syndrome type 5. These mutations lower the set-point for extracellular calcium sensing, thereby causing decreased parathyroid hormone secretion and disturbed renal calcium handling with hypercalciuria. Available therapies increase serum calcium levels but raise the risk of complications in affected patients. Symptom relief and the prevention of adverse outcome is currently very difficult to achieve. Calcilytics act as CaSR antagonists that attenuate its activity, thereby correcting the molecular defect of activating CaSR proteins in vitro and elevating serum calcium in mice and humans in vivo, and have emerged as the most promising therapeutics for the treatment of these rare and difficult to treat diseases.
Collapse
Affiliation(s)
- Bernhard Mayr
- Division of Endocrinology and Diabetes, Department of Medicine I, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Germany.
| | - Markus Glaudo
- Division of Endocrinology and Diabetes, Department of Medicine I, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| | - Christof Schöfl
- Division of Endocrinology and Diabetes, Department of Medicine I, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| |
Collapse
|
39
|
Guerreiro R, Brás J, Batista S, Pires P, Ribeiro MH, Almeida MR, Oliveira C, Hardy J, Santana I. Pseudohypoparathyroidism type I-b with neurological involvement is associated with a homozygous PTH1R mutation. GENES, BRAIN, AND BEHAVIOR 2016; 15:669-77. [PMID: 27415614 PMCID: PMC5026059 DOI: 10.1111/gbb.12308] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 07/11/2016] [Accepted: 07/11/2016] [Indexed: 12/16/2022]
Abstract
Pseudohypoparathyroidism type 1b (PHP1b) is characterized by hypocalcemia, hyperphosphatemia, increased levels of circulating parathyroid hormone (PTH), and no skeletal or developmental abnormalities. The goal of this study was to perform a full characterization of a familial case of PHP1b with neurological involvement and to identify the genetic cause of disease. The initial laboratory profile of the proband showed severe hypocalcemia, hyperphosphatemia and normal levels of PTH, which was considered to be compatible with primary hypoparathyroidism. With disease progression the patient developed cognitive disturbance, PTH levels were found to be slightly elevated and a picture of PTH resistance syndrome seemed more probable. The diagnosis of PHP1b was established after the study of family members and blunted urinary cAMP results were obtained in a PTH stimulation test. Integration of whole genome genotyping and exome sequencing data supported this diagnosis by revealing a novel homozygous missense mutation in PTH1R (p.Arg186His) completely segregating with the disease. Here, we demonstrate segregation of a novel mutation in PTH1R with a phenotype of PHP1b presenting with neurological symptoms, but no bone defects. This case represents the extreme end of the spectrum of cognitive impairment in PTH dysfunction and defines a possible novel form of PHP1b resulting from the impaired interaction between PTH and PTH1R.
Collapse
Affiliation(s)
- R Guerreiro
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK.
- Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal.
| | - J Brás
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
- Department of Medical Sciences, Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal
| | - S Batista
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - P Pires
- Hospital do Santo Espírito, Terceira, Portugal
| | - M H Ribeiro
- CNC - Center for Neuroscience and Cell Biology
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - M R Almeida
- CNC - Center for Neuroscience and Cell Biology
| | - C Oliveira
- CNC - Center for Neuroscience and Cell Biology
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - J Hardy
- Department of Molecular Neuroscience, UCL Institute of Neurology, London, UK
| | - I Santana
- Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
- CNC - Center for Neuroscience and Cell Biology
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
40
|
Geng Y, Mosyak L, Kurinov I, Zuo H, Sturchler E, Cheng TC, Subramanyam P, Brown AP, Brennan SC, Mun HC, Bush M, Chen Y, Nguyen TX, Cao B, Chang DD, Quick M, Conigrave AD, Colecraft HM, McDonald P, Fan QR. Structural mechanism of ligand activation in human calcium-sensing receptor. eLife 2016; 5. [PMID: 27434672 PMCID: PMC4977154 DOI: 10.7554/elife.13662] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 07/18/2016] [Indexed: 12/21/2022] Open
Abstract
Human calcium-sensing receptor (CaSR) is a G-protein-coupled receptor (GPCR) that maintains extracellular Ca2+ homeostasis through the regulation of parathyroid hormone secretion. It functions as a disulfide-tethered homodimer composed of three main domains, the Venus Flytrap module, cysteine-rich domain, and seven-helix transmembrane region. Here, we present the crystal structures of the entire extracellular domain of CaSR in the resting and active conformations. We provide direct evidence that L-amino acids are agonists of the receptor. In the active structure, L-Trp occupies the orthosteric agonist-binding site at the interdomain cleft and is primarily responsible for inducing extracellular domain closure to initiate receptor activation. Our structures reveal multiple binding sites for Ca2+ and PO43- ions. Both ions are crucial for structural integrity of the receptor. While Ca2+ ions stabilize the active state, PO43- ions reinforce the inactive conformation. The activation mechanism of CaSR involves the formation of a novel dimer interface between subunits. DOI:http://dx.doi.org/10.7554/eLife.13662.001 Calcium ions regulate many processes in the human body. The calcium-sensing receptor, called CaSR, is responsible for maintaining a stable level of calcium ions in the blood. This receptor can detect small changes in the concentration of calcium ions, and activates signalling events within the cell to restore the level of calcium ions back to normal. Abnormal activity of this receptor is associated with severe diseases in humans CaSR is found in the surface membrane of cells and belongs to a family of proteins called G-protein coupled receptors. Much of the protein extends out of the cell and interacts with calcium ions, phosphate ions and certain other molecules such as amino acids. However, it was not well understood how these small molecules bind to CaSR and how this activates the receptor. Geng et al. have now used a technique called X-ray crystallography to view the three-dimensional structure of the exterior domain of CaSR in its resting state and active state. These structures revealed that, contrary to expectations, calcium ions are not the main activator of the receptor. Instead, Geng et al. found that CaSR adopts an inactive state in the absence or presence of calcium ions, while the active state only forms when an amino acid is bound. Furthermore investigation showed that calcium ions are needed to stabilise the active form, while phosphate ions keep the inactive form stable. Geng et al. also identified the shape changes that must occur as CaSR transitions from its inactive to its active state. In particular, an amino acid binding to the exterior domain causes it to close like a venus flytrap, which is a crucial step in activating the receptor. Taken together, the findings show that the amino acids and calcium ions act jointly to fully activate CaSR. The next steps are to determine the structure of the entire receptor with and without its small molecule partners and to use these structures to design drugs that can alter CaSR’s activity in order to treat human diseases. DOI:http://dx.doi.org/10.7554/eLife.13662.002
Collapse
Affiliation(s)
- Yong Geng
- Department of Pharmacology, Columbia University, New York, United States.,Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Lidia Mosyak
- Department of Pharmacology, Columbia University, New York, United States
| | - Igor Kurinov
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, United States
| | - Hao Zuo
- Department of Pharmacology, Columbia University, New York, United States
| | - Emmanuel Sturchler
- Department of Molecular Therapeutics, The Scripps Translational Science Institute, Jupiter, United States
| | - Tat Cheung Cheng
- Department of Pharmacology, Columbia University, New York, United States
| | - Prakash Subramanyam
- Department of Physiology and Cellular Biophysics, Columbia University, New York, United States
| | - Alice P Brown
- School of Life and Environmental Sciences, University of Sydney, New South Wales, Australia
| | - Sarah C Brennan
- School of Life and Environmental Sciences, University of Sydney, New South Wales, Australia
| | - Hee-Chang Mun
- School of Life and Environmental Sciences, University of Sydney, New South Wales, Australia
| | - Martin Bush
- Department of Pharmacology, Columbia University, New York, United States
| | - Yan Chen
- Department of Pharmacology, Columbia University, New York, United States
| | - Trang X Nguyen
- Department of Psychiatry, Columbia University, New York, United States
| | - Baohua Cao
- Department of Pharmacology, Columbia University, New York, United States
| | - Donald D Chang
- Department of Physiology and Cellular Biophysics, Columbia University, New York, United States
| | - Matthias Quick
- Department of Psychiatry, Columbia University, New York, United States
| | - Arthur D Conigrave
- School of Life and Environmental Sciences, University of Sydney, New South Wales, Australia
| | - Henry M Colecraft
- Department of Physiology and Cellular Biophysics, Columbia University, New York, United States
| | - Patricia McDonald
- Department of Molecular Therapeutics, The Scripps Translational Science Institute, Jupiter, United States
| | - Qing R Fan
- Department of Pharmacology, Columbia University, New York, United States.,Department of Pathology and Cell Biology, Columbia University, New York, United States
| |
Collapse
|
41
|
Obermannova B, Sumnik Z, Dusatkova P, Cinek O, Grant M, Lebl J, Hendy GN. Novel calcium-sensing receptor cytoplasmic tail deletion mutation causing autosomal dominant hypocalcemia: molecular and clinical study. Eur J Endocrinol 2016; 174:K1-K11. [PMID: 26764418 DOI: 10.1530/eje-15-1216] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 01/12/2016] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Autosomal dominant hypocalcemia (ADH) is a rare disorder caused by activating mutations of the calcium-sensing receptor (CASR). The treatment of ADH patients with 1α-hydroxylated vitamin D derivatives can cause hypercalciuria leading to nephrocalcinosis. DESIGN AND METHODS We studied a girl who presented with hypoparathyroidism and asymptomatic hypocalcemia at age 2.5 years. Mutations of CASR were investigated by DNA sequencing. Functional analyses of mutant and WT CASRs were done in transiently transfected human embryonic kidney (HEK293) cells. RESULTS The proband and her father are heterozygous for an eight-nucleotide deletion c.2703_2710delCCTTGGAG in the CASR encoding the intracellular domain of the protein. Transient expression of CASR constructs in kidney cells in vitro suggested greater cell surface expression of the mutant receptor with a left-shifted extracellular calcium dose-response curve relative to that of the WT receptor consistent with gain of function. Initial treatment of the patient with calcitriol led to increased urinary calcium excretion. Evaluation for mosaicism in the paternal grandparents of the proband was negative. CONCLUSIONS We describe a novel naturally occurring deletion mutation within the CASR that apparently arose de novo in the father of the ADH proband. Functional analysis suggests that the cytoplasmic tail of the CASR contains determinants that regulate the attenuation of signal transduction. Early molecular analysis of the CASR gene in patients with isolated idiopathic hypoparathyroidism is recommended because of its relevance to clinical outcome and treatment choice. In ADH patients, calcium supplementation and low-dose cholecalciferol avoids hypocalcemic symptoms without compromising renal function.
Collapse
Affiliation(s)
- Barbora Obermannova
- Department of PediatricsSecond Faculty of Medicine, Charles University in Prague, University Hospital Motol V Uvalu 84, CZ-150 06 Prague, Czech RepublicLady Davis Institute for Medical ResearchSMBD-Jewish General Hospital, McGill University, Montréal, Québec, Canada H3T 1E2Experimental Therapeutics and MetabolismRoom No. EM1.3226 RI-McGill University Health Centre Glen Site, 1001 Décarie Boulevard, Montréal, Québec, Canada H4A 3J1Departments of MedicinePhysiology, and Human Genetics, McGill University, Montréal, Québec, Canada H4A 3J1
| | - Zdenek Sumnik
- Department of PediatricsSecond Faculty of Medicine, Charles University in Prague, University Hospital Motol V Uvalu 84, CZ-150 06 Prague, Czech RepublicLady Davis Institute for Medical ResearchSMBD-Jewish General Hospital, McGill University, Montréal, Québec, Canada H3T 1E2Experimental Therapeutics and MetabolismRoom No. EM1.3226 RI-McGill University Health Centre Glen Site, 1001 Décarie Boulevard, Montréal, Québec, Canada H4A 3J1Departments of MedicinePhysiology, and Human Genetics, McGill University, Montréal, Québec, Canada H4A 3J1
| | - Petra Dusatkova
- Department of PediatricsSecond Faculty of Medicine, Charles University in Prague, University Hospital Motol V Uvalu 84, CZ-150 06 Prague, Czech RepublicLady Davis Institute for Medical ResearchSMBD-Jewish General Hospital, McGill University, Montréal, Québec, Canada H3T 1E2Experimental Therapeutics and MetabolismRoom No. EM1.3226 RI-McGill University Health Centre Glen Site, 1001 Décarie Boulevard, Montréal, Québec, Canada H4A 3J1Departments of MedicinePhysiology, and Human Genetics, McGill University, Montréal, Québec, Canada H4A 3J1
| | - Ondrej Cinek
- Department of PediatricsSecond Faculty of Medicine, Charles University in Prague, University Hospital Motol V Uvalu 84, CZ-150 06 Prague, Czech RepublicLady Davis Institute for Medical ResearchSMBD-Jewish General Hospital, McGill University, Montréal, Québec, Canada H3T 1E2Experimental Therapeutics and MetabolismRoom No. EM1.3226 RI-McGill University Health Centre Glen Site, 1001 Décarie Boulevard, Montréal, Québec, Canada H4A 3J1Departments of MedicinePhysiology, and Human Genetics, McGill University, Montréal, Québec, Canada H4A 3J1
| | - Michael Grant
- Department of PediatricsSecond Faculty of Medicine, Charles University in Prague, University Hospital Motol V Uvalu 84, CZ-150 06 Prague, Czech RepublicLady Davis Institute for Medical ResearchSMBD-Jewish General Hospital, McGill University, Montréal, Québec, Canada H3T 1E2Experimental Therapeutics and MetabolismRoom No. EM1.3226 RI-McGill University Health Centre Glen Site, 1001 Décarie Boulevard, Montréal, Québec, Canada H4A 3J1Departments of MedicinePhysiology, and Human Genetics, McGill University, Montréal, Québec, Canada H4A 3J1
| | - Jan Lebl
- Department of PediatricsSecond Faculty of Medicine, Charles University in Prague, University Hospital Motol V Uvalu 84, CZ-150 06 Prague, Czech RepublicLady Davis Institute for Medical ResearchSMBD-Jewish General Hospital, McGill University, Montréal, Québec, Canada H3T 1E2Experimental Therapeutics and MetabolismRoom No. EM1.3226 RI-McGill University Health Centre Glen Site, 1001 Décarie Boulevard, Montréal, Québec, Canada H4A 3J1Departments of MedicinePhysiology, and Human Genetics, McGill University, Montréal, Québec, Canada H4A 3J1
| | - Geoffrey N Hendy
- Department of PediatricsSecond Faculty of Medicine, Charles University in Prague, University Hospital Motol V Uvalu 84, CZ-150 06 Prague, Czech RepublicLady Davis Institute for Medical ResearchSMBD-Jewish General Hospital, McGill University, Montréal, Québec, Canada H3T 1E2Experimental Therapeutics and MetabolismRoom No. EM1.3226 RI-McGill University Health Centre Glen Site, 1001 Décarie Boulevard, Montréal, Québec, Canada H4A 3J1Departments of MedicinePhysiology, and Human Genetics, McGill University, Montréal, Québec, Canada H4A 3J1 Department of PediatricsSecond Faculty of Medicine, Charles University in Prague, University Hospital Motol V Uvalu 84, CZ-150 06 Prague, Czech RepublicLady Davis Institute for Medical ResearchSMBD-Jewish General Hospital, McGill University, Montréal, Québec, Canada H3T 1E2Experimental Therapeutics and MetabolismRoom No. EM1.3226 RI-McGill University Health Centre Glen Site, 1001 Décarie Boulevard, Montréal, Québec, Canada H4A 3J1Departments of MedicinePhysiology, and Human Genetics, McGill University, Montréal, Québec, Canada H4A 3J1
| |
Collapse
|
42
|
Babinsky VN, Hannan FM, Gorvin CM, Howles SA, Nesbit MA, Rust N, Hanyaloglu AC, Hu J, Spiegel AM, Thakker RV. Allosteric Modulation of the Calcium-sensing Receptor Rectifies Signaling Abnormalities Associated with G-protein α-11 Mutations Causing Hypercalcemic and Hypocalcemic Disorders. J Biol Chem 2016; 291:10876-85. [PMID: 26994139 PMCID: PMC4865932 DOI: 10.1074/jbc.m115.696401] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Indexed: 11/06/2022] Open
Abstract
Germline loss- and gain-of-function mutations of G-protein α-11 (Gα11), which couples the calcium-sensing receptor (CaSR) to intracellular calcium (Ca2+i) signaling, lead to familial hypocalciuric hypercalcemia type 2 (FHH2) and autosomal dominant hypocalcemia type 2 (ADH2), respectively, whereas somatic Gα11 mutations mediate uveal melanoma development by constitutively up-regulating MAPK signaling. Cinacalcet and NPS-2143 are allosteric CaSR activators and inactivators, respectively, that ameliorate signaling disturbances associated with CaSR mutations, but their potential to modulate abnormalities of the downstream Gα11 protein is unknown. This study investigated whether cinacalcet and NPS-2143 may rectify Ca2+i alterations associated with FHH2- and ADH2-causing Gα11 mutations, and evaluated the influence of germline gain-of-function Gα11 mutations on MAPK signaling by measuring ERK phosphorylation, and assessed the effect of NPS-2143 on a uveal melanoma Gα11 mutant. WT and mutant Gα11 proteins causing FHH2, ADH2 or uveal melanoma were transfected in CaSR-expressing HEK293 cells, and Ca2+i and ERK phosphorylation responses measured by flow-cytometry and Alphascreen immunoassay following exposure to extracellular Ca2+ (Ca2+o) and allosteric modulators. Cinacalcet and NPS-2143 rectified the Ca2+i responses of FHH2- and ADH2-associated Gα11 loss- and gain-of-function mutations, respectively. ADH2-causing Gα11 mutations were demonstrated not to be constitutively activating and induced ERK phosphorylation following Ca2+o stimulation only. The increased ERK phosphorylation associated with ADH2 and uveal melanoma mutants was rectified by NPS-2143. These findings demonstrate that CaSR-targeted compounds can rectify signaling disturbances caused by germline and somatic Gα11 mutations, which respectively lead to calcium disorders and tumorigenesis; and that ADH2-causing Gα11 mutations induce non-constitutive alterations in MAPK signaling.
Collapse
Affiliation(s)
- Valerie N Babinsky
- From the Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LJ, United Kingdom
| | - Fadil M Hannan
- From the Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LJ, United Kingdom, Department of Musculoskeletal Biology, University of Liverpool, Liverpool L69 3GA, United Kingdom
| | - Caroline M Gorvin
- From the Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LJ, United Kingdom
| | - Sarah A Howles
- From the Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LJ, United Kingdom
| | - M Andrew Nesbit
- From the Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LJ, United Kingdom, Biomedical Sciences Research Institute, Ulster University, Coleraine BT52 1SA, United Kingdom
| | - Nigel Rust
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Aylin C Hanyaloglu
- Department of Surgery and Cancer, Institute of Reproductive Biology and Development, Imperial College London, London W12 0NN, United Kingdom
| | - Jianxin Hu
- Laboratory of Bioorganic Chemistry, NIDDK, National Institutes of Health, Bethesda, Maryland 20892, and
| | | | - Rajesh V Thakker
- From the Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LJ, United Kingdom,
| |
Collapse
|
43
|
Changcharoen B, Motosue M, Wong L, Arakaki R. A Woman and Her Father with Calcium-Sensing Receptor Mutation and Autosomal Dominant Hypocalcemia. AACE Clin Case Rep 2016. [DOI: 10.4158/ep15716.cr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
44
|
Aggarwal A, Höbaus J, Tennakoon S, Prinz-Wohlgenannt M, Graça J, Price SA, Heffeter P, Berger W, Baumgartner-Parzer S, Kállay E. Active vitamin D potentiates the anti-neoplastic effects of calcium in the colon: A cross talk through the calcium-sensing receptor. J Steroid Biochem Mol Biol 2016; 155:231-8. [PMID: 25758239 DOI: 10.1016/j.jsbmb.2015.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 02/06/2023]
Abstract
Epidemiological studies suggest an inverse correlation between dietary calcium (Ca(2+)) and vitamin D intake and the risk of colorectal cancer (CRC). It has been shown in vitro that the active vitamin D metabolite, 1,25-dihydroxyvitamin D3 (1,25-D3) can upregulate expression of the calcium-sensing receptor (CaSR). In the colon, CaSR has been suggested to regulate proliferation of colonocytes. However, during tumorigenesis colonic CaSR expression is downregulated and we hypothesized that the loss of CaSR could influence the anti-tumorigenic effects of Ca(2+) and vitamin D. Our aim was to assess the impact of CaSR expression and function on the anti-neoplastic effects of 1,25-D3 in colon cancer cell lines. We demonstrated that in the healthy colon of mice, high vitamin D diet (2500 IU/kg diet) increased expression of differentiation and apoptosis markers, decreased expression of proliferation markers and significantly upregulated CaSR mRNA expression, compared with low vitamin D diet (100 IU/kg diet). To determine the role of CaSR in this process, we transfected Caco2-15 and HT29 CRC cells with wild type CaSR (CaSR-WT) or a dominant negative CaSR mutant (CaSR-DN) and treated them with 1,25-D3 alone, or in combination with CaSR activators (Ca(2+) and NPS R-568). 1,25-D3 enhanced the anti-proliferative effects of Ca(2+) and induced differentiation and apoptosis only in cells with a functional CaSR, which were further enhanced in the presence of NPS R-568, a positive allosteric modulator of CaSR. The mutant CaSR inhibited the anti-tumorigenic effects of 1,25-D3 suggesting that the anti-neoplastic effects of 1,25-D3 are, at least in part, mediated by the CaSR. Taken together, our data provides molecular evidence to support the epidemiological observation that both, vitamin D and calcium are needed for protection against malignant transformation of the colon and that their effect is modulated by the presence of a functional CaSR. This article is part of a Special Issue entitled '17th Vitamin D Workshop'.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Adenocarcinoma/prevention & control
- Aniline Compounds/pharmacology
- Animals
- Caco-2 Cells
- Calcium/metabolism
- Calcium/pharmacology
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Colon/drug effects
- Colon/metabolism
- Colon/pathology
- Colonic Neoplasms/genetics
- Colonic Neoplasms/metabolism
- Colonic Neoplasms/pathology
- Colonic Neoplasms/prevention & control
- Dietary Supplements
- Gene Expression Regulation, Neoplastic
- HT29 Cells
- Humans
- Male
- Mice
- Mice, Transgenic
- Mutation
- Phenethylamines
- Propylamines
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Calcium-Sensing
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction
- Transfection
- Vitamin D/analogs & derivatives
- Vitamin D/pharmacology
Collapse
Affiliation(s)
- Abhishek Aggarwal
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Julia Höbaus
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Samawansha Tennakoon
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | | | - João Graça
- Safety Assessment, AstraZeneca, Macclesfield, UK
| | | | - Petra Heffeter
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Research Platform 'Translational Cancer Therapy Research', Vienna, Austria
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Research Platform 'Translational Cancer Therapy Research', Vienna, Austria
| | | | - Enikö Kállay
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
45
|
Graca JAZ, Schepelmann M, Brennan SC, Reens J, Chang W, Yan P, Toka H, Riccardi D, Price SA. Comparative expression of the extracellular calcium-sensing receptor in the mouse, rat, and human kidney. Am J Physiol Renal Physiol 2015; 310:F518-33. [PMID: 26661650 DOI: 10.1152/ajprenal.00208.2015] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 11/30/2015] [Indexed: 11/22/2022] Open
Abstract
The calcium-sensing receptor (CaSR) was cloned over 20 years ago and functionally demonstrated to regulate circulating levels of parathyroid hormone by maintaining physiological serum ionized calcium concentration ([Ca(2+)]). The receptor is highly expressed in the kidney; however, intrarenal and intraspecies distribution remains controversial. Recently, additional functions of the CaSR receptor in the kidney have emerged, including parathyroid hormone-independent effects. It is therefore critical to establish unequivocally the localization of the CaSR in the kidney to relate this to its proposed physiological roles. In this study, we determined CaSR expression in mouse, rat, and human kidneys using in situ hybridization, immunohistochemistry (using 8 different commercially available and custom-made antibodies), and proximity ligation assays. Negative results in mice with kidney-specific CaSR ablation confirmed the specificity of the immunohistochemistry signal. Both in situ hybridization and immunohistochemistry showed CaSR expression in the thick ascending limb, distal tubule, and collecting duct of all species, with the thick ascending limb showing the highest levels. Within the collecting ducts, there was significant heterogeneity of expression between cell types. In the proximal tubule, lower levels of immunoreactivity were detected by immunohistochemistry and proximity ligation assays. Proximity ligation assays were the only technique to demonstrate expression within glomeruli. This study demonstrated CaSR expression throughout the kidney with minimal discrepancy between species but with significant variation in the levels of expression between cell and tubule types. These findings clarify the intrarenal distribution of the CaSR and enable elucidation of the full physiological roles of the receptor within this organ.
Collapse
Affiliation(s)
- J A Z Graca
- Pathology Sciences, AstraZeneca R&D, Macclesfield, United Kingdom; School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - M Schepelmann
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - S C Brennan
- School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - J Reens
- Pathology Sciences, AstraZeneca R&D, Macclesfield, United Kingdom
| | - W Chang
- Department of Medicine, UCSF School of Medicine, San Francisco, California
| | - P Yan
- Division of Nephrology, Beth Israel Deaconess Medical Center, Boston, Massachusetts; and
| | - H Toka
- Division of Nephrology and Hyperension, Eastern Virginia Medical School, Norfolk, Virginia
| | - D Riccardi
- School of Biosciences, Cardiff University, Cardiff, United Kingdom;
| | - S A Price
- Pathology Sciences, AstraZeneca R&D, Macclesfield, United Kingdom
| |
Collapse
|
46
|
Liu C, Wu GQ, Fu XW, Mo XH, Zhao LH, Hu HM, Zhu SE, Hou YP. The Extracellular Calcium-Sensing Receptor (CASR) Regulates Gonadotropins-Induced Meiotic Maturation of Porcine Oocytes. Biol Reprod 2015; 93:131. [PMID: 26490840 DOI: 10.1095/biolreprod.115.128579] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 10/14/2015] [Indexed: 12/16/2022] Open
Abstract
Gonadotropins and epidermal growth factor (EGF) play crucial roles in promoting oocyte maturation. The regulatory network downstream of these key factors is not well understood. The present study was designed to investigate the role of the calcium-sensing receptor (CASR) in porcine oocyte in vitro maturation. CASR expression was up-regulated in oocytes matured in gonadotropin-containing medium. Cortical distribution of CASR was enhanced with gonadotropins but not EGF. Supplementation of a CASR agonist (NPS R-568) in the gonadotropin (FSH and/or LH)-containing maturation medium significantly enhanced oocyte nuclear maturation. Addition of NPS2390, a CASR antagonist, compromised oocyte nuclear maturation. Furthermore, increased cortical distribution and decreased expression of CASR was observed after the NPS R-568 treatment. Oocytes treated with NPS R-568 had higher concentration of CYCLIN B1, decreased reactive oxygen species, and increased glutathione levels, indicative of advanced cytoplasmic maturation. In contrast, NPS2390 treatment compromised oocyte cytoplasmic maturation. A higher blastocyst formation rate after parthenogenetic activation was observed when oocytes were matured in the presence of the CASR agonist, NPS R-568. MAPK3/1 phosphorylation was increased during in vitro maturation and after NPS R-568 treatment, and decreased following CASR antagonist supplementation. Taken together, our data showed that the CASR is a gonadotropin-regulated factor that promotes porcine oocyte maturation in a MAPK-dependent manner.
Collapse
Affiliation(s)
- Cong Liu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Guo-Quan Wu
- Yunnan Animal Science and Veterinary Institute, Kunming, Yunnan, China
| | - Xiang-Wei Fu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Xian-Hong Mo
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Li-Hong Zhao
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Hong-Mei Hu
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| | - Shi-En Zhu
- Key Laboratory of Animal Genetics, Breeding and Reproduction, Ministry of Agriculture and National Engineering Laboratory for Animal Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, China
| | - Yun-Peng Hou
- State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, China
| |
Collapse
|
47
|
Aggarwal A, Prinz-Wohlgenannt M, Tennakoon S, Höbaus J, Boudot C, Mentaverri R, Brown EM, Baumgartner-Parzer S, Kállay E. The calcium-sensing receptor: A promising target for prevention of colorectal cancer. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1853:2158-67. [PMID: 25701758 PMCID: PMC4549785 DOI: 10.1016/j.bbamcr.2015.02.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 02/07/2015] [Accepted: 02/09/2015] [Indexed: 02/07/2023]
Abstract
The inverse correlation between dietary calcium intake and the risk of colorectal cancer (CRC) is well known, but poorly understood. Expression of the calcium-sensing receptor (CaSR), a calcium-binding G protein-coupled receptor is downregulated in CRC leading us to hypothesize that the CaSR has tumor suppressive roles in the colon. The aim of this study was to understand whether restoration of CaSR expression could reduce the malignant phenotype in CRC. In human colorectal tumors, expression of the CaSR negatively correlated with proliferation markers whereas loss of CaSR correlated with poor tumor differentiation and reduced apoptotic potential. In vivo, dearth of CaSR significantly increased expression of proliferation markers and decreased levels of differentiation and apoptotic markers in the colons of CaSR/PTH double knock-out mice confirming the tumor suppressive functions of CaSR. In vitro CRC cells stably overexpressing wild-type CaSR showed significant reduction in proliferation, as well as increased differentiation and apoptotic potential. The positive allosteric modulator of CaSR, NPS R-568 further enhanced these effects, whereas treatment with the negative allosteric modulator, NPS 2143 inhibited these functions. Interestingly, the dominant-negative mutant (R185Q) was able to abrogate these effects. Our results demonstrate a critical tumor suppressive role of CaSR in the colon. Restoration of CaSR expression and function is linked to regulation of the balance between proliferation, differentiation, and apoptosis and provides a rationale for novel strategies in CRC therapy.
Collapse
MESH Headings
- Amino Acid Substitution
- Aniline Compounds/pharmacology
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Caco-2 Cells
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/pathology
- Colorectal Neoplasms/prevention & control
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Humans
- Male
- Mice
- Mice, Knockout
- Mutation, Missense
- Naphthalenes/pharmacology
- Phenethylamines
- Propylamines
- Receptors, Calcium-Sensing/antagonists & inhibitors
- Receptors, Calcium-Sensing/genetics
- Receptors, Calcium-Sensing/metabolism
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Tumor Suppressor Proteins/antagonists & inhibitors
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
Collapse
Affiliation(s)
- Abhishek Aggarwal
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | | | - Samawansha Tennakoon
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Julia Höbaus
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Cedric Boudot
- INSERM U1088, University of Picardie Jules Verne, Amiens, France
| | | | - Edward M Brown
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Boston, USA
| | | | - Enikö Kállay
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
48
|
Abstract
We have investigated how CaSR activation enhances sympathetic axon growth. CaSR activation promotes phosphorylation of ERK1 and ERK2. Inhibition of ERK1/ERK2 phosphorylation blocks CaSR-promoted axon growth. CaSR-promoted axon growth requires a discrete region of the cytoplasmic domain.
The extracellular calcium-sensing receptor (CaSR) is a G-protein coupled receptor that monitors the systemic extracellular free ionized calcium level ([Ca2+]o) in organs involved in systemic [Ca2+]o homeostasis. CaSR is widely expressed in the nervous system and its activation promotes axon and dendrite growth during development, but the mechanism by which it does this is not known. Here we show that enhanced axon growth and branching from cultured embryonic sympathetic neurons by activation of the endogenous CaSR depends on the presence of nerve growth factor (NGF). Our observation that activation of overexpressed CaSR promotes axon growth in NGF-free medium has enabled us to investigate CaSR downstream signaling contributing to axon growth in the absence of NGF signaling. We show that activation of overexpressed CaSR leads to activation of ERK1 and ERK2, and pharmacological inhibition of CaSR-dependent ERK1/ERK2 activation prevents CaSR-dependent axon growth. Analysis of axon growth from cultured neurons expressing deletion mutants of the CaSR cytoplasmic tail revealed that the region between alanine 877 and glycine 907 is required for promoting axon growth that is distinct from the high-affinity filamin-A binding site that has previously been implicated in ERK1/ERK2 activation.
Collapse
Affiliation(s)
- Thomas N Vizard
- School of Biosciences, Biomedical Building, Museum Avenue, Cardiff CF10 3US, UK.
| | - Michael Newton
- School of Biosciences, Biomedical Building, Museum Avenue, Cardiff CF10 3US, UK
| | - Laura Howard
- School of Biosciences, Biomedical Building, Museum Avenue, Cardiff CF10 3US, UK
| | - Sean Wyatt
- School of Biosciences, Biomedical Building, Museum Avenue, Cardiff CF10 3US, UK
| | - Alun M Davies
- School of Biosciences, Biomedical Building, Museum Avenue, Cardiff CF10 3US, UK
| |
Collapse
|
49
|
Mary A, Hénaut L, Boudot C, Six I, Brazier M, Massy ZA, Drüeke TB, Kamel S, Mentaverri R. Calcitriol prevents in vitro vascular smooth muscle cell mineralization by regulating calcium-sensing receptor expression. Endocrinology 2015; 156:1965-74. [PMID: 25763635 DOI: 10.1210/en.2014-1744] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Vascular calcification (VC) is a degenerative disease that contributes to cardiovascular morbidity and mortality. A negative relationship has been demonstrated between VC and calcium sensing receptor (CaSR) expression in the vasculature. Of interest, vitamin D response elements, which allow responsiveness to 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], are present in the promoters of the CaSR gene. We hypothesized that 1,25(OH)2D3, by modulating CaSR expression in vascular smooth muscle cells (VSMCs), might protect against VC. Human VSMCs were exposed to increasing concentrations of 1,25(OH)2D3 (0.01-10 nmol/L) in noncalcifying (1.8 mmol/L) or procalcifying Ca(2+)0 condition (5.0 mmol/L). Using quantitative RT-PCR and Western blotting we observed a significant increase in both CaSR mRNA and protein levels after exposure to 1.0 nmol/L 1,25(OH)2D3. This effect was associated with a maximal increase in CaSR expression at the cell surface after 48 hours of 1,25(OH)2D3 treatment, as assessed by flow cytometry. Down-regulation of the vitamin D receptor by small interfering RNA abolished these effects. In the procalcifying condition, 1.0 nmol/L 1,25(OH)2D3 blocked the Ca(2+)0-induced decrease in total and surface CaSR expression and protected against mineralization. Down-regulation of CaSR expression by CaSR small interfering RNA abolished this protective effect. 1,25(OH)2D3 concentrations of 0.5 and 5.0 nmol/L were also effective, but other (0.01, 0.1, and 10 nmol/L) concentrations did not modify CaSR expression and human VSMC mineralization. In conclusion, these findings suggest that nanomolar concentrations of 1,25(OH)2D3 induce a CaSR-dependent protection against VC. Both lower and higher concentrations are either ineffective or may even promote VC. Whether this also holds true in the clinical setting requires further study.
Collapse
Affiliation(s)
- Aurélien Mary
- INSERM Unit 1088 (A.M., L.H., C.B., I.B., M.B., Z.A.M., T.B.D., S.K., R.M.), University of Picardie Jules Vernes, 80000 Amiens, France; Department of Pharmacy (A.M.) and Department of Biochemistry (M.B., S.K., R.M.), Amiens University Medical Center, 80054 Amiens, France; Division of Nephrology (Z.A.M.), Ambroise Paré University Hospital, Assistance Publique-Hôpitaux de Paris, University Versailles Saint-Quentin-en-Yvelines, 92100 Boulogne Billancourt/Paris, France; and Multifaceted CaSR Initial Training Network (M.B., Z.A.M., S.K., R.M.)
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Baran N, ter Braak M, Saffrich R, Woelfle J, Schmitz U. Novel activating mutation of human calcium-sensing receptor in a family with autosomal dominant hypocalcaemia. Mol Cell Endocrinol 2015; 407:18-25. [PMID: 25766501 DOI: 10.1016/j.mce.2015.02.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 01/13/2015] [Accepted: 02/19/2015] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Autosomal dominant hypocalcaemia (ADH) is caused by activating mutations in the calcium sensing receptor gene (CaR) and characterised by mostly asymptomatic mild to moderate hypocalcaemia with low, inappropriately serum concentration of PTH. OBJECTIVE The purpose of the present study was to biochemically and functionally characterise a novel mutation of CaR. PATIENTS A female proband presenting with hypocalcaemia was diagnosed to have "idiopathic hypoparathyroidism" at the age of 10 with a history of muscle pain and cramps. Further examinations demonstrated hypocalcaemia in nine additional family members, affecting three generations. MAIN OUTCOME MEASURE P136L CaR mutation was predicted to cause gain of function of CaR. RESULTS Affected family members showed relevant hypocalcaemia (mean ± SD; 1.9 ± 0.1 mmol/l). Patient history included mild seizures and recurrent nephrolithiasis. Genetic analysis confirmed that hypocalcaemia cosegregated with a heterozygous mutation at codon 136 (CCC → CTC/Pro → Leu) in exon 3 of CaR confirming the diagnosis of ADH. For in vitro studies P136L mutant CaR was generated by site-directed mutagenesis and examined in transiently transfected HEK293 cells. Extracellular calcium stimulation of transiently transfected HEK293 cells showed significantly increased intracellular Ca(2+) mobilisation and MAPK activity for mutant P136L CaR compared to wild type CaR. CONCLUSIONS The present study gives insight about a novel activating mutation of CaR and confirms that the novel P136L-CaR mutation is responsible for ADH in this family.
Collapse
Affiliation(s)
- Natalia Baran
- Department of Endocrinology and Diabetology, University of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany; Department of Medicine V, University of Heidelberg, INF 410, 69120 Heidelberg, Germany.
| | - Michael ter Braak
- Institut of Pharmacology, University of Essen, Hufelandstr. 55, 45122 Essen, Germany
| | - Rainer Saffrich
- Department of Medicine V, University of Heidelberg, INF 410, 69120 Heidelberg, Germany
| | - Joachim Woelfle
- Pediatric Endocrinology Division, University of Bonn, Adenauerallee 119, 53113 Bonn, Germany
| | - Udo Schmitz
- Department of Endocrinology and Diabetology, University of Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| |
Collapse
|