1
|
Alhassen S, Hogenkamp D, Nguyen HA, Al Masri S, Abbott GW, Civelli O, Alachkar A. Ophthalmate is a new regulator of motor functions via CaSR: implications for movement disorders. Brain 2024; 147:3379-3394. [PMID: 38537648 PMCID: PMC11449132 DOI: 10.1093/brain/awae097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 02/17/2024] [Accepted: 03/12/2024] [Indexed: 10/05/2024] Open
Abstract
Dopamine's role as the principal neurotransmitter in motor functions has long been accepted. We broaden this conventional perspective by demonstrating the involvement of non-dopaminergic mechanisms. In mouse models of Parkinson's disease, we observed that L-DOPA elicited a substantial motor response even when its conversion to dopamine was blocked by inhibiting the enzyme aromatic amino acid decarboxylase (AADC). Remarkably, the motor activity response to L-DOPA in the presence of an AADC inhibitor (NSD1015) showed a delayed onset, yet greater intensity and longer duration, peaking at 7 h, compared to when L-DOPA was administered alone. This suggests an alternative pathway or mechanism, independent of dopamine signalling, mediating the motor functions. We sought to determine the metabolites associated with the pronounced hyperactivity observed, using comprehensive metabolomics analysis. Our results revealed that the peak in motor activity induced by NSD1015/L-DOPA in Parkinson's disease mice is associated with a surge (20-fold) in brain levels of the tripeptide ophthalmic acid (also known as ophthalmate in its anionic form). Interestingly, we found that administering ophthalmate directly to the brain rescued motor deficits in Parkinson's disease mice in a dose-dependent manner. We investigated the molecular mechanisms underlying ophthalmate's action and discovered, through radioligand binding and cAMP-luminescence assays, that ophthalmate binds to and activates the calcium-sensing receptor (CaSR). Additionally, our findings demonstrated that a CaSR antagonist inhibits the motor-enhancing effects of ophthalmate, further solidifying the evidence that ophthalmate modulates motor functions through the activation of the CaSR. The discovery of ophthalmate as a novel regulator of motor function presents significant potential to transform our understanding of brain mechanisms of movement control and the therapeutic management of related disorders.
Collapse
Affiliation(s)
- Sammy Alhassen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Derk Hogenkamp
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| | - Hung Anh Nguyen
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Saeed Al Masri
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Geoffrey W Abbott
- Bioelectricity Laboratory, Department of Physiology and Biophysics, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| | - Olivier Civelli
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA 92697, USA
| | - Amal Alachkar
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
- Institute for Genomics and Bioinformatics, School of Information and Computer Sciences, University of California Irvine, Irvine, CA 92697, USA
- UC Irvine Center for the Neurobiology of Learning and Memory, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
2
|
Matarage Don NJ, Padmavathi R, Khasro TD, Zaman MRU, Ji HF, Ram JL, Ahn YH. Glutathione-Based Photoaffinity Probe Identifies Caffeine as a Positive Allosteric Modulator of the Calcium-Sensing Receptor. ACS Chem Biol 2024; 19:1661-1670. [PMID: 38975966 PMCID: PMC11267565 DOI: 10.1021/acschembio.4c00335] [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: 05/11/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/09/2024]
Abstract
The calcium-sensing receptor (CaSR), abundantly expressed in the parathyroid gland and kidney, plays a central role in calcium homeostasis. In addition, CaSR exerts multimodal roles, including inflammation, muscle contraction, and bone remodeling, in other organs and tissues. The diverse functions of CaSR are mediated by many endogenous and exogenous ligands, including calcium, amino acids, glutathione, cinacalcet, and etelcalcetide, that have distinct binding sites in CaSR. However, strategies to evaluate ligand interactions with CaSR remain limited. Here, we developed a glutathione-based photoaffinity probe, DAZ-G, that analyzes ligand binding to CaSR. We showed that DAZ-G binds to the amino acid binding site in CaSR and acts as a positive allosteric modulator of CaSR. Oxidized and reduced glutathione and phenylalanine effectively compete with DAZ-G conjugation to CaSR, while calcium, cinacalcet, and etelcalcetide have cooperative effects. An unexpected finding was that caffeine effectively competes with DAZ-G's conjugation to CaSR and acts as a positive allosteric modulator of CaSR. The effective concentration of caffeine for CaSR activation (<10 μM) is easily attainable in plasma by ordinary caffeine consumption. Our report demonstrates the utility of a new chemical probe for CaSR and discovers a new protein target of caffeine, suggesting that caffeine consumption can modulate the diverse functions of CaSR.
Collapse
Affiliation(s)
| | - Rayavarapu Padmavathi
- Department
of Chemistry, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Talan D. Khasro
- Department
of Chemistry, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Md. Rumman U. Zaman
- Department
of Chemistry, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Hai-Feng Ji
- Department
of Chemistry, Drexel University, Philadelphia, Pennsylvania 19104, United States
| | - Jeffrey L. Ram
- Department
of Physiology, Wayne State University, Detroit, Michigan 48201, United States
| | - Young-Hoon Ahn
- Department
of Chemistry, Drexel University, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
3
|
Zhang J, Li Q, Liao P, Xiao R, Zhu L, Hu Q. Calcium sensing receptor: A promising therapeutic target in pulmonary hypertension. Life Sci 2024; 340:122472. [PMID: 38290572 DOI: 10.1016/j.lfs.2024.122472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/01/2024]
Abstract
Pulmonary hypertension (PH) is characterized by elevation of pulmonary arterial pressure and pulmonary vascular resistance. The increased pulmonary arterial pressure and pulmonary vascular resistance due to sustained pulmonary vasoconstriction and pulmonary vascular remodeling can lead to right heart failure and eventual death. A rise in intracellular Ca2+ concentration ([Ca2+]i) and enhanced pulmonary arterial smooth muscle cells (PASMCs) proliferation contribute to pulmonary vasoconstriction and pulmonary vascular remodeling. Recent studies demonstrated that extracellular calcium sensing receptor (CaSR) as a G-protein coupled receptor participates in [Ca2+]i increase induced by hypoxia in the experimental animals of PH and in PH patients. Pharmacological blockade or gene knockout of CaSR significantly attenuates the development of PH. This review will aim to discuss and update the pathogenicity of CaSR attributed to onset and progression in PH.
Collapse
Affiliation(s)
- Jiwei Zhang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinli Li
- Department of Clinical Laboratory Medicine, People's Hospital of Dongxihu District Wuhan City and Union Dongxihu Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Pu Liao
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Xiao
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liping Zhu
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qinghua Hu
- Key Laboratory of Pulmonary Diseases of Ministry of Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Jang Y, Lee H, Cho Y, Choi E, Jo S, Sohn HM, Kim BC, Ko YJ, Lim W. An LGR4 agonist activates the GSK‑3β pathway to inhibit RANK‑RANKL signaling during osteoclastogenesis in bone marrow‑derived macrophages. Int J Mol Med 2024; 53:10. [PMID: 38063193 PMCID: PMC10712694 DOI: 10.3892/ijmm.2023.5334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
The binding between receptor‑activated nuclear factor‑κB (RANK) and the RANK ligand (RANKL) during osteoclast development is an important target for drugs that treat osteoporosis. The leucine‑rich repeat‑containing G‑protein‑coupled receptor 4 (LGR4) acts as a negative regulator of RANK‑RANKL that suppresses canonical RANK signaling during osteoclast differentiation. Therefore, LGR4 agonists may be useful in inhibiting osteoclastogenesis and effectively treating osteoporosis. In the present study, bone marrow‑derived macrophages and a mouse model of RANKL‑induced bone loss were used to investigate the effect of mutant RANKL (MT RANKL), which was previously developed based on the crystal structure of the RANKL complex. In the present study, the binding affinity of wild‑type (WT) RANKL and MT RANKL to RANK and LGR4 was determined using microscale thermophoresis analysis, and the effect of the ligands on the AKT‑glycogen synthase kinase‑3β (GSK‑3β)‑nuclear factor of activated T cells, cytoplasmic, calcineurin‑dependent 1 (NFATc1) signaling cascade was investigated using western blotting and confocal microscopy. In addition, the expression of LGR4 and the colocalization of LGR4 with MT RANKL were analyzed in a mouse model of RANKL‑induced bone loss. The results showed that in osteoclast precursor cells, MT RANKL bound with high affinity to LGR4 and increased GSK‑3β phosphorylation independently of AKT, resulting in the inhibition of NFATc1 nuclear translocation. In the mouse model, MT RANKL colocalized with LGR4 and inhibited bone resorption. These results indicated that MT RANKL may inhibit RANKL‑induced osteoclastogenesis through an LGR4‑dependent pathway and this could be exploited to develop new therapies for osteoporosis.
Collapse
Affiliation(s)
- Yuria Jang
- Laboratory of Orthopedic Research, Chosun University Hospital, Gwangju 61452, Republic of Korea
- Department of Premedical Science, Gwangju 61452, Republic of Korea
| | - Hyeonjoon Lee
- Laboratory of Orthopedic Research, Chosun University Hospital, Gwangju 61452, Republic of Korea
- Department of Orthopaedic Surgery, College of Medicine, Gwangju 61452, Republic of Korea
| | - Yongjin Cho
- Laboratory of Orthopedic Research, Chosun University Hospital, Gwangju 61452, Republic of Korea
- Department of Orthopaedic Surgery, College of Medicine, Gwangju 61452, Republic of Korea
| | - Eunseo Choi
- Department of Physics, Chosun University, Gwangju 61452, Republic of Korea
| | - Suenghwan Jo
- Laboratory of Orthopedic Research, Chosun University Hospital, Gwangju 61452, Republic of Korea
- Department of Orthopaedic Surgery, College of Medicine, Gwangju 61452, Republic of Korea
| | - Hong Moon Sohn
- Laboratory of Orthopedic Research, Chosun University Hospital, Gwangju 61452, Republic of Korea
- Department of Orthopaedic Surgery, College of Medicine, Gwangju 61452, Republic of Korea
| | - Beom Chang Kim
- Laboratory of Orthopedic Research, Chosun University Hospital, Gwangju 61452, Republic of Korea
- Department of Premedical Science, Gwangju 61452, Republic of Korea
| | - Young Jong Ko
- Laboratory of Orthopedic Research, Chosun University Hospital, Gwangju 61452, Republic of Korea
- Department of Premedical Science, Gwangju 61452, Republic of Korea
| | - Wonbong Lim
- Laboratory of Orthopedic Research, Chosun University Hospital, Gwangju 61452, Republic of Korea
- Department of Premedical Science, Gwangju 61452, Republic of Korea
- Department of Orthopaedic Surgery, College of Medicine, Gwangju 61452, Republic of Korea
| |
Collapse
|
5
|
Lines KE, Gluck AK, Thongjuea S, Bountra C, Thakker RV, Gorvin CM. The bromodomain inhibitor JQ1+ reduces calcium-sensing receptor activity in pituitary cell lines. J Mol Endocrinol 2021; 67:83-94. [PMID: 34223822 PMCID: PMC8345903 DOI: 10.1530/jme-21-0030] [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: 06/30/2021] [Accepted: 07/05/2021] [Indexed: 12/05/2022]
Abstract
Corticotrophinomas represent 10% of all surgically removed pituitary adenomas, however, current treatment options are often not effective, and there is a need for improved pharmacological treatments. Recently, JQ1+, a bromodomain inhibitor that promotes gene transcription by binding acetylated histone residues and recruiting transcriptional machinery, has been shown to reduce proliferation in a murine corticotroph cell line, AtT20. RNA-Seq analysis of AtT20 cells following treatment with JQ1+ identified the calcium-sensing receptor (CaSR) gene as significantly downregulated, which was subsequently confirmed using real-time PCR and Western blot analysis. CaSR is a G protein-coupled receptor that plays a central role in calcium homeostasis but can elicit non-calcitropic effects in multiple tissues, including the anterior pituitary where it helps regulate hormone secretion. However, in AtT20 cells, CaSR activates a tumour-specific cAMP pathway that promotes ACTH and PTHrP hypersecretion. We hypothesised that the Casr promoter may harbour binding sites for BET proteins, and using chromatin immunoprecipitation (ChIP)-sequencing demonstrated that the BET protein Brd3 binds to the promoter of the Casr gene. Assessment of CaSR signalling showed that JQ1+ significantly reduced Ca2+e-mediated increases in intracellular calcium (Ca2+i) mobilisation and cAMP signalling. However, the CaSR-negative allosteric modulator, NPS-2143, was unable to reduce AtT20 cell proliferation, indicating that reducing CaSR expression rather than activity is likely required to reduce pituitary cell proliferation. Thus, these studies demonstrate that reducing CaSR expression may be a viable option in the treatment of pituitary tumours. Moreover, current strategies to reduce CaSR activity, rather than protein expression for cancer treatments, may be ineffective.
Collapse
Affiliation(s)
- Kate E Lines
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford,UK
- Correspondence should be addressed to K E Lines or C M Gorvin: or
| | - Anna K Gluck
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford,UK
| | - Supat Thongjuea
- Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Chas Bountra
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rajesh V Thakker
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford,UK
| | - Caroline M Gorvin
- Academic Endocrine Unit, Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford,UK
- Institute of Metabolism and Systems Research and Centre for Endocrinology, Diabetes and Metabolism, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK
- Correspondence should be addressed to K E Lines or C M Gorvin: or
| |
Collapse
|
6
|
Martin TJ, Sims NA, Seeman E. Physiological and Pharmacological Roles of PTH and PTHrP in Bone Using Their Shared Receptor, PTH1R. Endocr Rev 2021; 42:383-406. [PMID: 33564837 DOI: 10.1210/endrev/bnab005] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Indexed: 12/13/2022]
Abstract
Parathyroid hormone (PTH) and the paracrine factor, PTH-related protein (PTHrP), have preserved in evolution sufficient identities in their amino-terminal domains to share equivalent actions upon a common G protein-coupled receptor, PTH1R, that predominantly uses the cyclic adenosine monophosphate-protein kinase A signaling pathway. Such a relationship between a hormone and local factor poses questions about how their common receptor mediates pharmacological and physiological actions of the two. Mouse genetic studies show that PTHrP is essential for endochondral bone lengthening in the fetus and is essential for bone remodeling. In contrast, the main postnatal function of PTH is hormonal control of calcium homeostasis, with no evidence that PTHrP contributes. Pharmacologically, amino-terminal PTH and PTHrP peptides (teriparatide and abaloparatide) promote bone formation when administered by intermittent (daily) injection. This anabolic effect is remodeling-based with a lesser contribution from modeling. The apparent lesser potency of PTHrP than PTH peptides as skeletal anabolic agents could be explained by lesser bioavailability to PTH1R. By contrast, prolongation of PTH1R stimulation by excessive dosing or infusion, converts the response to a predominantly resorptive one by stimulating osteoclast formation. Physiologically, locally generated PTHrP is better equipped than the circulating hormone to regulate bone remodeling, which occurs asynchronously at widely distributed sites throughout the skeleton where it is needed to replace old or damaged bone. While it remains possible that PTH, circulating within a narrow concentration range, could contribute in some way to remodeling and modeling, its main physiological role is in regulating calcium homeostasis.
Collapse
Affiliation(s)
- T John Martin
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Natalie A Sims
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia.,The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Ego Seeman
- The University of Melbourne, Department of Medicine at Austin Health, Heidelberg, Victoria, Australia
| |
Collapse
|
7
|
Komaba H, Ketteler M, Cunningham J, Fukagawa M. Old and New Drugs for the Management of Bone Disorders in CKD. Calcif Tissue Int 2021; 108:486-495. [PMID: 33386480 DOI: 10.1007/s00223-020-00788-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/04/2020] [Indexed: 12/20/2022]
Abstract
Disturbances in mineral and bone metabolism are common in patients with chronic kidney disease (CKD), especially those undergoing dialysis. Renal osteodystrophy, which describes an alteration of bone morphology, is an important component of this systemic disorder and may explain the elevated risk of fracture which adversely affects morbidity and mortality. The most common form of renal osteodystrophy is high-turnover bone disease (osteitis fibrosa), which is induced by secondary hyperparathyroidism (SHPT). During the past decade, there has been considerable advances in the management of SHPT, with the introduction of the calcimimetic agents, the optimized use of nutritional and active vitamin D, and the accumulated experience with surgical parathyroidectomy. Studies supported that these advances could translate into improvement of renal bone disease and fracture prevention, as well as decreasing the risk of cardiovascular events and mortality. In this review, we summarize the available clinical evidence on the effect of old and new drugs on bone disorders in patients with CKD.
Collapse
Affiliation(s)
- Hirotaka Komaba
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, 143 Shimo-Kasuya, Isehara, 259-1193, Japan
- The Institute of Medical Sciences, Tokai University, Isehara, Japan
| | - Markus Ketteler
- Department of General Internal Medicine and Nephrology, Robert-Bosch-Krankenhaus, Stuttgart, Germany
| | | | - Masafumi Fukagawa
- Division of Nephrology, Endocrinology and Metabolism, Tokai University School of Medicine, 143 Shimo-Kasuya, Isehara, 259-1193, Japan.
| |
Collapse
|
8
|
Diao J, DeBono A, Josephs TM, Bourke JE, Capuano B, Gregory KJ, Leach K. Therapeutic Opportunities of Targeting Allosteric Binding Sites on the Calcium-Sensing Receptor. ACS Pharmacol Transl Sci 2021; 4:666-679. [PMID: 33860192 DOI: 10.1021/acsptsci.1c00046] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Indexed: 01/24/2023]
Abstract
The CaSR is a class C G protein-coupled receptor (GPCR) that acts as a multimodal chemosensor to maintain diverse homeostatic functions. The CaSR is a clinical therapeutic target in hyperparathyroidism and has emerged as a putative target in several other diseases. These include hyper- and hypocalcaemia caused either by mutations in the CASR gene or in genes that regulate CaSR signaling and expression, and more recently in asthma. The development of CaSR-targeting drugs is complicated by the fact that the CaSR possesses many different binding sites for endogenous and exogenous agonists and allosteric modulators. Binding sites for endogenous and exogenous ligands are located throughout the large CaSR protein and are interconnected in ways that we do not yet fully understand. This review summarizes our current understanding of CaSR physiology, signaling, and structure and how the many different binding sites of the CaSR may be targeted to treat disease.
Collapse
Affiliation(s)
- Jiayin Diao
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Aaron DeBono
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.,Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Tracy M Josephs
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Jane E Bourke
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, 9 Ancora Imparo Way, Clayton, Victoria 3800, Australia
| | - Ben Capuano
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Karen J Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.,Department of Pharmacology, Biomedicine Discovery Institute, Monash University, 9 Ancora Imparo Way, Clayton, Victoria 3800, Australia
| | - Katie Leach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria 3052, Australia.,Department of Pharmacology, Biomedicine Discovery Institute, Monash University, 9 Ancora Imparo Way, Clayton, Victoria 3800, Australia
| |
Collapse
|
9
|
Li X, Chen S, Hu Z, Chen D, Wang J, Li Z, Li Z, Cui H, Dai G, Liu L, Wang H, Zhang K, Zheng Z, Zhan Z, Liu H. Aberrant upregulation of CaSR promotes pathological new bone formation in ankylosing spondylitis. EMBO Mol Med 2020; 12:e12109. [PMID: 33259138 PMCID: PMC7721361 DOI: 10.15252/emmm.202012109] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 12/28/2022] Open
Abstract
Pathological new bone formation is a typical pathological feature in ankylosing spondylitis (AS), and the underlying molecular mechanism remains elusive. Previous studies have shown that the calcium‐sensing receptor (CaSR) is critical for osteogenic differentiation while also being highly involved in many inflammatory diseases. However, whether it plays a role in pathological new bone formation of AS has not been reported. Here, we report the first piece of evidence that expression of CaSR is aberrantly upregulated in entheseal tissues collected from AS patients and animal models with different hypothetical types of pathogenesis. Systemic inhibition of CaSR reduced the incidence of pathological new bone formation and the severity of the ankylosing phenotype in animal models. Activation of PLCγ signalling by CaSR promoted bone formation both in vitro and in vivo. In addition, various inflammatory cytokines induced upregulation of CaSR through NF‐κB/p65 and JAK/Stat3 pathways in osteoblasts. These novel findings suggest that inflammation‐induced aberrant upregulation of CaSR and activation of CaSR‐PLCγ signalling in osteoblasts act as mediators of inflammation, affecting pathological new bone formation in AS.
Collapse
Affiliation(s)
- Xiang Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Siwen Chen
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Zaiying Hu
- Department of Rheumatology and Immunology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dongying Chen
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jianru Wang
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Zemin Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Zihao Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Haowen Cui
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Guo Dai
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Lei Liu
- Department of Spine Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haitao Wang
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Kuibo Zhang
- Department of Spine Surgery, The Fifth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhaomin Zheng
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| | - Zhongping Zhan
- Department of Rheumatology and Immunology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hui Liu
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.,Guangdong Province Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
| |
Collapse
|
10
|
Ahmad R, Dalziel JE. G Protein-Coupled Receptors in Taste Physiology and Pharmacology. Front Pharmacol 2020; 11:587664. [PMID: 33390961 PMCID: PMC7774309 DOI: 10.3389/fphar.2020.587664] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 10/09/2020] [Indexed: 12/14/2022] Open
Abstract
Heterotrimeric G protein-coupled receptors (GPCRs) comprise the largest receptor family in mammals and are responsible for the regulation of most physiological functions. Besides mediating the sensory modalities of olfaction and vision, GPCRs also transduce signals for three basic taste qualities of sweet, umami (savory taste), and bitter, as well as the flavor sensation kokumi. Taste GPCRs reside in specialised taste receptor cells (TRCs) within taste buds. Type I taste GPCRs (TAS1R) form heterodimeric complexes that function as sweet (TAS1R2/TAS1R3) or umami (TAS1R1/TAS1R3) taste receptors, whereas Type II are monomeric bitter taste receptors or kokumi/calcium-sensing receptors. Sweet, umami and kokumi receptors share structural similarities in containing multiple agonist binding sites with pronounced selectivity while most bitter receptors contain a single binding site that is broadly tuned to a diverse array of bitter ligands in a non-selective manner. Tastant binding to the receptor activates downstream secondary messenger pathways leading to depolarization and increased intracellular calcium in TRCs, that in turn innervate the gustatory cortex in the brain. Despite recent advances in our understanding of the relationship between agonist binding and the conformational changes required for receptor activation, several major challenges and questions remain in taste GPCR biology that are discussed in the present review. In recent years, intensive integrative approaches combining heterologous expression, mutagenesis and homology modeling have together provided insight regarding agonist binding site locations and molecular mechanisms of orthosteric and allosteric modulation. In addition, studies based on transgenic mice, utilizing either global or conditional knock out strategies have provided insights to taste receptor signal transduction mechanisms and their roles in physiology. However, the need for more functional studies in a physiological context is apparent and would be enhanced by a crystallized structure of taste receptors for a more complete picture of their pharmacological mechanisms.
Collapse
Affiliation(s)
- Raise Ahmad
- Food Nutrition and Health Team, Food and Bio-based Products Group, AgResearch, Palmerston North, New Zealand
| | - Julie E Dalziel
- Food Nutrition and Health Team, Food and Bio-based Products Group, AgResearch, Palmerston North, New Zealand
| |
Collapse
|
11
|
Liu W, Sun J, Guo Y, Liu N, Ding X, Zhang X, Chi J, Kang N, Liu Y, Yin X. Calhex231 ameliorates myocardial fibrosis post myocardial infarction in rats through the autophagy-NLRP3 inflammasome pathway in macrophages. J Cell Mol Med 2020; 24:13440-13453. [PMID: 33043596 PMCID: PMC7701583 DOI: 10.1111/jcmm.15969] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/16/2020] [Accepted: 09/22/2020] [Indexed: 12/27/2022] Open
Abstract
The calcium‐sensing receptor (CaSR) is involved in the pathophysiology of many cardiovascular diseases, including myocardial infarction (MI) and hypertension. The role of Calhex231, a specific inhibitor of CaSR, in myocardial fibrosis following MI is still unclear. Using Wistar rats, we investigated whether Calhex231 ameliorates myocardial fibrosis through the autophagy‐NLRP3 inflammasome pathway in macrophages post myocardial infarction (MI). The rats were randomly divided into sham, MI and MI + Calhex231 groups. Compared with the sham rats, the MI rats consistently developed severe cardiac function, myocardial fibrosis and infiltration of inflammatory cells including macrophages. Moreover, inflammatory pathway including activation of NLRP3 inflammasome, IL‐1β and autophagy was significantly up‐regulated in myocardial tissue, infiltrated cardiac macrophages and peritoneal macrophages of the MI rats. These impacts were reversed by Calhex231. In vitro, studies revealed that calindol and rapamycin exacerbated MI‐induced autophagy and NLRP3 inflammasome activation in peritoneal macrophages. Calhex231 and 3‐Methyladenine (a specific inhibitor of autophagy) attenuated both autophagy and NLRP3 inflammasome activation; however, the caspase‐1 inhibitor Z‐YVAD‐FMK did not. Our study indicated that Calhex231 improved cardiac function and ameliorated myocardial fibrosis post MI, likely via the inhibition of autophagy‐mediated NLRP3 inflammasome activation; this provides a new therapeutic target for ventricular remodelling‐related cardiovascular diseases.
Collapse
Affiliation(s)
- Wenxiu Liu
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiaxing Sun
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yutong Guo
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Na Liu
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xue Ding
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Zhang
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinyu Chi
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ningning Kang
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yue Liu
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xinhua Yin
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
12
|
Hannan FM, Gorvin CM, Babinsky VN, Olesen MK, Stewart M, Wells S, Cox RD, Nemeth EF, Thakker RV. Calcilytic NPSP795 Increases Plasma Calcium and PTH in an Autosomal Dominant Hypocalcemia Type 1 Mouse Model. JBMR Plus 2020; 4:e10402. [PMID: 33103030 PMCID: PMC7574706 DOI: 10.1002/jbm4.10402] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 07/28/2020] [Indexed: 12/30/2022] Open
Abstract
Calcilytics are calcium‐sensing receptor (CaSR) antagonists that reduce the sensitivity of the CaSR to extracellular calcium. Calcilytics have the potential to treat autosomal dominant hypocalcemia type 1 (ADH1), which is caused by germline gain‐of‐function CaSR mutations and leads to symptomatic hypocalcemia, inappropriately low PTH concentrations, and hypercalciuria. To date, only one calcilytic compound, NPSP795, has been evaluated in patients with ADH1: Doses of up to 30 mg per patient have been shown to increase PTH concentrations, but did not significantly alter ionized blood calcium concentrations. The aim of this study was to further investigate NPSP795 for the treatment of ADH1 by undertaking in vitro and in vivo studies involving Nuf mice, which have hypocalcemia in association with a gain‐of‐function CaSR mutation, Leu723Gln. Treatment of HEK293 cells stably expressing the mutant Nuf (Gln723) CaSR with 20nM NPSP795 decreased extracellular Ca2+‐mediated intracellular calcium and phosphorylated ERK responses. An in vivo dose‐ranging study was undertaken by administering a s.c. bolus of NPSP795 at doses ranging from 0 to 30 mg/kg to heterozygous (Casr+/Nuf) and to homozygous (CasrNuf/Nuf) mice, and measuring plasma PTH responses at 30 min postdose. NPSP795 significantly increased plasma PTH concentrations in a dose‐dependent manner with the 30 mg/kg dose causing a maximal (≥10‐fold) rise in PTH. To determine whether NPSP795 can rectify the hypocalcemia of Casr+/Nuf and CasrNuf/Nuf mice, a submaximal dose (25 mg/kg) was administered, and plasma adjusted‐calcium concentrations measured over a 6‐hour period. NPSP795 significantly increased plasma adjusted‐calcium in Casr+/Nuf mice from 1.87 ± 0.03 mmol/L to 2.16 ± 0.06 mmol/L, and in CasrNuf/Nuf mice from 1.70 ± 0.03 mmol/L to 1.89 ± 0.05 mmol/L. Our findings show that NPSP795 elicits dose‐dependent increases in PTH and ameliorates the hypocalcemia in an ADH1 mouse model. Thus, calcilytics such as NPSP795 represent a potential targeted therapy for ADH1. © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Fadil M Hannan
- Academic Endocrine Unit, Radcliffe Department of Medicine Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford Oxford UK
| | - Caroline M Gorvin
- Academic Endocrine Unit, Radcliffe Department of Medicine Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford Oxford UK
| | - Valerie N Babinsky
- Academic Endocrine Unit, Radcliffe Department of Medicine Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford Oxford UK
| | - Mie K Olesen
- Academic Endocrine Unit, Radcliffe Department of Medicine Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford Oxford UK
| | - Michelle Stewart
- MRC Mammalian Genetics Unit and Mary Lyon Centre MRC Harwell Institute, Harwell Science and Innovation Campus Oxford UK
| | - Sara Wells
- MRC Mammalian Genetics Unit and Mary Lyon Centre MRC Harwell Institute, Harwell Science and Innovation Campus Oxford UK
| | - Roger D Cox
- MRC Mammalian Genetics Unit and Mary Lyon Centre MRC Harwell Institute, Harwell Science and Innovation Campus Oxford UK
| | | | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine Oxford Centre for Diabetes, Endocrinology and Metabolism (OCDEM), University of Oxford Oxford UK
| |
Collapse
|
13
|
Illuminating the allosteric modulation of the calcium-sensing receptor. Proc Natl Acad Sci U S A 2020; 117:21711-21722. [PMID: 32817431 DOI: 10.1073/pnas.1922231117] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Many membrane receptors are regulated by nutrients. However, how these nutrients control a single receptor remains unknown, even in the case of the well-studied calcium-sensing receptor CaSR, which is regulated by multiple factors, including ions and amino acids. Here, we developed an innovative cell-free Förster resonance energy transfer (FRET)-based conformational CaSR biosensor to clarify the main conformational changes associated with activation. By allowing a perfect control of ambient nutrients, this assay revealed that Ca2+ alone fully stabilizes the active conformation, while amino acids behave as pure positive allosteric modulators. Based on the identification of Ca2+ activation sites, we propose a molecular basis for how these different ligands cooperate to control CaSR activation. Our results provide important information on CaSR function and improve our understanding of the effects of genetic mutations responsible for human diseases. They also provide insights into how a receptor can integrate signals from various nutrients to better adapt to the cell response.
Collapse
|
14
|
Leach K, Hannan FM, Josephs TM, Keller AN, Møller TC, Ward DT, Kallay E, Mason RS, Thakker RV, Riccardi D, Conigrave AD, Bräuner-Osborne H. International Union of Basic and Clinical Pharmacology. CVIII. Calcium-Sensing Receptor Nomenclature, Pharmacology, and Function. Pharmacol Rev 2020; 72:558-604. [PMID: 32467152 PMCID: PMC7116503 DOI: 10.1124/pr.119.018531] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The calcium-sensing receptor (CaSR) is a class C G protein-coupled receptor that responds to multiple endogenous agonists and allosteric modulators, including divalent and trivalent cations, L-amino acids, γ-glutamyl peptides, polyamines, polycationic peptides, and protons. The CaSR plays a critical role in extracellular calcium (Ca2+ o) homeostasis, as demonstrated by the many naturally occurring mutations in the CaSR or its signaling partners that cause Ca2+ o homeostasis disorders. However, CaSR tissue expression in mammals is broad and includes tissues unrelated to Ca2+ o homeostasis, in which it, for example, regulates the secretion of digestive hormones, airway constriction, cardiovascular effects, cellular differentiation, and proliferation. Thus, although the CaSR is targeted clinically by the positive allosteric modulators (PAMs) cinacalcet, evocalcet, and etelcalcetide in hyperparathyroidism, it is also a putative therapeutic target in diabetes, asthma, cardiovascular disease, and cancer. The CaSR is somewhat unique in possessing multiple ligand binding sites, including at least five putative sites for the "orthosteric" agonist Ca2+ o, an allosteric site for endogenous L-amino acids, two further allosteric sites for small molecules and the peptide PAM, etelcalcetide, and additional sites for other cations and anions. The CaSR is promiscuous in its G protein-coupling preferences, and signals via Gq/11, Gi/o, potentially G12/13, and even Gs in some cell types. Not surprisingly, the CaSR is subject to biased agonism, in which distinct ligands preferentially stimulate a subset of the CaSR's possible signaling responses, to the exclusion of others. The CaSR thus serves as a model receptor to study natural bias and allostery. SIGNIFICANCE STATEMENT: The calcium-sensing receptor (CaSR) is a complex G protein-coupled receptor that possesses multiple orthosteric and allosteric binding sites, is subject to biased signaling via several different G proteins, and has numerous (patho)physiological roles. Understanding the complexities of CaSR structure, function, and biology will aid future drug discovery efforts seeking to target this receptor for a diversity of diseases. This review summarizes what is known to date regarding key structural, pharmacological, and physiological features of the CaSR.
Collapse
Affiliation(s)
- Katie Leach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Fadil M Hannan
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Tracy M Josephs
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Andrew N Keller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Thor C Møller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Donald T Ward
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Enikö Kallay
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Rebecca S Mason
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Rajesh V Thakker
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Daniela Riccardi
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Arthur D Conigrave
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Hans Bräuner-Osborne
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| |
Collapse
|
15
|
Guo D, He H, Hou T. Purification and characterization of positive allosteric regulatory peptides of calcium sensing receptor (CaSR) from desalted duck egg white. Food Chem 2020; 325:126919. [PMID: 32387992 DOI: 10.1016/j.foodchem.2020.126919] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/21/2020] [Accepted: 04/25/2020] [Indexed: 02/07/2023]
Abstract
HPLC-ESI-MS/MS, molecular docking simulation and in situ single-pass intestinal perfusion (SPIP) study were used to identify, select, and confirm the binding affinities between peptides identified from desalted duck egg white peptides (DPs) and calcium sensing receptor (CaSR), respectively. F3 fraction from DPs possessed superior calcium binding activity (P < 0.05), and 16 peptides enriched aromatic amino acids and other 33 peptides were identified. FAE, FNE, INSW, FDPE and NFE presented well binding affinities with CaSR in molecular docking. Additionally, SPIP results showed that NFE and INSW significantly reduced the increased PTH levels by 45.8% and 48.8%, respectively (P < 0.05), and increased calcium percent absorption, calcium absorption rate constant (Ka) and calcium effective permeability (Peff) (P < 0.05), as well as up-regulated mRNA levels of CaSR (P < 0.05). Moreover, NFE and INSW could interact with the VFT domain of CaSR, which exhibited the potential activities in regulation of CaSR.
Collapse
Affiliation(s)
- Danjun Guo
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Hui He
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| | - Tao Hou
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
16
|
Josephs TM, Keller AN, Khajehali E, DeBono A, Langmead CJ, Conigrave AD, Capuano B, Kufareva I, Gregory KJ, Leach K. Negative allosteric modulators of the human calcium-sensing receptor bind to overlapping and distinct sites within the 7-transmembrane domain. Br J Pharmacol 2020; 177:1917-1930. [PMID: 31881094 PMCID: PMC7070164 DOI: 10.1111/bph.14961] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 12/06/2019] [Accepted: 12/09/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND PURPOSE Negative allosteric modulators (NAMs) that target the calcium-sensing receptor (CaS receptor) were originally developed for the treatment of osteoporosis by stimulating the release of endogenous parathyroid hormone, but failed in human clinical trials. Several chemically and structurally distinct NAM scaffolds have been described, but it is not known how these different scaffolds interact with the CaS receptor to inhibit receptor signalling in response to agonists. EXPERIMENTAL APPROACH In the present study, we used a mutagenesis approach combined with analytical pharmacology and computational modelling to probe the binding sites of four distinct NAM scaffolds. KEY RESULTS Although all four scaffolds bind to the 7-transmembrane and/or extracellular or intracellular loops, they occupy distinct regions, as previously shown for positive allosteric modulators of the CaS receptor. Furthermore, different NAM scaffolds mediate negative allosteric modulation via distinct amino acid networks. CONCLUSION AND IMPLICATIONS These findings aid our understanding of how different NAMs bind to and inhibit the CaS receptor. Elucidation of allosteric binding sites in the CaS receptor has implications for the discovery of novel allosteric modulators.
Collapse
Affiliation(s)
- Tracy M. Josephs
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| | - Andrew N. Keller
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| | - Elham Khajehali
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| | - Aaron DeBono
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| | - Christopher J. Langmead
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| | - Arthur D. Conigrave
- School of Life and Environmental SciencesUniversity of SydneySydneyNSWAustralia
| | - Ben Capuano
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| | - Irina Kufareva
- Skaggs School of Pharmacy & Pharmaceutical SciencesUniversity of CaliforniaSan DiegoCAUSA
| | - Karen J. Gregory
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| | - Katie Leach
- Drug Discovery Biology and Department of Pharmacology, Monash Institute of Pharmaceutical SciencesMonash UniversityParkvilleVICAustralia
| |
Collapse
|
17
|
Das S, Clézardin P, Kamel S, Brazier M, Mentaverri R. The CaSR in Pathogenesis of Breast Cancer: A New Target for Early Stage Bone Metastases. Front Oncol 2020; 10:69. [PMID: 32117726 PMCID: PMC7013091 DOI: 10.3389/fonc.2020.00069] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/15/2020] [Indexed: 12/11/2022] Open
Abstract
The Ca2+-sensing receptor (CaSR) is a class-C G protein-coupled receptor which plays a pivotal role in calciotropic processes, primarily in regulating parathyroid hormone secretion to maintain systemic calcium homeostasis. Among its non-calciotropic roles, where the CaSR sits at the intersection of myriad processes, it has steadily garnered attention as an oncogene or tumor suppressor in different organs. In maternal breast tissues the CaSR promotes lactation but in breast cancer it acts as an oncoprotein and has been shown to drive the pathogenesis of skeletal metastases from breast cancer. Even though research has made great strides in treating primary breast cancer, there is an unmet need when it comes to treatment of metastatic breast cancer. This review focuses on how the CaSR leads to the pathogenesis of breast cancer by contrasting its role in healthy tissues and tumorigenesis, and by drawing brief parallels with the tissues where it has been implicated as an oncogene. A class of compounds called calcilytics, which are CaSR antagonists, have also been surveyed in the instances where they have been used to target the receptor in cancerous tissues and constitute a proof of principle for repurposing them. Current clinical therapies for treating bone metastases from breast cancer are limited to targeting osteoclasts and a deeper understanding of the CaSR signaling nexus in this context can bolster them or lead to novel therapeutic interventions.
Collapse
Affiliation(s)
- Souvik Das
- MP3CV, EA7517, CURS, University of Picardie Jules Verne, Amiens, France
| | - Philippe Clézardin
- INSERM, Research Unit UMR_S1033, LyOS, Faculty of Medicine Lyon-Est, University of Lyon 1, Lyon, France
- Department of Oncology and Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Said Kamel
- MP3CV, EA7517, CURS, University of Picardie Jules Verne, Amiens, France
- Department of Biochemistry, Amiens-Picardie University Hospital, Amiens, France
- Faculty of Pharmacy, University of Picardie Jules Verne, Amiens, France
| | - Michel Brazier
- MP3CV, EA7517, CURS, University of Picardie Jules Verne, Amiens, France
- Department of Biochemistry, Amiens-Picardie University Hospital, Amiens, France
- Faculty of Pharmacy, University of Picardie Jules Verne, Amiens, France
| | - Romuald Mentaverri
- MP3CV, EA7517, CURS, University of Picardie Jules Verne, Amiens, France
- Department of Biochemistry, Amiens-Picardie University Hospital, Amiens, France
- Faculty of Pharmacy, University of Picardie Jules Verne, Amiens, France
| |
Collapse
|
18
|
Tokonami N, Olinger E, Debaix H, Houillier P, Devuyst O. The excretion of uromodulin is modulated by the calcium-sensing receptor. Kidney Int 2019; 94:882-886. [PMID: 30348305 DOI: 10.1016/j.kint.2018.07.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 06/28/2018] [Accepted: 07/05/2018] [Indexed: 11/16/2022]
Abstract
Uromodulin is produced in the thick ascending limb, but little is known about regulation of its excretion in urine. Using mouse and cellular models, we demonstrate that excretion of uromodulin by thick ascending limb cells is increased or decreased upon inactivation or activation of the calcium-sensing receptor (CaSR), respectively. These effects reflect changes in uromodulin trafficking and likely involve alterations in intracellular cyclic adenosine monophosphate (cAMP) levels. Administration of the CaSR agonist cinacalcet led to a rapid reduction of urinary uromodulin excretion in healthy subjects. Modulation of uromodulin excretion by the CaSR may be clinically relevant considering the increasing use of CaSR modulators.
Collapse
Affiliation(s)
- Natsuko Tokonami
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Eric Olinger
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Huguette Debaix
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Pascal Houillier
- Sorbonne Université, UPMC Univ Paris 06, INSERM, Université Paris Descartes, UMR_S 1138 Team 3, Centre de Recherche des Cordeliers, Paris, France
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
19
|
Szczoczarz A, Marchwińska A, Dyś A, Boblewski K, Lehmann A, Lewko B, Rybczyńska A. Verapamil prevents the effect of calcium-sensing receptor activation on the blood glucose and insulin levels in rats. Pharmacol Rep 2019; 71:478-484. [PMID: 31003161 DOI: 10.1016/j.pharep.2019.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/14/2018] [Accepted: 01/04/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND The Ca2+ triggered insulin exocytosis in β cells of the pancreatic islets may be the result of Ca2+ influx through L-type voltage dependent calcium channels (VDCC) localized in the plasma membrane, as well as of liberation of Ca2+ from intracellular storages, induced by activation of the calcium receptor (CaR) coupled with the PLC enzyme present in the pancreatic islets. The present study was designated to determine, in in vivo experiments, the effects of CaR activation by R-568 and inhibition of the receptor by NPS 2143 on the plasma glucose and insulin levels in the presence of verapamil, a calcium channel blocker. METHODS Wistar rats, after fasting for 14 h before the experiment, were anesthetized with inactin and loaded ip with 1 g/kg glucose. RESULTS In comparison to the control group, the verapamil-induced blockade of the calcium channels in glucose loaded animals increased the blood glucose level and decreased the insulin level, whereas CaR activation with R-568 induced opposite effects. However, in the presence of verapamil, R-568 did not change the concentration of glucose or insulin versus the control animals. Verapamil infusion did not alter elevated glucose concentration in the NPS 2143 animals. At the same time, verapamil reduced the plasma insulin level and potentiated the drop of insulin concentration induced by NPS 2143. CONCLUSION The observations suggest that under the in vivo conditions, calcium channel blockade may prevent changes in the blood glucose and insulin concentrations induced by the CaR activation.
Collapse
Affiliation(s)
- Anna Szczoczarz
- Department of Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Marchwińska
- Department of Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Dyś
- Department of Laboratory Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Konrad Boblewski
- Department of Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| | - Artur Lehmann
- Department of Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| | - Barbara Lewko
- Department of Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| | - Apolonia Rybczyńska
- Department of Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland.
| |
Collapse
|
20
|
Antagonism of Ca 2+-sensing receptors by NPS 2143 is transiently masked by p38 activation in mouse brain bEND.3 endothelial cells. Naunyn Schmiedebergs Arch Pharmacol 2019; 392:823-832. [PMID: 30826858 DOI: 10.1007/s00210-019-01637-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 02/22/2019] [Indexed: 10/27/2022]
Abstract
Ca2+-sensing receptors (CaSR) are G protein-coupled receptors which are activated by a rise in extracellular Ca2+. CaSR activation has been known to inhibit parathyroid hormone release and stimulate calcitonin release from parathyroid glands and thyroid parafollicular C cells, respectively. The roles of CaSR in other cell types including endothelial cells (EC) are much less understood. In this work, we demonstrated protein and functional expression of CaSR in mouse cerebral EC (bEND.3). Unexpectedly, CaSR response (high Ca2+-elicited cytosolic [Ca2+] elevation) was unaffected by edelfosine or U73122 but strongly suppressed by SK&F 96365, ruthenium red, and 2-aminoethoxydiphenyl borate (2-APB), suggesting involvement of TRPV and TRPC channels but not Gq-phospholipase C. Acute application of NPS2143, a negative allosteric modulator of CaSR, suppressed CaSR response. However, a 40-min NPS2143 pre-treatment surprisingly enhanced CaSR response. After 4-24 h of application, this enhancement faded away and suppression of CaSR response was observed again. Similar results were obtained when La3+ and Sr2+ were used as CaSR agonists. The transient NPS 2143 enhancement effect was abolished by SB203580, a p38 inhibitor. Consistently, NPS 2143 triggered a transient p38 activation. Taken together, results suggest that in bEND.3 cells, NPS 2143 caused acute suppression of CaSR response, but then elicited a transient enhancement of CaSR response in a p38-dependent manner. NPS 2143 effects on CaSR in bEND.3 cells therefore depended on drug exposure time. These findings warrant cautious use of this agent as a CaSR modulator and potential cardiovascular drug.
Collapse
|
21
|
Zhang X, Jin T, Shi N, Yao L, Yang X, Han C, Wen L, Du D, Szatmary P, Mukherjee R, Liu T, Xia Q, Criddle DN, Huang W, Chvanov M, Sutton R. Mechanisms of Pancreatic Injury Induced by Basic Amino Acids Differ Between L-Arginine, L-Ornithine, and L-Histidine. Front Physiol 2019; 9:1922. [PMID: 30697165 PMCID: PMC6341295 DOI: 10.3389/fphys.2018.01922] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 12/20/2018] [Indexed: 02/05/2023] Open
Abstract
Pancreatic acinar cells require high rates of amino acid uptake for digestive enzyme synthesis, but excessive concentrations can trigger acute pancreatitis (AP) by mechanisms that are not well understood. We have used three basic natural amino acids L-arginine, L-ornithine, and L-histidine to determine mechanisms of amino acid-induced pancreatic injury and whether these are common to all three amino acids. Caffeine markedly inhibited necrotic cell death pathway activation in isolated pancreatic acinar cells induced by L-arginine, but not L-ornithine, whereas caffeine accelerated L-histidine-induced cell death. Both necroptosis inhibitors of RIPK1 and RIPK3 and a necroptosis activator/apoptosis inhibitor z-VAD increased cell death caused by L-histidine, but not L-arginine or L-ornithine. Cyclophilin D knock-out (Ppif-/-) significantly attenuated cell death induced by L-histidine, but not L-arginine, or L-ornithine. Allosteric modulators of calcium-sensing receptor (CaSR) and G-protein coupled receptor class C group 6 member A (GPRC6A) had inhibitory effects on cell death induced by L-arginine but not L-ornithine or L-histidine. We developed a novel amino acid-induced AP murine model with high doses of L-histidine and confirmed AP severity was significantly reduced in Ppif-/- vs. wild type mice. In L-arginine-induced AP neither Ppif-/-, caffeine, or allosteric modulators of CaSR or GPRC6A reduced pancreatic damage, even though CaSR inhibition with NPS-2143 significantly reduced pancreatic and systemic injury in caerulein-induced AP. These findings demonstrate marked differences in the mechanisms of pancreatic injury induced by different basic amino acids and suggest the lack of effect of treatments on L-arginine-induced AP may be due to conversion to L-ornithine in the urea cycle.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
- Liverpool Pancreatitis Study Group, Royal Liverpool University Hospital, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Tao Jin
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
- Liverpool Pancreatitis Study Group, Royal Liverpool University Hospital, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Na Shi
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Linbo Yao
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Xinmin Yang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Chenxia Han
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Li Wen
- Liverpool Pancreatitis Study Group, Royal Liverpool University Hospital, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Dan Du
- West China-Washington Mitochondria and Metabolism Centre, West China Hospital of Sichuan University, Chengdu, China
| | - Peter Szatmary
- Liverpool Pancreatitis Study Group, Royal Liverpool University Hospital, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Rajarshi Mukherjee
- Liverpool Pancreatitis Study Group, Royal Liverpool University Hospital, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Tingting Liu
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Xia
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - David N. Criddle
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, United Kingdom
| | - Wei Huang
- Department of Integrated Traditional Chinese and Western Medicine, Sichuan Provincial Pancreatitis Centre and West China-Liverpool Biomedical Research Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Michael Chvanov
- Department of Cellular and Molecular Physiology, University of Liverpool, Liverpool, United Kingdom
| | - Robert Sutton
- Liverpool Pancreatitis Study Group, Royal Liverpool University Hospital, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
22
|
Hannan FM, Kallay E, Chang W, Brandi ML, Thakker RV. The calcium-sensing receptor in physiology and in calcitropic and noncalcitropic diseases. Nat Rev Endocrinol 2018; 15:33-51. [PMID: 30443043 PMCID: PMC6535143 DOI: 10.1038/s41574-018-0115-0] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Ca2+-sensing receptor (CaSR) is a dimeric family C G protein-coupled receptor that is expressed in calcitropic tissues such as the parathyroid glands and the kidneys and signals via G proteins and β-arrestin. The CaSR has a pivotal role in bone and mineral metabolism, as it regulates parathyroid hormone secretion, urinary Ca2+ excretion, skeletal development and lactation. The importance of the CaSR for these calcitropic processes is highlighted by loss-of-function and gain-of-function CaSR mutations that cause familial hypocalciuric hypercalcaemia and autosomal dominant hypocalcaemia, respectively, and also by the fact that alterations in parathyroid CaSR expression contribute to the pathogenesis of primary and secondary hyperparathyroidism. Moreover, the CaSR is an established therapeutic target for hyperparathyroid disorders. The CaSR is also expressed in organs not involved in Ca2+ homeostasis: it has noncalcitropic roles in lung and neuronal development, vascular tone, gastrointestinal nutrient sensing, wound healing and secretion of insulin and enteroendocrine hormones. Furthermore, the abnormal expression or function of the CaSR is implicated in cardiovascular and neurological diseases, as well as in asthma, and the CaSR is reported to protect against colorectal cancer and neuroblastoma but increase the malignant potential of prostate and breast cancers.
Collapse
Affiliation(s)
- Fadil M Hannan
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Enikö Kallay
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Wenhan Chang
- Endocrine Research Unit, Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, CA, USA
| | - Maria Luisa Brandi
- Metabolic Bone Diseases Unit, Department of Surgery and Translational Medicine, University of Florence, Florence, Italy.
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
23
|
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: 11] [Impact Index Per Article: 1.8] [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
|
24
|
Wang S, Qiu L, Song H, Dang N. NPS - 2143 (hydrochloride) inhibits melanoma cancer cell proliferation and induces autophagy and apoptosis. Med Sci (Paris) 2018; 34 Focus issue F1:87-93. [PMID: 30403181 DOI: 10.1051/medsci/201834f115] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Melanoma is a common and aggressive skin cancer caused by the oncogenic transformation of melanocytes. NPS-2143 (hydrochloride) is a calcification drug that acts as an antagonist of the calcium-sensing receptor (CaSR) and consequently stimulates the release of parathyroid hormone. In the present work, we treated cells from the human melanoma cell line M14 to investigate the effects of NPS-2143 on melanoma cells and elucidate their underlying mechanisms. We observed that NPS-2143 inhibits the survival and proliferation of M14 cells and suppresses the migration and proliferation of M14 cells by inducing apoptosis. The Bax/Bcl‑2 ratio in M14 cells was enhanced by the NPS-2143 treatment, suggesting that the mitochondrial apoptotic pathway was activated. The expression and phosphorylation of proteins involved in the PI3K signaling pathway were altered by NPS-2143 treatment. Our data show that NPS-2143 impacts the viability and induces the apoptosis of melanoma M14 cells through its impact on the PI3K signaling pathway. It suggests that NPS-2143 could represent a promising candidate for melanoma treatment.
Collapse
Affiliation(s)
- Shumei Wang
- Department of Community Medicine, Jinan Central Hospital affiliated to Shandong University, No.105 Jiefang Road, Jinan 250013, Shandong Province, China
| | - Liyun Qiu
- Department of Pharmacy, Jinan Central Hospital affiliated to Shandong University, No.105 Jiefang Road, Jinan 250013, Shandong Province, China
| | - Haiyan Song
- Department of Dermatology, Jinan Central Hospital affiliated to Shandong University, No.105 Jiefang Road, Jinan 250013, Shandong Province, China
| | - Ningning Dang
- Department of Dermatology, Jinan Central Hospital affiliated to Shandong University, No.105 Jiefang Road, Jinan 250013, Shandong Province, China
| |
Collapse
|
25
|
Hannan FM, Olesen MK, Thakker RV. Calcimimetic and calcilytic therapies for inherited disorders of the calcium-sensing receptor signalling pathway. Br J Pharmacol 2018; 175:4083-4094. [PMID: 29127708 PMCID: PMC6177618 DOI: 10.1111/bph.14086] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 10/24/2017] [Accepted: 10/30/2017] [Indexed: 12/15/2022] Open
Abstract
The calcium-sensing receptor (CaS receptor) plays a pivotal role in extracellular calcium homeostasis, and germline loss-of-function and gain-of-function mutations cause familial hypocalciuric hypercalcaemia (FHH) and autosomal dominant hypocalcaemia (ADH), respectively. CaS receptor signal transduction in the parathyroid glands is probably regulated by G-protein subunit α11 (Gα11 ) and adaptor-related protein complex-2 σ-subunit (AP2σ), and recent studies have identified germline mutations of these proteins as a cause of FHH and/or ADH. Calcimimetics and calcilytics are positive and negative allosteric modulators of the CaS receptor that have potential efficacy for symptomatic forms of FHH and ADH. Cellular studies have demonstrated that these compounds correct signalling and/or trafficking defects caused by mutant CaS receptor, Gα11 or AP2σ proteins. Moreover, mouse model studies indicate that calcilytics can rectify the hypocalcaemia and hypercalciuria associated with ADH, and patient-based studies reveal calcimimetics to ameliorate symptomatic hypercalcaemia caused by FHH. Thus, calcimimetics and calcilytics represent targeted therapies for inherited disorders of the CaS receptor signalling pathway. LINKED ARTICLES This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.
Collapse
Affiliation(s)
- Fadil M Hannan
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic DiseaseUniversity of LiverpoolLiverpoolUK
- Academic Endocrine Unit, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Mie K Olesen
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic DiseaseUniversity of LiverpoolLiverpoolUK
- Academic Endocrine Unit, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of MedicineUniversity of OxfordOxfordUK
| |
Collapse
|
26
|
Nemeth EF, Van Wagenen BC, Balandrin MF. Discovery and Development of Calcimimetic and Calcilytic Compounds. PROGRESS IN MEDICINAL CHEMISTRY 2018; 57:1-86. [PMID: 29680147 DOI: 10.1016/bs.pmch.2017.12.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
The extracellular calcium receptor (CaR) is a G protein-coupled receptor (GPCR) and the pivotal molecule regulating systemic Ca2+ homeostasis. The CaR was a challenging target for drug discovery because its physiological ligand is an inorganic ion (Ca2+) rather than a molecule so there was no structural template to guide medicinal chemistry. Nonetheless, small molecules targeting this receptor were discovered. Calcimimetics are agonists or positive allosteric modulators of the CaR, while calcilytics are antagonists and all to date are negative allosteric modulators. The calcimimetic cinacalcet was the first allosteric modulator of a GPCR to achieve regulatory approval and is a first-in-class treatment for secondary hyperparathyroidism in patients on dialysis, and for hypercalcemia in some forms of primary hyperparathyroidism. It is also useful in treating some rare genetic diseases that cause hypercalcemia. Two other calcimimetics are now on the market (etelcalcetide) or under regulatory review (evocalcet). Calcilytics stimulate the secretion of parathyroid hormone and were initially developed as treatments for osteoporosis. Three different calcilytics of two different chemotypes failed in clinical trials due to lack of efficacy. Calcilytics are now being repurposed and might be useful in treating hypoparathyroidism and several rare genetic diseases causing hypocalcemia. The challenges ahead for medicinal chemists are to design compounds that select conformations of the CaR that preferentially target a particular signalling pathway and/or that affect the CaR in a tissue-selective manner.
Collapse
|
27
|
Diepenhorst N, Rueda P, Cook AE, Pastoureau P, Sabatini M, Langmead CJ. G protein-coupled receptors as anabolic drug targets in osteoporosis. Pharmacol Ther 2017; 184:1-12. [PMID: 29080701 DOI: 10.1016/j.pharmthera.2017.10.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Osteoporosis is a progressive bone disorder characterised by imbalance between bone building (anabolism) and resorption (catabolism). Most therapeutics target inhibition of osteoclast-mediated bone resorption, but more recent attention in early drug discovery has focussed on anabolic targets in osteoblasts or their precursors. Two marketed agents that display anabolic properties, strontium ranelate and teriparatide, mediate their actions via the G protein-coupled calcium-sensing and parathyroid hormone-1 receptors, respectively. This review explores their activity, the potential for improved therapeutics targeting these receptors and other putative anabolic GPCR targets, including Smoothened, Wnt/Frizzled, relaxin family peptide, adenosine, cannabinoid, prostaglandin and sphingosine-1-phosphate receptors.
Collapse
Affiliation(s)
- Natalie Diepenhorst
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, VIC 3052, Australia
| | - Patricia Rueda
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, VIC 3052, Australia
| | - Anna E Cook
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, VIC 3052, Australia
| | - Philippe Pastoureau
- Therapeutic Innovation Pole of Immuno-Inflammatory Diseases, Institut de Recherches Servier, Suresnes, France
| | - Massimo Sabatini
- Therapeutic Innovation Pole of Immuno-Inflammatory Diseases, Institut de Recherches Servier, Suresnes, France
| | - Christopher J Langmead
- Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, VIC 3052, Australia.
| |
Collapse
|
28
|
Abstract
Cancellous bone has high metabolic activity compared to many other bone compartments and can be affected not only by changes in physeal activity but also by perturbations in homeostasis caused by changes in physiology or on-target pharmacology. Examples of several types of resulting morphologic findings were presented; if known, the pathways causing morphologic changes were discussed.
Collapse
Affiliation(s)
- Kathryn E Gropp
- 1 Drug Safety Research and Development, Pfizer Inc., Groton, Connecticut, USA
| |
Collapse
|
29
|
Balenga N, Azimzadeh P, Hogue JA, Staats PN, Shi Y, Koh J, Dressman H, Olson JA. Orphan Adhesion GPCR GPR64/ADGRG2 Is Overexpressed in Parathyroid Tumors and Attenuates Calcium-Sensing Receptor-Mediated Signaling. J Bone Miner Res 2017; 32:654-666. [PMID: 27760455 PMCID: PMC7211037 DOI: 10.1002/jbmr.3023] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 09/30/2016] [Accepted: 10/17/2016] [Indexed: 12/19/2022]
Abstract
Abnormal feedback of serum calcium to parathyroid hormone (PTH) secretion is the hallmark of primary hyperparathyroidism (PHPT). Although the molecular pathogenesis of parathyroid neoplasia in PHPT has been linked to abnormal expression of genes involved in cell growth (e.g., cyclin D1, retinoblastoma, and β-catenin), the molecular basis of abnormal calcium sensing by calcium-sensing receptor (CaSR) and PTH hypersecretion in PHPT are incompletely understood. Through gene expression profiling, we discovered that an orphan adhesion G protein-coupled receptor (GPCR), GPR64/ADGRG2, is expressed in human normal parathyroid glands and is overexpressed in parathyroid tumors from patients with PHPT. Using immunohistochemistry, Western blotting, and coimmunoprecipitation, we found that GPR64 is expressed on the cell surface of parathyroid cells, is overexpressed in parathyroid tumors, and physically interacts with the CaSR. By using reporter gene assay and GPCR second messenger readouts we identified Gαs, 3',5'-cyclic adenosine monophosphate (cAMP), protein kinase A, and cAMP response element binding protein (CREB) as the signaling cascade downstream of GPR64. Furthermore, we found that an N-terminally truncated human GPR64 is constitutively active and a 15-amino acid-long peptide C-terminal to the GPCR proteolysis site (GPS) of GPR64 activates this receptor. Functional characterization of GPR64 demonstrated its ability to increase PTH release from human parathyroid cells at a range of calcium concentrations. We discovered that the truncated constitutively active, but not the full-length GPR64 physically interacts with CaSR and attenuates the CaSR-mediated intracellular Ca2+ signaling and cAMP suppression in HEK293 cells. Our results indicate that GPR64 may be a physiologic regulator of PTH release that is dysregulated in parathyroid tumors, and suggest a role for GPR64 in pathologic calcium sensing in PHPT. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Nariman Balenga
- Division of General & Oncologic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pedram Azimzadeh
- Division of General & Oncologic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joyce A Hogue
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Paul N Staats
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Yuhong Shi
- Division of General & Oncologic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - James Koh
- Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Holly Dressman
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | - John A Olson
- Division of General & Oncologic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
30
|
Rybczyńska A, Marchwińska A, Dyś A, Boblewski K, Lehmann A, Lewko B. Activity of the calcium-sensing receptor influences blood glucose and insulin levels in rats. Pharmacol Rep 2017; 69:709-713. [PMID: 28551530 DOI: 10.1016/j.pharep.2017.01.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 01/30/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND The calcium-sensing receptor (CaR) has been found not only in parathyroid glands but also in other tissues, e.g. in β cells of the pancreatic islets. Therefore, CaR might likely mediate the mechanism of insulin secretion. The present study was designed to examine the in vivo effects of R-568, a CaR agonist, and NPS2143, a CaR inhibitor, on plasma insulin and blood glucose concentrations. METHODS Wistar rats, after fasting for 14h before the experiment, were anesthetized with inactin and loaded ip with 1g/kg glucose. RESULTS 20, 120 and 180min after iv R-568 administration, plasma insulin increased markedly (by approximately 30%), in glucose-loaded rats, as compared to the control animals. Simultaneously, 180min after R-568 administration, a significant drop by approximately 12% in blood glucose was observed. In contrast, administration of R-568 in rats not given glucose, did not influence the blood glucose or plasma insulin concentrations vs. the control group. Administration of NPS2143 increased the blood glucose level markedly (by about 18% vs. control group) at 180 and 210min of the experiment. Simultaneously, a significant decrease of insulin concentration was observed vs. control group (by about 18 and 23%, respectively). CONCLUSION We suggest that modulation of the CaR activity may participate in the mechanisms which mediate insulin secretion in rats.
Collapse
Affiliation(s)
- Apolonia Rybczyńska
- Department of Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland.
| | - Aleksandra Marchwińska
- Department of Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Dyś
- Department of Laboratory Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Konrad Boblewski
- Department of Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| | - Artur Lehmann
- Department of Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| | - Barbara Lewko
- Department of Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
31
|
Gorvin CM, Hannan FM, Howles SA, Babinsky VN, Piret SE, Rogers A, Freidin AJ, Stewart M, Paudyal A, Hough TA, Nesbit MA, Wells S, Vincent TL, Brown SD, Cox RD, Thakker RV. G α11 mutation in mice causes hypocalcemia rectifiable by calcilytic therapy. JCI Insight 2017; 2:e91103. [PMID: 28194447 PMCID: PMC5291742 DOI: 10.1172/jci.insight.91103] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 01/03/2017] [Indexed: 12/11/2022] Open
Abstract
Heterozygous germline gain-of-function mutations of G-protein subunit α11 (Gα11), a signaling partner for the calcium-sensing receptor (CaSR), result in autosomal dominant hypocalcemia type 2 (ADH2). ADH2 may cause symptomatic hypocalcemia with low circulating parathyroid hormone (PTH) concentrations. Effective therapies for ADH2 are currently not available, and a mouse model for ADH2 would help in assessment of potential therapies. We hypothesized that a previously reported dark skin mouse mutant (Dsk7) - which has a germline hypermorphic Gα11 mutation, Ile62Val - may be a model for ADH2 and allow evaluation of calcilytics, which are CaSR negative allosteric modulators, as a targeted therapy for this disorder. Mutant Dsk7/+ and Dsk7/Dsk7 mice were shown to have hypocalcemia and reduced plasma PTH concentrations, similar to ADH2 patients. In vitro studies showed the mutant Val62 Gα11 to upregulate CaSR-mediated intracellular calcium and MAPK signaling, consistent with a gain of function. Treatment with NPS-2143, a calcilytic compound, normalized these signaling responses. In vivo, NPS-2143 induced a rapid and marked rise in plasma PTH and calcium concentrations in Dsk7/Dsk7 and Dsk7/+ mice, which became normocalcemic. Thus, these studies have established Dsk7 mice, which harbor a germline gain-of-function Gα11 mutation, as a model for ADH2 and have demonstrated calcilytics as a potential targeted therapy.
Collapse
Affiliation(s)
- Caroline M. Gorvin
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Fadil M. Hannan
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Sarah A. Howles
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Valerie N. Babinsky
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Sian E. Piret
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Angela Rogers
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Andrew J. Freidin
- ARUK Centre for Osteoarthritis Pathogenesis, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Michelle Stewart
- Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Harwell Science and Innovation Campus, United Kingdom
| | - Anju Paudyal
- Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Harwell Science and Innovation Campus, United Kingdom
| | - Tertius A. Hough
- Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Harwell Science and Innovation Campus, United Kingdom
| | - M. Andrew Nesbit
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- Biomedical Sciences Research Institute, Ulster University, Coleraine, United Kingdom
| | - Sara Wells
- Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Harwell Science and Innovation Campus, United Kingdom
| | - Tonia L. Vincent
- ARUK Centre for Osteoarthritis Pathogenesis, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Stephen D.M. Brown
- Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Harwell Science and Innovation Campus, United Kingdom
| | - Roger D. Cox
- Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre, MRC Harwell Institute, Harwell Science and Innovation Campus, United Kingdom
| | - Rajesh V. Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
32
|
Leach K, Gregory KJ. Molecular insights into allosteric modulation of Class C G protein-coupled receptors. Pharmacol Res 2017; 116:105-118. [DOI: 10.1016/j.phrs.2016.12.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 11/18/2016] [Accepted: 12/07/2016] [Indexed: 12/23/2022]
|
33
|
Müller WEG, Wang X, Schröder HC. New Target Sites for Treatment of Osteoporosis. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2017; 55:187-219. [PMID: 28238039 DOI: 10.1007/978-3-319-51284-6_6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the last few years, much progress has been achieved in the discovery of new drug target sites for treatment of osteoporotic disorders, one of the main challenging diseases with a large burden for the public health systems. Among these new agents promoting bone formation, shifting the impaired equilibrium between bone anabolism and bone catabolism in the direction of bone synthesis are inorganic polymers, in particular inorganic polyphosphates that show strong stimulatory effects on the expression of bone anabolic marker proteins and hydroxyapatite formation. The bone-forming activity of these polymers can even be enhanced by combination with certain small molecules like quercetin, or if given as functionally active particles with certain divalent cations like strontium ions even showing by itself biological activity. This chapter summarizes recent developments in the search and development of novel anti-osteoporotic agents, with a particular focus on therapeutic approaches based on the potential application of inorganic polymers and combinations.
Collapse
Affiliation(s)
- Werner E G Müller
- ERC Advanced Investigator Group, Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, 55128, Mainz, Germany. .,NanotecMARIN GmbH, Duesbergweg 6, 55128, Mainz, Germany.
| | - Xiaohong Wang
- ERC Advanced Investigator Group, Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, 55128, Mainz, Germany.,NanotecMARIN GmbH, Duesbergweg 6, 55128, Mainz, Germany
| | - Heinz C Schröder
- ERC Advanced Investigator Group, Institute for Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, Duesbergweg 6, 55128, Mainz, Germany.,NanotecMARIN GmbH, Duesbergweg 6, 55128, Mainz, Germany
| |
Collapse
|
34
|
PIXE study on the effects of parathyroid hormone on elemental content in rat bones. Phys Med 2016; 32:1615-1620. [PMID: 27899269 DOI: 10.1016/j.ejmp.2016.11.117] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 11/22/2016] [Accepted: 11/22/2016] [Indexed: 11/22/2022] Open
Abstract
Parathyroid hormone (PTH) has attracted considerable interest as a bone anabolic agent. PTH plays a central role in regulating calcium phosphate metabolism and its increases in production in response to low serum calcium levels. A continuous hypersecretion of PTH, as occurs in primary hyperparathyroidism, leads to bone resorption. In this study, the effect of different doses of parathyroid hormone (PTH) on bone mineral content (BMC) in rats was investigated by particle-induced X-ray emission (PIXE). This study will help in investigating further the toxicity of extremely high doses of PTH on BMC. For this study, PTH at doses of 15, 45, or 135μg/kg/day were applied to 9-month-old male and female Sprague Dawley (SD) rats. The concentrations of calcium (Ca), phosphorus (P), strontium (Sr), and zinc (Zn) were measured for bone treatment of PTH. From the results of the research, it was revealed that the biomechanical characteristics of the bone as well as the bone mass were enhanced after the treatment. It was further found that the concentrations of other elements also increased, excluding Zn. This research proved that PTH assists in the treatment of osteoporosis as revealed by the characteristics of different elements. PIXE can be used to determine the concentrations of bone mineral content.
Collapse
|
35
|
Abstract
Antiresorptive agents for osteoporosis are a cornerstone of therapy, but anabolic drugs have recently increased our options. By directly stimulating bone formation, anabolic agents reduce fracture incidence by improving bone qualities as well as increasing bone mass. The anabolic agent currently approved for osteoporosis, teriparatide (recombinant human parathyroid hormone[1–34]), has emerged as a major approach for selected patients with osteoporosis. Parathyroid hormone(1–84) is also available in Europe. Teriparatide increases bone density and bone turnover, improves microarchitecture and changes bone size. The incidence of vertebral and nonvertebral fractures is reduced. A current concept in the mechanism of teriparatide action is related to its effect of stimulating processes associated with bone formation before processes associated with bone resorption. This sequence of events has led to the concept of the anabolic window, the period of time when teriparatide is maximally anabolic. Newer approaches to the use of teriparatide alone and in combination with antiresorptive agents has led to ways in which the anabolic window can be expanded.
Collapse
Affiliation(s)
- John P Bilezikian
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, NY 10032 USA, Tel.: +1 212 305 6238, Fax: +1 212 305 6486,
| |
Collapse
|
36
|
Guo Q, Xu H, Yang X, Zhao D, Liu S, Sun X, Huang JA. Notch activation of Ca 2+-sensing receptor mediates hypoxia-induced pulmonary hypertension. Hypertens Res 2016; 40:117-129. [PMID: 27581537 DOI: 10.1038/hr.2016.118] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/15/2016] [Accepted: 07/25/2016] [Indexed: 12/13/2022]
Abstract
A recent study from our group demonstrated that the Ca2+-sensing receptor (CaSR) was upregulated and that the extracellular Ca2+-induced increase in the cytosolic Ca2+ concentration [Ca2+]cyt was enhanced in pulmonary arterial smooth muscle cells (PASMCs) from patients with idiopathic pulmonary arterial hypertension. Here, we examined whether hypoxia-induced activation of Notch signaling leads to the activation and upregulation of CaSR in hypoxia-induced pulmonary hypertension (HPH). The activation of Notch signaling with Jag-1, a Notch ligand, can activate the function and increase the expression of CaSR in acute and chronic hypoxic PASMCs. Downregulation of Notch3 with a siRNA attenuates the extracellular Ca2+-induced increase in [Ca2+]cyt and the increase in hypoxia-induced PASMC proliferation in acute hypoxic rat PASMCs. Furthermore, we tested the prevention and rescue effects of a γ-secretase inhibitor (DAPT) in HPH rats. For the Jag-1-treated group, right ventricular systolic pressure (RVSP), right heart hypertrophy (RV/LV+S ratio), and the level of right ventricular myocardial fibrosis were higher than the hypoxia alone group. Meanwhile, DAPT treatment prevented and rescued pulmonary hypertension in HPH rats. The Notch activation of CaSR mediates hypoxia-induced pulmonary hypertension. Understanding the new molecular mechanisms that regulate [Ca2+]cyt and PASMC proliferation is critical to elucidating the pathogenesis of HPH and the development of novel therapies for pulmonary hypertension.
Collapse
Affiliation(s)
- Qiang Guo
- Department of Medicine, Respiratory, Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hua Xu
- Department of Medicine, Respiratory, Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xinjing Yang
- Department of Medicine, Respiratory, Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Daguo Zhao
- Department of Medicine, Respiratory, Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shenlang Liu
- Department of Medicine, Respiratory, Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xue Sun
- Department of Medicine, Respiratory, Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jian-An Huang
- Department of Medicine, Respiratory, Emergency and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
37
|
Liang GB, Zhou C, Huo X, Wang H, Yang X, Huang S, Wang H, Wilkinson H, Luo L, Tang W, Sutton D, Li H, Zaller D, Meinke PT. Discovery of novel dihydrobenzofuran cyclopropane carboxylic acid based calcium sensing receptor antagonists for the treatment of osteoporosis. Bioorg Med Chem Lett 2016; 26:4077-80. [DOI: 10.1016/j.bmcl.2016.06.073] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/06/2016] [Accepted: 06/25/2016] [Indexed: 01/12/2023]
|
38
|
Martin TJ. Parathyroid Hormone-Related Protein, Its Regulation of Cartilage and Bone Development, and Role in Treating Bone Diseases. Physiol Rev 2016; 96:831-71. [DOI: 10.1152/physrev.00031.2015] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although parathyroid hormone-related protein (PTHrP) was discovered as a cancer-derived hormone, it has been revealed as an important paracrine/autocrine regulator in many tissues, where its effects are context dependent. Thus its location and action in the vasculature explained decades-long observations that injection of PTH into animals rapidly lowered blood pressure by producing vasodilatation. Its roles have been specified in development and maturity in cartilage and bone as a crucial regulator of endochondral bone formation and bone remodeling, respectively. Although it shares actions with parathyroid hormone (PTH) through the use of their common receptor, PTHR1, PTHrP has other actions mediated by regions within the molecule beyond the amino-terminal sequence that resembles PTH, including the ability to promote placental transfer of calcium from mother to fetus. A striking feature of the physiology of PTHrP is that it possesses structural features that equip it to be transported in and out of the nucleus, and makes use of a specific nuclear import mechanism to do so. Evidence from mouse genetic experiments shows that PTHrP generated locally in bone is essential for normal bone remodeling. Whereas the main physiological function of PTH is the hormonal regulation of calcium metabolism, locally generated PTHrP is the important physiological mediator of bone remodeling postnatally. Thus the use of intermittent injection of PTH as an anabolic therapy for bone appears to be a pharmacological application of the physiological function of PTHrP. There is much current interest in the possibility of developing PTHrP analogs that might enhance the therapeutic anabolic effects.
Collapse
Affiliation(s)
- T. John Martin
- St Vincent's Institute of Medical Research, Department of Medicine, University of Melbourne, St Vincent's Hospital, Melbourne, Australia
| |
Collapse
|
39
|
Liu L, Wang C, Lin Y, Xi Y, Li H, Shi S, Li H, Zhang W, Zhao Y, Tian Y, Xu C, Wang L. Suppression of calcium‑sensing receptor ameliorates cardiac hypertrophy through inhibition of autophagy. Mol Med Rep 2016; 14:111-20. [PMID: 27176663 PMCID: PMC4918534 DOI: 10.3892/mmr.2016.5279] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 03/22/2016] [Indexed: 01/16/2023] Open
Abstract
The calcium-sensing receptor (CaSR) releases intracellular calcium ([Ca2+]i) by accumulating inositol phosphate. Changes in [Ca2+]i initiate myocardial hypertrophy. Furthermore, autophagy associated with [Ca2+]i. Autophagy has previously been demonstrated to participate in the hypertrophic process. The current study investigated whether suppression of CaSR affects the hypertrophic response via modulating autophagy. Isoproterenol (ISO) was used to induce cardiac hypertrophy in Wistar rats. Hypertrophic status was determined by echocardiographic assessment, hematoxylin and eosin, and Masson's staining. The protein expression levels of CaSR and autophagy level were observed. Changes of hypertrophy and autophagy indicators were observed following intravenous injection of a CaSR inhibitor. An ISO-induced cardiomyocyte hypertrophy model was established and used determine the involvement of GdCl3. [Ca2+]i was determined using Fluo-4/AM dye followed by confocal microscopy. The expression levels of various active proteins were analyzed by western blotting. The size of the heart, expression levels of CaSR and autophagy level were markedly increased in hypertrophic myocardium. In addition, the present study demonstrated that the indicators of hypertrophy and autophagy were effectively suppressed by CaSR inhibitor. Furthermore, similar effects were demonstrated in neonatal rat hypertrophic cardiomyocytes treated with ISO. It was also observed that CaSR regulates the Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ)-AMP-activated protein kinase (AMPK)-mammalian target of rapamycin (mTOR) signaling pathway induced by ISO in cardiomyocytes. Furthermore, the AMPK inhibition significantly reduced the autophagy level following CaSR stimulation (P<0.05). The results of the present demonstrated that inhibition of CaSR may ameliorate cardiac hypertrophy induced by ISO and the effect may be associated with the inhibition of autophagy and suppression of the CaMKKβ-AMPK-mTOR signaling pathway.
Collapse
Affiliation(s)
- Lei Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Chao Wang
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yan Lin
- Department of Pathophysiology, Qiqihar Medical College, Qiqihar, Heilongjiang 161042, P.R. China
| | - Yuhui Xi
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Hong Li
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Sa Shi
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Hongzhu Li
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yajun Zhao
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Ye Tian
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Changqing Xu
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Lina Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| |
Collapse
|
40
|
Mayr B, Schnabel D, Dörr HG, Schöfl C. GENETICS IN ENDOCRINOLOGY: Gain and loss of function mutations of the calcium-sensing receptor and associated proteins: current treatment concepts. Eur J Endocrinol 2016; 174:R189-208. [PMID: 26646938 DOI: 10.1530/eje-15-1028] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 12/08/2015] [Indexed: 12/26/2022]
Abstract
The calcium-sensing receptor (CASR) is the main calcium sensor in the maintenance of calcium metabolism. Mutations of the CASR, the G protein alpha 11 (GNA11) and the adaptor-related protein complex 2 sigma 1 subunit (AP2S1) genes can shift the set point for calcium sensing causing hyper- or hypo-calcemic disorders. Therapeutic concepts for these rare diseases range from general therapies of hyper- and hypo-calcemic conditions to more pathophysiology oriented approaches such as parathyroid hormone (PTH) substitution and allosteric CASR modulators. Cinacalcet is a calcimimetic that enhances receptor function and has gained approval for the treatment of hyperparathyroidism. Calcilytics in turn attenuate CASR activity and are currently under investigation for the treatment of various diseases. We conducted a literature search for reports about treatment of patients harboring inactivating or activating CASR, GNA11 or AP2S1 mutants and about in vitro effects of allosteric CASR modulators on mutated CASR. The therapeutic concepts for patients with familial hypocalciuric hypercalcemia (FHH), neonatal hyperparathyroidism (NHPT), neonatal severe hyperparathyroidism (NSHPT) and autosomal dominant hypocalcemia (ADH) are reviewed. FHH is usually benign, but symptomatic patients benefit from cinacalcet. In NSHPT patients pamidronate effectively lowers serum calcium, but most patients require parathyroidectomy. In some patients cinacalcet can obviate the need for surgery, particularly in heterozygous NHPT. Symptomatic ADH patients respond to vitamin D and calcium supplementation but this may increase calciuria and renal complications. PTH treatment can reduce relative hypercalciuria. None of the currently available therapies for ADH, however, prevent tissue calcifications and complications, which may become possible with calcilytics that correct the underlying pathophysiologic defect.
Collapse
Affiliation(s)
- Bernhard Mayr
- Division of Endocrinology and DiabetesDepartment of Medicine I, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Ulmenweg 18, 91054 Erlangen, GermanyCenter for Chronic Sick ChildrenPediatric Endocrinology and Diabetes, Charité University Medicine Berlin, Berlin, GermanyDivision of Paediatric Endocrinology and DiabetesDepartment of Paediatrics, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Dirk Schnabel
- Division of Endocrinology and DiabetesDepartment of Medicine I, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Ulmenweg 18, 91054 Erlangen, GermanyCenter for Chronic Sick ChildrenPediatric Endocrinology and Diabetes, Charité University Medicine Berlin, Berlin, GermanyDivision of Paediatric Endocrinology and DiabetesDepartment of Paediatrics, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Helmuth-Günther Dörr
- Division of Endocrinology and DiabetesDepartment of Medicine I, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Ulmenweg 18, 91054 Erlangen, GermanyCenter for Chronic Sick ChildrenPediatric Endocrinology and Diabetes, Charité University Medicine Berlin, Berlin, GermanyDivision of Paediatric Endocrinology and DiabetesDepartment of Paediatrics, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Christof Schöfl
- Division of Endocrinology and DiabetesDepartment of Medicine I, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Ulmenweg 18, 91054 Erlangen, GermanyCenter for Chronic Sick ChildrenPediatric Endocrinology and Diabetes, Charité University Medicine Berlin, Berlin, GermanyDivision of Paediatric Endocrinology and DiabetesDepartment of Paediatrics, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
41
|
Nemeth EF, Goodman WG. Calcimimetic and Calcilytic Drugs: Feats, Flops, and Futures. Calcif Tissue Int 2016; 98:341-58. [PMID: 26319799 DOI: 10.1007/s00223-015-0052-z] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/10/2015] [Indexed: 01/28/2023]
Abstract
The actions of extracellular Ca(2+) in regulating parathyroid gland and kidney functions are mediated by the extracellular calcium receptor (CaR), a G protein-coupled receptor. The CaR is one of the essential molecules maintaining systemic Ca(2+) homeostasis and is a molecular target for drugs useful in treating bone and mineral disorders. Ligands that activate the CaR are termed calcimimetics and are classified as either agonists (type I) or positive allosteric modulators (type II); calcimimetics inhibit the secretion of parathyroid hormone (PTH). Cinacalcet is a type II calcimimetic that is used to treat secondary hyperparathyroidism in patients receiving dialysis and to treat hypercalcemia in some forms of primary hyperparathyroidism. The use of cinacalcet among patients with secondary hyperparathyroidism who are managed with dialysis effectively lowers circulating PTH levels, reduces serum phosphorus and FGF23 concentrations, improves bone histopathology, and may diminish skeletal fracture rates and the need for parathyroidectomy. A second generation type II calcimimetic (AMG 416) is currently under regulatory review. Calcilytics are CaR antagonists that stimulate the secretion of PTH. Several calcilytic compounds have been evaluated as orally active anabolic therapies for postmenopausal osteoporosis but clinical development of all of them has been abandoned because they lacked clinical efficacy. Calcilytics might be repurposed for new indications like autosomal dominant hypocalcemia or other disorders beyond those involving systemic Ca(2+) homeostasis.
Collapse
Affiliation(s)
- E F Nemeth
- MetisMedica, 13 Poplar Plains Road, Toronto, ON, M4V 2M7, Canada.
| | - W G Goodman
- , 22102 Palais Place, Calabasas, CA, 91302, USA
| |
Collapse
|
42
|
Leach K, Gregory KJ, Kufareva I, Khajehali E, Cook AE, Abagyan R, Conigrave AD, Sexton PM, Christopoulos A. Towards a structural understanding of allosteric drugs at the human calcium-sensing receptor. Cell Res 2016; 26:574-92. [PMID: 27002221 DOI: 10.1038/cr.2016.36] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 12/18/2015] [Accepted: 01/28/2016] [Indexed: 12/19/2022] Open
Abstract
Drugs that allosterically target the human calcium-sensing receptor (CaSR) have substantial therapeutic potential, but are currently limited. Given the absence of high-resolution structures of the CaSR, we combined mutagenesis with a novel analytical approach and molecular modeling to develop an "enriched" picture of structure-function requirements for interaction between Ca(2+)o and allosteric modulators within the CaSR's 7 transmembrane (7TM) domain. An extended cavity that accommodates multiple binding sites for structurally diverse ligands was identified. Phenylalkylamines bind to a site that overlaps with a putative Ca(2+)o-binding site and extends towards an extracellular vestibule. In contrast, the structurally and pharmacologically distinct AC-265347 binds deeper within the 7TM domains. Furthermore, distinct amino acid networks were found to mediate cooperativity by different modulators. These findings may facilitate the rational design of allosteric modulators with distinct and potentially pathway-biased pharmacological effects.
Collapse
Affiliation(s)
- Katie Leach
- Drug Discovery Biology, Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Karen J Gregory
- Drug Discovery Biology, Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92037, USA
| | - Elham Khajehali
- Drug Discovery Biology, Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Anna E Cook
- Drug Discovery Biology, Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Ruben Abagyan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92037, USA
| | - Arthur D Conigrave
- School of Molecular Bioscience, Charles Perkins Centre, University of Sydney, NSW 2006, Australia
| | - Patrick M Sexton
- Drug Discovery Biology, Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Arthur Christopoulos
- Drug Discovery Biology, Department of Pharmacology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, VIC 3052, Australia
| |
Collapse
|
43
|
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: 3.5] [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
|
44
|
Santa Maria C, Cheng Z, Li A, Wang J, Shoback D, Tu CL, Chang W. Interplay between CaSR and PTH1R signaling in skeletal development and osteoanabolism. Semin Cell Dev Biol 2016; 49:11-23. [PMID: 26688334 PMCID: PMC4761456 DOI: 10.1016/j.semcdb.2015.12.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 12/05/2015] [Indexed: 12/01/2022]
Abstract
Parathyroid hormone (PTH)-related peptide (PTHrP) controls the pace of pre- and post-natal growth plate development by activating the PTH1R in chondrocytes, while PTH maintains mineral and skeletal homeostasis by modulating calciotropic activities in kidneys, gut, and bone. The extracellular calcium-sensing receptor (CaSR) is a member of family C, G protein-coupled receptor, which regulates mineral and skeletal homeostasis by controlling PTH secretion in parathyroid glands and Ca(2+) excretion in kidneys. Recent studies showed the expression of CaSR in chondrocytes, osteoblasts, and osteoclasts and confirmed its non-redundant roles in modulating the recruitment, proliferation, survival, and differentiation of the cells. This review emphasizes the actions of CaSR and PTH1R signaling responses in cartilage and bone and discusses how these two signaling cascades interact to control growth plate development and maintain skeletal metabolism in physiological and pathological conditions. Lastly, novel therapeutic regimens that exploit interrelationship between the CaSR and PTH1R are proposed to produce more robust osteoanabolism.
Collapse
Affiliation(s)
- Christian Santa Maria
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Zhiqiang Cheng
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Alfred Li
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Jiali Wang
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Dolores Shoback
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Chia-Ling Tu
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Wenhan Chang
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA.
| |
Collapse
|
45
|
Hannan FM, Walls GV, Babinsky VN, Nesbit MA, Kallay E, Hough TA, Fraser WD, Cox RD, Hu J, Spiegel AM, Thakker RV. The Calcilytic Agent NPS 2143 Rectifies Hypocalcemia in a Mouse Model With an Activating Calcium-Sensing Receptor (CaSR) Mutation: Relevance to Autosomal Dominant Hypocalcemia Type 1 (ADH1). Endocrinology 2015; 156:3114-21. [PMID: 26052899 PMCID: PMC4541614 DOI: 10.1210/en.2015-1269] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Autosomal dominant hypocalcemia type 1 (ADH1) is caused by germline gain-of-function mutations of the calcium-sensing receptor (CaSR) and may lead to symptomatic hypocalcemia, inappropriately low serum PTH concentrations and hypercalciuria. Negative allosteric CaSR modulators, known as calcilytics, have been shown to normalize the gain-of-function associated with ADH-causing CaSR mutations in vitro and represent a potential targeted therapy for ADH1. However, the effectiveness of calcilytic drugs for the treatment of ADH1-associated hypocalcemia remains to be established. We have investigated NPS 2143, a calcilytic compound, for the treatment of ADH1 by in vitro and in vivo studies involving a mouse model, known as Nuf, which harbors a gain-of-function CaSR mutation, Leu723Gln. Wild-type (Leu723) and Nuf mutant (Gln723) CaSRs were expressed in HEK293 cells, and the effect of NPS 2143 on their intracellular calcium responses was determined by flow cytometry. NPS 2143 was also administered as a single ip bolus to wild-type and Nuf mice and plasma concentrations of calcium and PTH, and urinary calcium excretion measured. In vitro administration of NPS 2143 decreased the intracellular calcium responses of HEK293 cells expressing the mutant Gln723 CaSR in a dose-dependent manner, thereby rectifying the gain-of-function associated with the Nuf mouse CaSR mutation. Intraperitoneal injection of NPS 2143 in Nuf mice led to significant increases in plasma calcium and PTH without elevating urinary calcium excretion. These studies of a mouse model with an activating CaSR mutation demonstrate NPS 2143 to normalize the gain-of-function causing ADH1 and improve the hypocalcemia associated with this disorder.
Collapse
Affiliation(s)
- Fadil M Hannan
- Academic Endocrine Unit (F.M.H., G.V.W., V.N.B., M.A.N., E.K., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom; Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre (T.A.H., R.D.C.), MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, United Kingdom; Department of Medicine (W.D.F.), Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, United Kingdom; Laboratory of Bioorganic Chemistry (J.H.), National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892; and Albert Einstein College of Medicine (A.M.S.), Bronx, New York 10461
| | - Gerard V Walls
- Academic Endocrine Unit (F.M.H., G.V.W., V.N.B., M.A.N., E.K., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom; Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre (T.A.H., R.D.C.), MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, United Kingdom; Department of Medicine (W.D.F.), Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, United Kingdom; Laboratory of Bioorganic Chemistry (J.H.), National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892; and Albert Einstein College of Medicine (A.M.S.), Bronx, New York 10461
| | - Valerie N Babinsky
- Academic Endocrine Unit (F.M.H., G.V.W., V.N.B., M.A.N., E.K., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom; Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre (T.A.H., R.D.C.), MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, United Kingdom; Department of Medicine (W.D.F.), Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, United Kingdom; Laboratory of Bioorganic Chemistry (J.H.), National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892; and Albert Einstein College of Medicine (A.M.S.), Bronx, New York 10461
| | - M Andrew Nesbit
- Academic Endocrine Unit (F.M.H., G.V.W., V.N.B., M.A.N., E.K., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom; Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre (T.A.H., R.D.C.), MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, United Kingdom; Department of Medicine (W.D.F.), Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, United Kingdom; Laboratory of Bioorganic Chemistry (J.H.), National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892; and Albert Einstein College of Medicine (A.M.S.), Bronx, New York 10461
| | - Enikö Kallay
- Academic Endocrine Unit (F.M.H., G.V.W., V.N.B., M.A.N., E.K., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom; Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre (T.A.H., R.D.C.), MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, United Kingdom; Department of Medicine (W.D.F.), Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, United Kingdom; Laboratory of Bioorganic Chemistry (J.H.), National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892; and Albert Einstein College of Medicine (A.M.S.), Bronx, New York 10461
| | - Tertius A Hough
- Academic Endocrine Unit (F.M.H., G.V.W., V.N.B., M.A.N., E.K., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom; Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre (T.A.H., R.D.C.), MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, United Kingdom; Department of Medicine (W.D.F.), Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, United Kingdom; Laboratory of Bioorganic Chemistry (J.H.), National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892; and Albert Einstein College of Medicine (A.M.S.), Bronx, New York 10461
| | - William D Fraser
- Academic Endocrine Unit (F.M.H., G.V.W., V.N.B., M.A.N., E.K., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom; Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre (T.A.H., R.D.C.), MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, United Kingdom; Department of Medicine (W.D.F.), Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, United Kingdom; Laboratory of Bioorganic Chemistry (J.H.), National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892; and Albert Einstein College of Medicine (A.M.S.), Bronx, New York 10461
| | - Roger D Cox
- Academic Endocrine Unit (F.M.H., G.V.W., V.N.B., M.A.N., E.K., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom; Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre (T.A.H., R.D.C.), MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, United Kingdom; Department of Medicine (W.D.F.), Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, United Kingdom; Laboratory of Bioorganic Chemistry (J.H.), National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892; and Albert Einstein College of Medicine (A.M.S.), Bronx, New York 10461
| | - Jianxin Hu
- Academic Endocrine Unit (F.M.H., G.V.W., V.N.B., M.A.N., E.K., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom; Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre (T.A.H., R.D.C.), MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, United Kingdom; Department of Medicine (W.D.F.), Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, United Kingdom; Laboratory of Bioorganic Chemistry (J.H.), National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892; and Albert Einstein College of Medicine (A.M.S.), Bronx, New York 10461
| | - Allen M Spiegel
- Academic Endocrine Unit (F.M.H., G.V.W., V.N.B., M.A.N., E.K., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom; Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre (T.A.H., R.D.C.), MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, United Kingdom; Department of Medicine (W.D.F.), Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, United Kingdom; Laboratory of Bioorganic Chemistry (J.H.), National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892; and Albert Einstein College of Medicine (A.M.S.), Bronx, New York 10461
| | - Rajesh V Thakker
- Academic Endocrine Unit (F.M.H., G.V.W., V.N.B., M.A.N., E.K., R.V.T.), Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, OX3 7LJ, United Kingdom; Medical Research Council (MRC) Mammalian Genetics Unit and Mary Lyon Centre (T.A.H., R.D.C.), MRC Harwell, Harwell Science and Innovation Campus, Oxfordshire, OX11 0RD, United Kingdom; Department of Medicine (W.D.F.), Norwich Medical School, University of East Anglia, Norwich, NR4 7TJ, United Kingdom; Laboratory of Bioorganic Chemistry (J.H.), National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892; and Albert Einstein College of Medicine (A.M.S.), Bronx, New York 10461
| |
Collapse
|
46
|
Massy ZA, Hénaut L, Larsson TE, Vervloet MG. Calcium-sensing receptor activation in chronic kidney disease: effects beyond parathyroid hormone control. Semin Nephrol 2015; 34:648-59. [PMID: 25498383 DOI: 10.1016/j.semnephrol.2014.10.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Secondary hyperparathyroidism (SHPT) is an important complication of advanced chronic kidney disease (CKD). Cinacalcet, an allosteric modulator of the calcium-sensing receptor (CaSR) expressed in parathyroid glands, is the only calcimimetic approved to treat SHPT in patients on dialysis. By enhancing CaSR sensitivity for plasma extracellular calcium (Ca(2+)0), cinacalcet reduces serum parathyroid hormone, Ca(2+)0, and serum inorganic phosphorous concentrations, allowing better control of SHPT and CKD-mineral and bone disorders. Of interest, the CaSR also is expressed in a variety of tissues where its activation regulates diverse cellular processes, including secretion, apoptosis, and proliferation. Thus, the existence of potential off-target effects of cinacalcet cannot be neglected. This review summarizes our current knowledge concerning the potential role(s) of the CaSR expressed in various tissues in CKD-related disorders, independently of parathyroid hormone control.
Collapse
Affiliation(s)
- Ziad A Massy
- Inserm U-1088, University of Picardie Jules Verne, Amiens, France; Division of Nephrology, Ambroise Paré Hospital, Paris-Ile-de-France-Ouest University (University of Versailles Saint-Quentin-En-Yvelines), Paris-Boulogne Billancourt, France.
| | - Lucie Hénaut
- Inserm U-1088, University of Picardie Jules Verne, Amiens, France
| | - Tobias E Larsson
- Department of Clinical Science, Intervention and Technology, Renal Unit, Karolinska Institutet, Stockholm, Sweden; Department of Nephrology, Karolinska University Hospital, Stockholm, Sweden
| | - Marc G Vervloet
- Department of Nephrology and Institute of Cardiovascular Research VU (Institute for Cardiovascular Research of the Vrije Universiteit of Amsterdam), VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
47
|
Brandenburg VM, Floege J. Adynamic bone disease-bone and beyond. NDT Plus 2015; 1:135-47. [PMID: 25983860 PMCID: PMC4421169 DOI: 10.1093/ndtplus/sfn040] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2008] [Accepted: 03/18/2008] [Indexed: 11/13/2022] Open
Affiliation(s)
- Vincent M Brandenburg
- Department of Nephrology and Clinical Immunology , RWTH University Hospital Aachen , Pauwelsstrasse 30, Aachen, D-52057 , Germany
| | - Jürgen Floege
- Department of Nephrology and Clinical Immunology , RWTH University Hospital Aachen , Pauwelsstrasse 30, Aachen, D-52057 , Germany
| |
Collapse
|
48
|
Abstract
The extracellular calcium-sensing receptor, CaSR, is a member of the G protein-coupled receptor superfamily and has a critical role in modulating Ca(2+) homeostasis via its role in the parathyroid glands and kidneys. New evidence suggests that CaSR expression in cartilage and bone also directly regulates skeletal homeostasis. This Review discusses the role of CaSR in chondrocytes, through which CaSR contributes to the development of the cartilaginous growth plate, as well as in osteoblasts and osteoclasts, through which CaSR has effects on skeletal development and bone turnover in young and mature animals. The interaction of skeletal CaSR activation with parathyroid hormone (PTH), which is secreted by the parathyroid gland, can lead to net bone formation in trabecular bone or net bone resorption in cortical bone. Allosteric modulators of CaSR are beneficial in some clinical conditions, with effects that are mediated by the ability of these agents to alter levels of PTH and improve Ca(2+) homeostasis. However, further insights into the action of CaSR in bone cells might lead to CaSR-based drugs that maximize not only the effects of the receptor on the parathyroid glands and kidneys but also on bone.
Collapse
Affiliation(s)
- David Goltzman
- Department of Medicine, McGill University, 687 Pine Avenue West, Montreal, QC H3A 1A1, Canada
| | - Geoffrey N Hendy
- Department of Medicine, McGill University, 687 Pine Avenue West, Montreal, QC H3A 1A1, Canada
| |
Collapse
|
49
|
Hernández-Bedolla MA, Carretero-Ortega J, Valadez-Sánchez M, Vázquez-Prado J, Reyes-Cruz G. Chemotactic and proangiogenic role of calcium sensing receptor is linked to secretion of multiple cytokines and growth factors in breast cancer MDA-MB-231 cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:166-82. [DOI: 10.1016/j.bbamcr.2014.10.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/11/2014] [Accepted: 10/15/2014] [Indexed: 12/18/2022]
|
50
|
Letz S, Haag C, Schulze E, Frank-Raue K, Raue F, Hofner B, Mayr B, Schöfl C. Amino alcohol- (NPS-2143) and quinazolinone-derived calcilytics (ATF936 and AXT914) differentially mitigate excessive signalling of calcium-sensing receptor mutants causing Bartter syndrome Type 5 and autosomal dominant hypocalcemia. PLoS One 2014; 9:e115178. [PMID: 25506941 PMCID: PMC4266668 DOI: 10.1371/journal.pone.0115178] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 11/19/2014] [Indexed: 11/24/2022] Open
Abstract
INTRODUCTION Activating calcium sensing receptor (CaSR) mutations cause autosomal dominant hypocalcemia (ADH) characterized by low serum calcium, inappropriately low PTH and relative hypercalciuria. Four activating CaSR mutations cause additional renal wasting of sodium, chloride and other salts, a condition called Bartter syndrome (BS) type 5. Until today there is no specific medical treatment for BS type 5 and ADH. We investigated the effects of different allosteric CaSR antagonists (calcilytics) on activating CaSR mutants. METHODS All 4 known mutations causing BS type 5 and five ADH mutations were expressed in HEK 293T cells and receptor signalling was studied by measurement of intracellular free calcium in response to extracellular calcium ([Ca2+]o). To investigate the effect of calcilytics, cells were stimulated with 3 mM [Ca2+]o in the presence or absence of NPS-2143, ATF936 or AXT914. RESULTS All BS type 5 and ADH mutants showed enhanced signalling activity to [Ca2+]o with left shifted dose response curves. In contrast to the amino alcohol NPS-2143, which was only partially effective, the quinazolinone calcilytics ATF936 and AXT914 significantly mitigated excessive cytosolic calcium signalling of all BS type 5 and ADH mutants studied. When these mutants were co-expressed with wild-type CaSR to approximate heterozygosity in patients, ATF936 and AXT914 were also effective on all mutants. CONCLUSION The calcilytics ATF936 and AXT914 are capable of attenuating enhanced cytosolic calcium signalling activity of CaSR mutations causing BS type 5 and ADH. Quinazolinone calcilytics might therefore offer a novel treatment option for patients with activating CaSR mutations.
Collapse
Affiliation(s)
- Saskia Letz
- Division of Endocrinology and Diabetes, Department of Medicine I, Universitätsklinikum Erlangen, Erlangen, Germany
| | | | | | | | | | - Benjamin Hofner
- Department of Medical Informatics, Biometry and Epidemiology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Bernhard Mayr
- Division of Endocrinology and Diabetes, Department of Medicine I, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Christof Schöfl
- Division of Endocrinology and Diabetes, Department of Medicine I, Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|