1
|
Ranieri M, Angelini I, D'Agostino M, Di Mise A, Centrone M, Venneri M, Ferrulli A, Mastrodonato M, Tamma G, Endo I, Fukumoto S, Matsumoto T, Valenti G. In vivo treatment with calcilytic of CaSR knock-in mice ameliorates renal phenotype reversing downregulation of the vasopressin-AQP2 pathway. J Physiol 2024; 602:3207-3224. [PMID: 38367250 DOI: 10.1113/jp284233] [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: 10/17/2023] [Accepted: 01/30/2024] [Indexed: 02/19/2024] Open
Abstract
High concentrations of urinary calcium counteract vasopressin action via the activation of the Calcium-Sensing Receptor (CaSR) expressed in the luminal membrane of the collecting duct cells, which impairs the trafficking of aquaporin-2 (AQP2). In line with these findings, we provide evidence that, with respect to wild-type mice, CaSR knock-in (KI) mice mimicking autosomal dominant hypocalcaemia, display a significant decrease in the total content of AQP2 associated with significantly higher levels of AQP2 phosphorylation at Ser261, a phosphorylation site involved in AQP2 degradation. Interestingly, KI mice also had significantly higher levels of phosphorylated p38MAPK, a downstream effector of CaSR and known to phosphorylate AQP2 at Ser261. Moreover, ATF1 phosphorylated at Ser63, a transcription factor downstream of p38MAPK, was significantly higher in KI. In addition, KI mice had significantly higher levels of AQP2-targeting miRNA137 consistent with a post-transcriptional downregulation of AQP2. In vivo treatment of KI mice with the calcilytic JTT-305, a CaSR antagonist, increased AQP2 expression and reduced AQP2-targeting miRNA137 levels in KI mice. Together, these results provide direct evidence for a critical role of CaSR in impairing both short-term vasopressin response by increasing AQP2-pS261, as well as AQP2 abundance, via the p38MAPK-ATF1-miR137 pathway. KEY POINTS: Calcium-Sensing Receptor (CaSR) activating mutations are the main cause of autosomal dominant hypocalcaemia (ADH) characterized by inappropriate renal calcium excretion leading to hypocalcaemia and hypercalciuria. Current treatments of ADH patients with parathyroid hormone, although improving hypocalcaemia, do not improve hypercalciuria or nephrocalcinosis. In vivo treatment with calcilytic JTT-305/MK-5442 ameliorates most of the ADH phenotypes of the CaSR knock-in mice including hypercalciuria or nephrocalcinosis and reverses the downregulation of the vasopressin-sensitive aquaporin-2 (AQP2) expression, providing direct evidence for a critical role of CaSR in impairing vasopressin response. The beneficial effect of calcilytic in reducing the risk of renal calcification may occur in a parathyroid hormone-independent action through vasopressin-dependent inhibition of cAMP synthesis in the thick ascending limb and in the collecting duct. The amelioration of most of the abnormalities in calcium metabolism including hypercalciuria, renal calcification, and AQP2-mediated osmotic water reabsorption makes calcilytic a good candidate as a novel therapeutic agent for ADH.
Collapse
Affiliation(s)
- Marianna Ranieri
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Italy
| | - Ines Angelini
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Italy
| | | | - Annarita Di Mise
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Italy
| | - Mariangela Centrone
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Italy
| | - Maria Venneri
- Istituti Clinici Scientifici Maugeri SPA SB IRCCS, Bari, Italy
| | - Angela Ferrulli
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Italy
| | - Maria Mastrodonato
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Italy
| | - Grazia Tamma
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Italy
| | - Itsuro Endo
- Department of Bioregulatory Sciences, Tokushima University, Tokushima, Japan
| | - Seiji Fukumoto
- Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Toshio Matsumoto
- Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Giovanna Valenti
- Department of Biosciences, Biotechnologies and Environment, University of Bari, Italy
| |
Collapse
|
2
|
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
|
3
|
Kalinkovich A, Livshits G. Biased and allosteric modulation of bone cell-expressing G protein-coupled receptors as a novel approach to osteoporosis therapy. Pharmacol Res 2021; 171:105794. [PMID: 34329703 DOI: 10.1016/j.phrs.2021.105794] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/20/2021] [Accepted: 07/25/2021] [Indexed: 12/16/2022]
Abstract
On the cellular level, osteoporosis (OP) is a result of imbalanced bone remodeling, in which osteoclastic bone resorption outcompetes osteoblastic bone formation. Currently available OP medications include both antiresorptive and bone-forming drugs. However, their long-term use in OP patients, mainly in postmenopausal women, is accompanied by severe side effects. Notably, the fundamental coupling between bone resorption and formation processes underlies the existence of an undesirable secondary outcome that bone anabolic or anti-resorptive drugs also reduce bone formation. This drawback requires the development of anti-OP drugs capable of selectively stimulating osteoblastogenesis and concomitantly reducing osteoclastogenesis. We propose that the application of small synthetic biased and allosteric modulators of bone cell receptors, which belong to the G-protein coupled receptors (GPCR) family, could be the key to resolving the undesired anti-OP drug selectivity. This approach is based on the capacity of these GPCR modulators, unlike the natural ligands, to trigger signaling pathways that promote beneficial effects on bone remodeling while blocking potentially deleterious effects. Under the settings of OP, an optimal anti-OP drug should provide fine-tuned regulation of downstream effects, for example, intermittent cyclic AMP (cAMP) elevation, preservation of Ca2+ balance, stimulation of osteoprotegerin (OPG) and estrogen production, suppression of sclerostin secretion, and/or preserved/enhanced canonical β-catenin/Wnt signaling pathway. As such, selective modulation of GPCRs involved in bone remodeling presents a promising approach in OP treatment. This review focuses on the evidence for the validity of our hypothesis.
Collapse
Affiliation(s)
- Alexander Kalinkovich
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel
| | - Gregory Livshits
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6905126, Israel; Adelson School of Medicine, Ariel University, Ariel 4077625, Israel.
| |
Collapse
|
4
|
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
|
5
|
Yarova PL, Huang P, Schepelmann MW, Bruce R, Ecker R, Nica R, Telezhkin V, Traini D, Gomes Dos Reis L, Kidd EJ, Ford WR, Broadley KJ, Kariuki BM, Corrigan CJ, Ward JPT, Kemp PJ, Riccardi D. Characterization of Negative Allosteric Modulators of the Calcium-Sensing Receptor for Repurposing as a Treatment of Asthma. J Pharmacol Exp Ther 2020; 376:51-63. [PMID: 33115824 DOI: 10.1124/jpet.120.000281] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/05/2020] [Indexed: 12/16/2022] Open
Abstract
Asthma is still an incurable disease, and there is a recognized need for novel small-molecule therapies for people with asthma, especially those poorly controlled by current treatments. We previously demonstrated that calcium-sensing receptor (CaSR) negative allosteric modulators (NAMs), calcilytics, uniquely suppress both airway hyperresponsiveness (AHR) and inflammation in human cells and murine asthma surrogates. Here we assess the feasibility of repurposing four CaSR NAMs, which were originally developed for oral therapy for osteoporosis and previously tested in the clinic as a novel, single, and comprehensive topical antiasthma therapy. We address the hypotheses, using murine asthma surrogates, that topically delivered CaSR NAMs 1) abolish AHR; 2) are unlikely to cause unwanted systemic effects; 3) are suitable for topical application; and 4) inhibit airway inflammation to the same degree as the current standard of care, inhaled corticosteroids, and, furthermore, inhibit airway remodeling. All four CaSR NAMs inhibited poly-L-arginine-induced AHR in naïve mice and suppressed both AHR and airway inflammation in a murine surrogate of acute asthma, confirming class specificity. Repeated exposure to inhaled CaSR NAMs did not alter blood pressure, heart rate, or serum calcium concentrations. Optimal candidates for repurposing were identified based on anti-AHR/inflammatory activities, pharmacokinetics/pharmacodynamics, formulation, and micronization studies. Whereas both inhaled CaSR NAMs and inhaled corticosteroids reduced airways inflammation, only the former prevented goblet cell hyperplasia in a chronic asthma model. We conclude that inhaled CaSR NAMs are likely a single, safe, and effective topical therapy for human asthma, abolishing AHR, suppressing airways inflammation, and abrogating some features of airway remodeling. SIGNIFICANCE STATEMENT: Calcium-sensing receptor (CaSR) negative allosteric modulators (NAMs) reduce airway smooth muscle hyperresponsiveness, reverse airway inflammation as efficiently as topical corticosteroids, and suppress airway remodeling in asthma surrogates. CaSR NAMs, which were initially developed for oral therapy of osteoporosis proved inefficacious for this indication despite being safe and well tolerated. Here we show that structurally unrelated CaSR NAMs are suitable for inhaled delivery and represent a one-stop, steroid-free approach to asthma control and prophylaxis.
Collapse
Affiliation(s)
- Polina L Yarova
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Ping Huang
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Martin W Schepelmann
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Richard Bruce
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Rupert Ecker
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Robert Nica
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Vsevolod Telezhkin
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Daniela Traini
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Larissa Gomes Dos Reis
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Emma J Kidd
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - William R Ford
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Kenneth J Broadley
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Benson M Kariuki
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Christopher J Corrigan
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Jeremy P T Ward
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Paul J Kemp
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| | - Daniela Riccardi
- Schools of Biosciences (P.L.Y., P.H., M.W.S., R.B., P.J.K., D.R.), Pharmacy (E.J.K., W.R.F., K.J.B.), and Chemistry (B.M.K.), Cardiff University, Cardiff, United Kingdom; Institute for Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (M.W.S.); TissueGnostics GmbH, Vienna, Austria (R.E., R.N.); School of Dental Sciences, University of Newcastle, United Kingdom (V.T.); Woolcock Institute of Medical Research, The University of Sydney, Sydney, Australia (D.T., L.G.d.R.); and School of Immunology & Microbial Sciences, King's College London, London, United Kingdom (C.J.C., J.P.T.W.)
| |
Collapse
|
6
|
Wang M, Xia F, Wei Y, Wei X. Molecular mechanisms and clinical management of cancer bone metastasis. Bone Res 2020; 8:30. [PMID: 32793401 PMCID: PMC7391760 DOI: 10.1038/s41413-020-00105-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/03/2019] [Accepted: 10/23/2019] [Indexed: 02/05/2023] Open
Abstract
As one of the most common metastatic sites of malignancies, bone has a unique microenvironment that allows metastatic tumor cells to grow and flourish. The fenestrated capillaries in the bone, bone matrix, and bone cells, including osteoblasts and osteoclasts, together maintain the homeostasis of the bone microenvironment. In contrast, tumor-derived factors act on bone components, leading to subsequent bone resorption or excessive bone formation. The various pathways involved also provide multiple targets for therapeutic strategies against bone metastases. In this review, we summarize the current understanding of the mechanism of bone metastases. Based on the general process of bone metastases, we specifically highlight the complex crosstalk between tumor cells and the bone microenvironment and the current management of cancer bone metastases.
Collapse
Affiliation(s)
- Manni Wang
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| | - Fan Xia
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan P.R. China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Targets, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041 Sichuan P.R. China
| |
Collapse
|
7
|
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
|
8
|
Zhang Q, Nettleship I, Schmelzer E, Gerlach J, Zhang X, Wang J, Liu C. Tissue Engineering and Regenerative Medicine Therapies for Cell Senescence in Bone and Cartilage. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:64-78. [DOI: 10.1089/ten.teb.2019.0215] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Qinghao Zhang
- Department of Materials Science and Engineering, East China University of Science and Technology, Shanghai, P.R. China
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Ian Nettleship
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Eva Schmelzer
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jorg Gerlach
- Department of Surgery, McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Xuewei Zhang
- Department of Materials Science and Engineering, East China University of Science and Technology, Shanghai, P.R. China
| | - Jing Wang
- Department of Materials Science and Engineering, East China University of Science and Technology, Shanghai, P.R. China
| | - Changsheng Liu
- Department of Materials Science and Engineering, East China University of Science and Technology, Shanghai, P.R. China
| |
Collapse
|
9
|
Kosiba AA, Wang Y, Chen D, Wong CKC, Gu J, Shi H. The roles of calcium-sensing receptor (CaSR) in heavy metals-induced nephrotoxicity. Life Sci 2019; 242:117183. [PMID: 31874167 DOI: 10.1016/j.lfs.2019.117183] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/15/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023]
Abstract
The kidney is a vital organ responsible for regulating water, electrolyte and acid-base balance as well as eliminating toxic substances from the blood in the body. Exposure of humans to heavy metals in their natural and occupational environments, foods, water, and drugs has serious implications on the kidney's health. The accumulation of heavy metals in the kidney has been linked to acute or chronic renal injury, kidney stones or even renal cancer, at the expense of expensive treatment options. Therefore, unearthing novel biomarkers and potential therapeutic agents or targets against kidney injury for efficient treatment are imperative. The calcium-sensing receptor (CaSR), a G-protein-coupled receptor (GPCR) is typically expressed in the parathyroid glands and renal tubules. It modulates parathyroid hormone secretion according to the serum calcium (Ca2+) concentration. In the kidney, it modulates electrolyte and water excretion by regulating the function of diverse tubular segments. Notably, CaSR lowers passive and active Ca2+ reabsorption in distal tubules, which facilitates phosphate reabsorption in proximal tubules and stimulates proton and water excretion in collecting ducts. Moreover, at the cellular level, modulation of the CaSR regulates cytosolic Ca2+ levels, reactive oxygen species (ROS) generation and the mitogen-activated protein kinase (MAPK) signaling cascades as well as autophagy and the suppression of apoptosis, an effect predominantly triggered by heavy metals. In this regard, we present a review on the CaSR at the cellular level and its potential as a therapeutic target for the development of new and efficient drugs against heavy metals-induced nephrotoxicity.
Collapse
Affiliation(s)
- Anthony A Kosiba
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yanwei Wang
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Dongfeng Chen
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China; Department of Rheumatology and Inflammation Research, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Chris Kong Chu Wong
- Department of Biology, Hong Kong Baptist University, Hong Kong Special Administrative Region
| | - Jie Gu
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| | - Haifeng Shi
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
10
|
Roberts MS, Gafni RI, Brillante B, Guthrie LC, Streit J, Gash D, Gelb J, Krusinska E, Brennan SC, Schepelmann M, Riccardi D, Bin Khayat ME, Ward DT, Nemeth EF, Rosskamp R, Collins MT. Treatment of Autosomal Dominant Hypocalcemia Type 1 With the Calcilytic NPSP795 (SHP635). J Bone Miner Res 2019; 34:1609-1618. [PMID: 31063613 PMCID: PMC6744344 DOI: 10.1002/jbmr.3747] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/05/2019] [Accepted: 03/21/2019] [Indexed: 12/20/2022]
Abstract
Autosomal dominant hypocalcemia type 1 (ADH1) is a rare form of hypoparathyroidism caused by heterozygous, gain-of-function mutations of the calcium-sensing receptor gene (CAR). Individuals are hypocalcemic with inappropriately low parathyroid hormone (PTH) secretion and relative hypercalciuria. Calcilytics are negative allosteric modulators of the extracellular calcium receptor (CaR) and therefore may have therapeutic benefits in ADH1. Five adults with ADH1 due to four distinct CAR mutations received escalating doses of the calcilytic compound NPSP795 (SHP635) on 3 consecutive days. Pharmacokinetics, pharmacodynamics, efficacy, and safety were assessed. Parallel in vitro testing with subject CaR mutations assessed the effects of NPSP795 on cytoplasmic calcium concentrations (Ca2+i ), and ERK and p38MAPK phosphorylation. These effects were correlated with clinical responses to administration of NPSP795. NPSP795 increased plasma PTH levels in a concentration-dependent manner up to 129% above baseline (p = 0.013) at the highest exposure levels. Fractional excretion of calcium (FECa) trended down but not significantly so. Blood ionized calcium levels remained stable during NPSP795 infusion despite fasting, no calcitriol supplementation, and little calcium supplementation. NPSP795 was generally safe and well-tolerated. There was significant variability in response clinically across genotypes. In vitro, all mutant CaRs were half-maximally activated (EC50 ) at lower concentrations of extracellular calcium (Ca2+o ) compared to wild-type (WT) CaR; NPSP795 exposure increased the EC50 for all CaR activity readouts. However, the in vitro responses to NPSP795 did not correlate with any clinical parameters. NPSP795 increased plasma PTH levels in subjects with ADH1 in a dose-dependent manner, and thus, serves as proof-of-concept that calcilytics could be an effective treatment for ADH1. Albeit all mutations appear to be activating at the CaR, in vitro observations were not predictive of the in vivo phenotype or the response to calcilytics, suggesting that other parameters impact the response to the drug. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Mary Scott Roberts
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Rachel I Gafni
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Beth Brillante
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Lori C Guthrie
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Jamie Streit
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| | - David Gash
- NPS Pharmaceuticals, Inc., Bedminster, NJ, USA
| | - Jeff Gelb
- NPS Pharmaceuticals, Inc., Bedminster, NJ, USA
| | | | - Sarah C Brennan
- School of Biosciences, Cardiff University, Cardiff, UK.,School of Life and Environmental Science, University of Sydney, Sydney, NSW, Australia
| | - Martin Schepelmann
- School of Biosciences, Cardiff University, Cardiff, UK.,Institute of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | | | - Mohd Ezuan Bin Khayat
- Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, UK.,Faculty of Biotechnology and Biomolecular Sciences, University Putra Malaysia, Selangor, Malaysia
| | - Donald T Ward
- Institute of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | | | | | - Michael T Collins
- Skeletal Disorders and Mineral Homeostasis Section, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
11
|
Pal S, Porwal K, Khanna K, Gautam MK, Malik MY, Rashid M, Macleod RJ, Wahajuddin M, Parameswaran V, Bellare JR, Chattopadhyay N. Oral dosing of pentoxifylline, a pan-phosphodiesterase inhibitor restores bone mass and quality in osteopenic rabbits by an osteogenic mechanism: A comparative study with human parathyroid hormone. Bone 2019; 123:28-38. [PMID: 30858147 DOI: 10.1016/j.bone.2019.03.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 03/06/2019] [Accepted: 03/08/2019] [Indexed: 10/27/2022]
Abstract
The non-selective phosphodiesterase inhibitor pentoxifylline (PTX) is used for the treatment of intermittent claudication due to artery occlusion. Previous studies in rodents have reported salutary effects of the intraperitoneal administration of PTX in segmental bone defect and fracture healing, as well as stimulation of bone formation. We determined the effect of orally dosed PTX in skeletally mature ovariectomized (OVX) rabbits with osteopenia. The half-maximal effective concentration (EC50) of PTX in rabbit bone marrow stromal cells was 3.07 ± 1.37 nM. The plasma PTX level was 2.05 ± 0.522 nM after a single oral dose of 12.5mg/kg, which was one-sixth of the adult human dose of PTX. Four months of daily oral dosing of PTX at 12.5 mg/kg to osteopenic rabbits completely restored bone mineral density, bone mineral content (BMC), microarchitecture and bone strength to the level of the sham-operated (ovary intact) group. The bone strength to BMC relationship between PTX and sham was similar. The bone restorative effect of PTX was observed in both axial and appendicular bones. In osteopenic rabbits, PTX increased serum amino-terminal propeptide, mineralized nodule formation by stromal cells and osteogenic gene expression in bone. PTX reversed decreased calcium weight percentage and poor crystal packing found in osteopenic rabbits. Furthermore, similar to parathyroid hormone (PTH), PTX had no effect on bone resorption. Taken together, our data show that PTX completely restored bone mass, bone strength and bone mineral properties by an anabolic mechanism. PTX has the potential to become an oral osteogenic drug for the treatment of post-menopausal osteoporosis.
Collapse
Affiliation(s)
- Subhashis Pal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, India
| | - Konica Porwal
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, India
| | - Kunal Khanna
- Department of Chemical Engineering, Indian Institute of Technology-Bombay, Mumbai 400076, India
| | - Manoj Kumar Gautam
- Department of Mechanical Engineering, Indian Institute of Technology-Kanpur, Kanpur 208016, India
| | | | - Mamunur Rashid
- Division of Pharmaceutics, CDRI-CSIR, Lucknow 226031, India
| | - R John Macleod
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | | | - Jayesh R Bellare
- Department of Chemical Engineering, Indian Institute of Technology-Bombay, Mumbai 400076, India
| | - Naibedya Chattopadhyay
- Division of Endocrinology, CSIR-Central Drug Research Institute, Council of Scientific and Industrial Research, Lucknow 226031, India.
| |
Collapse
|
12
|
Feng R, Ding F, Mi XH, Liu SF, Jiang AL, Liu BH, Lian Y, Shi Q, Wang YJ, Zhang Y. Protective Effects of Ligustroflavone, an Active Compound from Ligustrum lucidum, on Diabetes-Induced Osteoporosis in Mice: A Potential Candidate as Calcium-Sensing Receptor Antagonist. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:457-476. [PMID: 30834778 DOI: 10.1142/s0192415x1950023x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Ligustroflavone is one major compound contained in active fraction from Fructus Ligustri Lucidi (the fruit of Ligustrum lucidum), which could regulate parathyroid hormone (PTH) levels and improve calcium balance by acting on calcium-sensing receptors (CaSR). This study aimed to explore the potency of ligustroflavone as a CaSR antagonist and its protective effects against diabetic osteoporosis in mice. LF interacted well with the allosteric site of CaSR shown by molecular docking analysis, increased PTH release of primary parathyroid gland cells and suppressed extracellular calcium influx in HEK-293 cells. The serum level of PTH attained peak value at 2 h and maintained high during the period of 1 h and 3 h than that before treatment in mice after a single dose of LF. Treatment of diabetic mice with LF inhibited the decrease in calcium level of serum and bone and the enhancement in urinary calcium excretion as well as elevated circulating PTH levels. Trabecular bone mineral density and micro-architecture were markedly improved in diabetic mice upon to LF treatment for 8 weeks. LF reduced CaSR mRNA and protein expression in the kidneys of diabetic mice. Taken together, ligustroflavone could transiently increase PTH level and regulate calcium metabolism as well as prevent osteoporosis in diabetic mice, suggesting that ligustroflavone might be an effective antagonist on CaSR.
Collapse
Affiliation(s)
- Rui Feng
- * Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P. R. China
| | - Fan Ding
- * Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P. R. China
| | - Xiu-Hua Mi
- † Section of Nephrology, Yangpu Traditional Chinese Medicine Hospital, Shanghai 200090, P. R. China
| | - Shu-Fen Liu
- * Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P. R. China.,‡ Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai 200032, P. R. China
| | - Ai-Ling Jiang
- § School of Medical Instrument and Food Engineering, University of Shanghai for Science and Technology, Shanghai 200093, P. R. China
| | - Bi-Hui Liu
- ¶ Department of Orthopaedic, Shenzhen Pingle Orthopaedic Hospital, Shenzhen 518000, P. R. China
| | - Yin Lian
- ¶ Department of Orthopaedic, Shenzhen Pingle Orthopaedic Hospital, Shenzhen 518000, P. R. China
| | - Qi Shi
- * Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P. R. China.,‡ Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai 200032, P. R. China
| | - Yong-Jun Wang
- * Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P. R. China.,‡ Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai 200032, P. R. China
| | - Yan Zhang
- * Spine Disease Research Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, P. R. China.,‡ Key Laboratory of Theory and Therapy of Muscles and Bones, Ministry of Education, Shanghai 200032, P. R. China
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Dede AD, Makras P, Anastasilakis AD. Investigational anabolic agents for the treatment of osteoporosis: an update on recent developments. Expert Opin Investig Drugs 2017; 26:1137-1144. [DOI: 10.1080/13543784.2017.1371136] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Anastasia D Dede
- Department of Endocrinology and Diabetes, Chelsea and Westminster Hospital, London, UK
- Laboratory for Research of the Musculoskeletal System ‘Th. Garofalidis,’ KAT Hospital, Medical School, Athens, Greece
| | - Polyzois Makras
- Department of Endocrinology and Diabetes, 251 Hellenic Air Force & VA General Hospital, Athens, Greece
| | | |
Collapse
|
16
|
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
|
17
|
Montesi M, Panseri S, Dapporto M, Tampieri A, Sprio S. Sr-substituted bone cements direct mesenchymal stem cells, osteoblasts and osteoclasts fate. PLoS One 2017; 12:e0172100. [PMID: 28196118 PMCID: PMC5308610 DOI: 10.1371/journal.pone.0172100] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 01/31/2017] [Indexed: 12/14/2022] Open
Abstract
Strontium-substituted apatitic bone cements enriched with sodium alginate were developed as a potential modulator of bone cells fate. The biological impact of the bone cement were investigated in vitro through the study of the effect of the nanostructured apatitic composition and the doping of strontium on mesenchymal stem cells, pre-osteoblasts and osteoclasts behaviours. Up to 14 days of culture the bone cells viability, proliferation, morphology and gene expression profiles were evaluated. The results showed that different concentrations of strontium were able to evoke a cell-specific response, in fact an inductive effect on mesenchymal stem cells differentiation and pre-osteoblasts proliferation and an inhibitory effect on osteoclasts activity were observed. Moreover, the apatitic structure of the cements provided a biomimetic environment suitable for bone cells growth. Therefore, the combination of biological features of this bone cement makes it as promising biomaterials for tissue regeneration.
Collapse
Affiliation(s)
- Monica Montesi
- Institute of Science and Technology for Ceramics, National Research Council, Faenza, Ravenna, Italy
| | - Silvia Panseri
- Institute of Science and Technology for Ceramics, National Research Council, Faenza, Ravenna, Italy
| | - Massimiliano Dapporto
- Institute of Science and Technology for Ceramics, National Research Council, Faenza, Ravenna, Italy
| | - Anna Tampieri
- Institute of Science and Technology for Ceramics, National Research Council, Faenza, Ravenna, Italy
| | - Simone Sprio
- Institute of Science and Technology for Ceramics, National Research Council, Faenza, Ravenna, Italy
| |
Collapse
|
18
|
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
|
19
|
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]
|
20
|
Sha NN, Zhao YJ, Zhao DF, Mok DKW, Shi Q, Wang YJ, Zhang Y. Effect of the water fraction isolated from Fructus Ligustri Lucidi extract on bone metabolismviaantagonizing a calcium-sensing receptor in experimental type 1 diabetic rats. Food Funct 2017; 8:4703-4712. [PMID: 29165475 DOI: 10.1039/c7fo01259d] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Osteoprotective effects of FLL water fraction by potentially regulating vitamin D metabolism and calcium transporters as well as CaSR.
Collapse
Affiliation(s)
- Nan-Nan Sha
- Spine Disease Research Institute
- Longhua Hospital
- Shanghai University of Traditional Chinese Medicine
- Shanghai 200032
- China
| | - Yong-Jian Zhao
- Spine Disease Research Institute
- Longhua Hospital
- Shanghai University of Traditional Chinese Medicine
- Shanghai 200032
- China
| | - Dong-Feng Zhao
- Spine Disease Research Institute
- Longhua Hospital
- Shanghai University of Traditional Chinese Medicine
- Shanghai 200032
- China
| | - Daniel Kam-Wah Mok
- Department of Applied Biology and Chemical Technology
- The Hong Kong Polytechnic University
- Kowloon
- China
| | - Qi Shi
- Spine Disease Research Institute
- Longhua Hospital
- Shanghai University of Traditional Chinese Medicine
- Shanghai 200032
- China
| | - Yong-Jun Wang
- Spine Disease Research Institute
- Longhua Hospital
- Shanghai University of Traditional Chinese Medicine
- Shanghai 200032
- China
| | - Yan Zhang
- Spine Disease Research Institute
- Longhua Hospital
- Shanghai University of Traditional Chinese Medicine
- Shanghai 200032
- China
| |
Collapse
|
21
|
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
|
22
|
Mayr B, Glaudo M, Schöfl C. Activating Calcium-Sensing Receptor Mutations: Prospects for Future Treatment with Calcilytics. Trends Endocrinol Metab 2016; 27:643-652. [PMID: 27339034 DOI: 10.1016/j.tem.2016.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 05/17/2016] [Accepted: 05/20/2016] [Indexed: 12/20/2022]
Abstract
Activating mutations of the G protein-coupled receptor, calcium-sensing receptor (CaSR), cause autosomal dominant hypocalcemia and Bartter syndrome type 5. These mutations lower the set-point for extracellular calcium sensing, thereby causing decreased parathyroid hormone secretion and disturbed renal calcium handling with hypercalciuria. Available therapies increase serum calcium levels but raise the risk of complications in affected patients. Symptom relief and the prevention of adverse outcome is currently very difficult to achieve. Calcilytics act as CaSR antagonists that attenuate its activity, thereby correcting the molecular defect of activating CaSR proteins in vitro and elevating serum calcium in mice and humans in vivo, and have emerged as the most promising therapeutics for the treatment of these rare and difficult to treat diseases.
Collapse
Affiliation(s)
- Bernhard Mayr
- Division of Endocrinology and Diabetes, Department of Medicine I, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Germany.
| | - Markus Glaudo
- Division of Endocrinology and Diabetes, Department of Medicine I, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| | - Christof Schöfl
- Division of Endocrinology and Diabetes, Department of Medicine I, Universitätsklinikum Erlangen, Friedrich-Alexander University Erlangen-Nuremberg, Germany
| |
Collapse
|
23
|
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
|
24
|
Dong B, Endo I, Ohnishi Y, Kondo T, Hasegawa T, Amizuka N, Kiyonari H, Shioi G, Abe M, Fukumoto S, Matsumoto T. Calcilytic Ameliorates Abnormalities of Mutant Calcium-Sensing Receptor (CaSR) Knock-In Mice Mimicking Autosomal Dominant Hypocalcemia (ADH). J Bone Miner Res 2015; 30:1980-93. [PMID: 25967373 DOI: 10.1002/jbmr.2551] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 04/16/2015] [Accepted: 05/07/2015] [Indexed: 02/01/2023]
Abstract
Activating mutations of calcium-sensing receptor (CaSR) cause autosomal dominant hypocalcemia (ADH). ADH patients develop hypocalcemia, hyperphosphatemia, and hypercalciuria, similar to the clinical features of hypoparathyroidism. The current treatment of ADH is similar to the other forms of hypoparathyroidism, using active vitamin D3 or parathyroid hormone (PTH). However, these treatments aggravate hypercalciuria and renal calcification. Thus, new therapeutic strategies for ADH are needed. Calcilytics are allosteric antagonists of CaSR, and may be effective for the treatment of ADH caused by activating mutations of CaSR. In order to examine the effect of calcilytic JTT-305/MK-5442 on CaSR harboring activating mutations in the extracellular and transmembrane domains in vitro, we first transfected a mutated CaSR gene into HEK cells. JTT-305/MK-5442 suppressed the hypersensitivity to extracellular Ca(2+) of HEK cells transfected with the CaSR gene with activating mutations in the extracellular and transmembrane domains. We then selected two activating mutations locating in the extracellular (C129S) and transmembrane (A843E) domains, and generated two strains of CaSR knock-in mice to build an ADH mouse model. Both mutant mice mimicked almost all the clinical features of human ADH. JTT-305/MK-5442 treatment in vivo increased urinary cAMP excretion, improved serum and urinary calcium and phosphate levels by stimulating endogenous PTH secretion, and prevented renal calcification. In contrast, PTH(1-34) treatment normalized serum calcium and phosphate but could not reduce hypercalciuria or renal calcification. CaSR knock-in mice exhibited low bone turnover due to the deficiency of PTH, and JTT-305/MK-5442 as well as PTH(1-34) increased bone turnover and bone mineral density (BMD) in these mice. These results demonstrate that calcilytics can reverse almost all the phenotypes of ADH including hypercalciuria and renal calcification, and suggest that calcilytics can become a novel therapeutic agent for ADH.
Collapse
Affiliation(s)
- Bingzi Dong
- Department of Medicine and Bioregulatory Sciences, University of Tokushima Graduate School of Medical Sciences, Tokushima, Japan
| | - Itsuro Endo
- Department of Medicine and Bioregulatory Sciences, University of Tokushima Graduate School of Medical Sciences, Tokushima, Japan
| | - Yukiyo Ohnishi
- Department of Medicine and Bioregulatory Sciences, University of Tokushima Graduate School of Medical Sciences, Tokushima, Japan
| | - Takeshi Kondo
- Department of Medicine and Bioregulatory Sciences, University of Tokushima Graduate School of Medical Sciences, Tokushima, Japan
| | - Tomoka Hasegawa
- Department of Developmental Biology of Hard Tissue, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Norio Amizuka
- Department of Developmental Biology of Hard Tissue, Hokkaido University Graduate School of Dental Medicine, Sapporo, Japan
| | - Hiroshi Kiyonari
- Animal Resource Development Unit, RIKEN Center for Life Science Technologies, Kobe, Japan
| | - Go Shioi
- Genetic Engineering Team, RIKEN Center for Life Science Technologies, Kobe, Japan
| | - Masahiro Abe
- Department of Medicine and Bioregulatory Sciences, University of Tokushima Graduate School of Medical Sciences, Tokushima, Japan
| | - Seiji Fukumoto
- Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| | - Toshio Matsumoto
- Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
Kim JY, Ho H, Kim N, Liu J, Tu CL, Yenari MA, Chang W. Calcium-sensing receptor (CaSR) as a novel target for ischemic neuroprotection. Ann Clin Transl Neurol 2014; 1:851-66. [PMID: 25540800 PMCID: PMC4265057 DOI: 10.1002/acn3.118] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Revised: 08/14/2014] [Accepted: 08/15/2014] [Indexed: 01/04/2023] Open
Abstract
Object Ischemic brain injury is the leading cause for death and long-term disability in patients who suffer cardiac arrest and embolic stroke. Excitotoxicity and subsequent Ca2+-overload lead to ischemic neuron death. We explore a novel mechanism concerning the role of the excitatory extracellular calcium-sensing receptor (CaSR) in the induction of ischemic brain injury. Method Mice were exposed to forebrain ischemia and the actions of CaSR were determined after its genes were ablated specifically in hippocampal neurons or its activities were inhibited pharmacologically. Since the CaSR forms a heteromeric complex with the inhibitory type B γ-aminobutyric acid receptor 1 (GABABR1), we compared neuronal responses to ischemia in mice deficient in CaSR, GABABR1, or both, and in mice injected locally or systemically with a specific CaSR antagonist (or calcilytic) in the presence or absence of a GABABR1 agonist (baclofen). Results Both global and focal brain ischemia led to CaSR overexpression and GABABR1 downregulation in injured neurons. Genetic ablation of Casr genes or blocking CaSR activities by calcilytics rendered robust neuroprotection and preserved learning and memory functions in ischemic mice, partly by restoring GABABR1 expression. Concurrent ablation of Gabbr1 gene blocked the neuroprotection caused by the Casr gene knockout. Coinjection of calcilytics with baclofen synergistically enhanced neuroprotection. This combined therapy remained robust when given 6 h after ischemia. Interpretation Our study demonstrates a novel receptor interaction, which contributes to ischemic neuron death through CaSR upregulation and GABABR1 downregulation, and feasibility of neuroprotection by concurrently targeting these two receptors.
Collapse
Affiliation(s)
- Jong Youl Kim
- Endocrine Unit, University of California San Francisco and Veterans Affairs Medical Center San Francisco, California, 94121 ; Department of Neurology, University of California San Francisco and Veterans Affairs Medical Center San Francisco, California, 94121
| | - Hanson Ho
- Endocrine Unit, University of California San Francisco and Veterans Affairs Medical Center San Francisco, California, 94121
| | - Nuri Kim
- Department of Neurology, University of California San Francisco and Veterans Affairs Medical Center San Francisco, California, 94121
| | - Jialing Liu
- Neurological Surgery, University of California San Francisco and Veterans Affairs Medical Center San Francisco, California, 94121
| | - Chia-Ling Tu
- Endocrine Unit, University of California San Francisco and Veterans Affairs Medical Center San Francisco, California, 94121
| | - Midori A Yenari
- Department of Neurology, University of California San Francisco and Veterans Affairs Medical Center San Francisco, California, 94121
| | - Wenhan Chang
- Endocrine Unit, University of California San Francisco and Veterans Affairs Medical Center San Francisco, California, 94121
| |
Collapse
|
28
|
Fitzpatrick LA, Wooddell M, Dabrowski CE, Cicconetti G, Gordon DN. Bone mineral density changes following discontinuation of ronacaleret treatment: off-treatment extension of a randomized, dose-finding phase II trial. Bone 2014; 67:104-8. [PMID: 24794093 DOI: 10.1016/j.bone.2014.04.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Revised: 04/21/2014] [Accepted: 04/22/2014] [Indexed: 10/25/2022]
Abstract
CONTEXT Parathyroidectomy in patients with hyperparathyroidism can produce subsequent increases in bone mineral density (BMD). Ronacaleret, a selective calcium-sensing receptor antagonist that stimulates endogenous parathyroid hormone release, induced mild hyperparathyroidism. OBJECTIVE The aim of this study is to evaluate whether BMD changes after cessation of ronacaleret treatment. DESIGN Observational, off-treatment, extension of a randomized, placebo-controlled, dose-ranging phase II trial. SETTING Fifteen academic centers in seven countries. PATIENTS Postmenopausal women with low BMD; 171 out of 569 women in the parent study were enrolled in the extension study. INTERVENTIONS Subjects were treated with ronacaleret 100mg (n=16), 200mg (n=38), 300mg (n=35), or 400mg (n=32) once daily, alendronate 70mg (n=17) once weekly, or matching placebo (n=33) for 10-12months; BMD was measured after discontinuation of ronacaleret or alendronate treatment. MAIN OUTCOME MEASURE Mean percent change in lumbar spine areal BMD by dual-energy X-ray absorptiometry at 6-12months after discontinuing ronacaleret or alendronate compared with the 10- to 12-month BMD measurement of the parent study. RESULTS At the lumbar spine, all doses of ronacaleret resulted in gains in BMD while on treatment. These increases in BMD were maintained or increased after discontinuation of ronacaleret. All doses of ronacaleret caused bone loss at the total hip while on active treatment. However, there was an attenuation of this loss in the off-treatment extension study. CONCLUSION The gain in BMD at the lumbar spine was maintained post-treatment and the loss of BMD at the total hip was attenuated. We hypothesize that there may have been some bone remineralization after cessation of ronacaleret.
Collapse
|
29
|
John MR, Harfst E, Loeffler J, Belleli R, Mason J, Bruin GJM, Seuwen K, Klickstein LB, Mindeholm L, Widler L, Kneissel M. AXT914 a novel, orally-active parathyroid hormone-releasing drug in two early studies of healthy volunteers and postmenopausal women. Bone 2014; 64:204-10. [PMID: 24769332 DOI: 10.1016/j.bone.2014.04.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 03/25/2014] [Accepted: 04/14/2014] [Indexed: 11/17/2022]
Abstract
Antagonism of the calcium-sensing receptor in the parathyroid gland leads to parathyroid hormone (PTH) release. Calcilytics are a new class of molecules designed to exploit this mechanism. In order to mimic the known bone-anabolic pharmacokinetic (PK) profile of s.c. administered PTH, such molecules must trigger sharp, transient and robust release of PTH. The results of two early clinical studies with the orally-active calcilytic AXT914, a quinazolin-2ne derivative are reported. These were GCP-compliant, single and multiple dose studies of PK/PD and tolerability in healthy volunteers and postmenopausal women. The first study, examined single ascending doses (4 to 120 mg) and limited multiple doses (60 or 120 mgq.d. for 12 days) of AXT914. The second study was a randomized, double-blind, active- and placebo-controlled, 4-week repeat-dose parallel group study of healthy postmenopausal women (45 and 60 mg AXT914, placebo, 20 μg Forteo/teriparatide/PTH(1-34) fragment). AXT914 was well tolerated at all doses and reproducibly induced the desired PTH-release profiles. Yet, 4 weeks of 45 or 60 mg AXT914 did not result in the expected changes in circulating bone biomarkers seen with teriparatide. However total serum calcium levels increased above baseline in the 45 and 60 mg AXT914 treatment groups (8.0% and 10.7%, respectively), compared to that in the teriparatide and placebo groups (1.3% and 1.0%, respectively). Thus the trial was terminated after a planned interim analysis due to lack of effect on bone formation biomarkers and dose-limiting effects on serum calcium. In conclusion, AXT914 was well tolerated but the observed transient and reproducible PTH-release after repeat oral administration of AXT914 which showed an exposure profile close to that of s c. PTH, did not translate into a bone anabolic response and was associated with a persistent dose-related increase in serum calcium concentrations.
Collapse
Affiliation(s)
- Markus R John
- Novartis Pharma AG, Basel, Switzerland, Novartis Corporation, East Hanover, NJ, USA
| | - Evita Harfst
- Novartis Pharma AG, Basel, Switzerland, Novartis Corporation, East Hanover, NJ, USA
| | - Juergen Loeffler
- Novartis Pharma AG, Basel, Switzerland, Novartis Corporation, East Hanover, NJ, USA
| | - Rossella Belleli
- Novartis Pharma AG, Basel, Switzerland, Novartis Corporation, East Hanover, NJ, USA
| | - June Mason
- Novartis Pharma AG, Basel, Switzerland, Novartis Corporation, East Hanover, NJ, USA
| | - Gerard J M Bruin
- Novartis Institutes for Biomedical Research, Basel, Switzerland and Cambridge, MA, USA
| | - Klaus Seuwen
- Novartis Institutes for Biomedical Research, Basel, Switzerland and Cambridge, MA, USA
| | - Lloyd B Klickstein
- Novartis Institutes for Biomedical Research, Basel, Switzerland and Cambridge, MA, USA
| | - Linda Mindeholm
- Novartis Pharma AG, Basel, Switzerland, Novartis Corporation, East Hanover, NJ, USA
| | - Leo Widler
- Novartis Institutes for Biomedical Research, Basel, Switzerland and Cambridge, MA, USA
| | - Michaela Kneissel
- Novartis Institutes for Biomedical Research, Basel, Switzerland and Cambridge, MA, USA.
| |
Collapse
|
30
|
Abstract
Osteoporosis is a chronic disease of the osseous system characterized by decreased bone strength and increased fracture risk. It is due to an imbalance in the dynamic ongoing processes of bone formation and bone resorption. Currently available osteoporosis therapies like bisphosphonates, selective estrogen receptor modulators (SERMs), and denosumab are anti-resorptive agents. Parathyroid hormone analogs like teriparatide are the only anabolic agents currently approved for osteoporosis treatment. The side-effects and limited efficacy of the presently available therapies has encouraged extensive research into the pathophysiology of the disease and newer drug targets for its treatment. The novel anti-resorptive agents being developed are newer SERMs, osteoprotegerin, c-src (cellular-sarcoma) kinase inhibitors, αVβ3 integrin antagonists, cathepsin K inhibitors, chloride channel inhibitors, and nitrates. Upcoming anabolic agents include calcilytics, antibodies against sclerostin and Dickkopf-1, statins, matrix extracellular phosphoglycoprotein fragments activin inhibitiors, and endo-cannabinoid agonists. Many of these new drugs are still in development. This article provides an insight into the emerging drugs for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Garima Bhutani
- Department of Pharmacology, Pt. B.D. Sharma Post Graduate Institute of Medical Sciences, Rohtak, Haryana, India
| | - Mahesh Chander Gupta
- Department of Pharmacology, Pt. B.D. Sharma Post Graduate Institute of Medical Sciences, Rohtak, Haryana, India
| |
Collapse
|
31
|
Scott JPR, Sale C, Greeves JP, Casey A, Dutton J, Fraser WD. Treadmill running reduces parathyroid hormone concentrations during recovery compared with a nonexercising control group. J Clin Endocrinol Metab 2014; 99:1774-82. [PMID: 24476072 DOI: 10.1210/jc.2013-3027] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Lower PTH concentrations reported in the hours after acute, endurance exercise compared with preexercise levels might be influenced by factors such as circadian fluctuations. OBJECTIVE The objective of the study was to compare postexercise PTH concentrations with a nonexercising control group. DESIGN AND SETTING A laboratory-based study with a crossover design, comparing a 60-minute (at 10:30 am) bout of treadmill running at 65% of the maximal rate of oxygen uptake (exercise) with semirecumbent rest (CON). Blood samples were obtained immediately before (baseline 10:15 am) and after (11:30 am) exercise and during recovery (12:30 am, 1:30 pm, and 2:15 pm). PARTICIPANTS Ten physically active men (mean ± 1 SD, age 26 ± 5 y; body mass 78.3 ± 5.8 kg; maximal rate of oxygen uptake 57.3 ± 6.9 mL/kg(-1) · min(-1)) participated in the study. MAIN OUTCOME MEASURES PTH, albumin-adjusted calcium, and phosphate concentrations were measured. RESULTS PTH concentrations increased (+85%, P < .01) during exercise and were higher than in CON immediately at the end of exercise (4.5 ± 1.9 vs 2.6 ± 0.9 pmol/L(-1), P < .05). In the postexercise period (12:30-2:15 pm), PTH was not different compared with baseline but was lower compared with CON at 1:30 pm (-22%; P < .01) and tended to be lower at both 12:30 pm (-12%; P = .063) and 2:15 pm (-13%; P = .057). Exercise did not significantly affect the albumin-adjusted calcium concentrations, whereas phosphate was higher than CON immediately after exercise (1.47 ± 0.17 vs 1.03 ± 0.17 pmol/L(-1), P < .001) and was lower at 1:30 pm (-16%: P < .05). CONCLUSIONS Lower PTH concentrations after acute endurance running compared with a rested control condition suggest a true effect of exercise.
Collapse
Affiliation(s)
- Jonathan P R Scott
- Human Sciences (J.P.R.S., A.C.), QinetiQ Ltd, Farnborough GU14 0LX, United Kingdom; Biomedical, Life, and Health Sciences Research Centre (C.S.), School of Science and Technology, Nottingham Trent University, NG1 4BU Nottingham, United Kingdom; Department of Occupational Medicine (J.P.G.), Headquarters Army Recruiting and Training Division, Upavon SN9 6BE, Wiltshire, United Kingdom; Department of Musculoskeletal Biology (J.D.), University of Liverpool, Liverpool L69 7ZX, United Kingdom; and Norwich Medical School (W.D.F.), University of East Anglia, Norwich NR4 7TJ United Kingdom
| | | | | | | | | | | |
Collapse
|
32
|
Kim JY, Kim N, Yenari MA, Chang W. Hypothermia and pharmacological regimens that prevent overexpression and overactivity of the extracellular calcium-sensing receptor protect neurons against traumatic brain injury. J Neurotrauma 2014; 30:1170-6. [PMID: 23360235 DOI: 10.1089/neu.2012.2691] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Traumatic brain injury (TBI) leads to acute functional deficit in the brain. Molecular events underlying TBI remain unclear. In mouse brains, we found controlled cortical impact (CCI) injury induced overexpression of the extracellular calcium-sensing receptor (CaSR), which is known to stimulate neuronal activity and accumulation of intracellular Ca(2+) and concurrent down-regulation of type B or metabotropic GABA receptor 1 (GABA-B-R1), a prominent inhibitory pathway in the brain. These changes in protein expression preceded and were closely associated with the loss of brain tissue, as indicated by the increased size of cortical cavity at impact sites, and the development of motor deficit, as indicated by the increased frequency of right-biased swing and turn in the CCI mice. Mild hypothermia, an established practice of neuroprotection for brain ischemia, partially but significantly blunted all of the above effects of CCI. Administration of CaSR antagonist NPS89636 mimicked hypothermia to reduce loss of brain tissue and motor functions in the CCI mice. These data together support the concept that CaSR overexpression and overactivity play a causal role in potentiating TBI potentially by stimulating excitatory neuronal responses and by interfering with inhibitory GABA-B-R signaling and that the CaSR could be a novel target for neuroprotection against TBI.
Collapse
Affiliation(s)
- Jong Youl Kim
- University of California, San Francisco, California, USA
| | | | | | | |
Collapse
|
33
|
Saidak Z, Marie PJ. Strontium signaling: Molecular mechanisms and therapeutic implications in osteoporosis. Pharmacol Ther 2012; 136:216-26. [DOI: 10.1016/j.pharmthera.2012.07.009] [Citation(s) in RCA: 202] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Accepted: 07/06/2012] [Indexed: 12/15/2022]
|
34
|
Abstract
INTRODUCTION The biological process of fracture healing is complex with influences that are both patient-dependent and related to the trauma experienced and stability of the fracture. Fracture healing complications negatively affect the patient's quality of life, even more when fractures occur in the elderly osteoporotic patients. AREAS COVERED In the polytherapy for bone regeneration, a high success rate was obtained with the use of growth factors, osteogenic cells, and osteoconductive factors. There have been high expectations that treatment with drugs active on bone remodeling would be efficient for acceleration of fracture healing. A literature search was undertaken using wording like "drug or pharmacology of fracture healing." This report will review the systemic pharmacological agents for which clinical trials documenting their efficacy on bone healing have been carried out or are underway. EXPERT OPINION At present the use of systemic pharmacological agents to enhance fracture healing in the clinical setting is still controversial. However, future clinical trials will offer the possibility to obtain data that will make possible the registration of a drug as a "healer."
Collapse
Affiliation(s)
- Maria Luisa Brandi
- University of Florence, Department of Surgery and Translational Medicine, Mineral and Bone Metabolic Diseases Unit, Largo Palagi, 1, 50100 Florence, Italy.
| |
Collapse
|
35
|
Fisher JE, Scott K, Wei N, Zhao JZ, Cusick T, Tijerina M, Karanam B, Duong L, Glantschnig H. Pharmacodynamic responses to combined treatment regimens with the calcium sensing receptor antagonist JTT-305/MK-5442 and alendronate in osteopenic ovariectomized rats. Bone 2012; 50:1332-42. [PMID: 22445539 DOI: 10.1016/j.bone.2012.03.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Revised: 02/20/2012] [Accepted: 03/05/2012] [Indexed: 11/26/2022]
Abstract
Parathyroid hormone (PTH) is the anabolic standard of care for patients with severe osteoporosis. The CaSR allosteric antagonist JTT-305/MK-5442, a PTH secretagogue, could offer an oral osteoanabolic treatment alternative for postmenopausal women with osteoporosis. Here we disclose the pharmacokinetic profile of JTT-305/MK-5442 and its activity on bone remodeling in ovariectomized (OVX) osteopenic rats. Daily treatments (0.3 to 2.4 mg/kg/d) for 12 weeks resulted in plateaued BMD increases (3.8 to 5.3%) at axial and appendicular skeletal sites. However, treatment effects were not statistically significant, in agreement with effects seen in animals treated with low dose PTH (1-84) (5 μg/kg/d). In a consecutive study we tested JTT-305/MK-5442 effects on bone formation in OVX-rats challenged with combined alendronate (ALN) treatment paradigms. At 7 month, JTT-305/MK-5442 treatment significantly increased BMD in lumbar vertebrae (LV), while no change in BMD was observed in femora or tibiae. ALN add-on co-treatment produced incremental increases in LV, distal femur (DF) and proximal tibia (PT) BMD over the respective ALN control. Histological analyses confirmed modest increases in mineralized surface (MS/BS) and bone formation rate (30.5±1.9%) on trabecular surfaces by JTT-305/MK-5442. As expected, ALN administration profoundly reduced bone formation, however, JTT-305/MK-5442 significantly stimulated MS/BS and BFR in ALN treated groups. In summary, JTT-305/MK-5442 acts as a PTH secretagogue in the osteopenic OVX-rat, eliciting consistent, though modest effects on remediation of BMD due to estrogen depletion. Induction of bone formation by JTT-305/MK-5442 at trabecular bone surfaces appears to be resilient to ALN-mediated suppression of bone formation. This study provides for the first time, a mechanistic evaluation of combination treatment of a PTH secretagogue with ALN.
Collapse
Affiliation(s)
- John E Fisher
- Bone Biology, Merck Research Laboratories, West Point, PA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Hämmerle SP, Mindeholm L, Launonen A, Kiese B, Loeffler R, Harfst E, Azria M, Arnold M, John MR. The single dose pharmacokinetic profile of a novel oral human parathyroid hormone formulation in healthy postmenopausal women. Bone 2012; 50:965-73. [PMID: 22289659 DOI: 10.1016/j.bone.2012.01.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 01/10/2012] [Accepted: 01/13/2012] [Indexed: 11/25/2022]
Abstract
Parathyroid hormone (PTH), currently the only marketed anabolic treatment for osteoporosis, is available as the full-length hormone, human PTH1-84, or as the human PTH1-34 fragment (teriparatide). Both must be administered as a daily subcutaneous (sc) injection. A new oral formulation of human PTH1-34 (PTH134) is being developed as a more convenient option for patients. In this single-center, partially-blinded, incomplete cross-over study, the safety, tolerability, and exposure of oral PTH134 (teriparatide combined with 2 different quantities of the absorption enhancer 5-CNAC) were assessed in 32 healthy postmenopausal women. 16 subjects were randomized to receive 4 single doses out of 6 different treatments: placebo, teriparatide 20 μg sc, or 1, 2.5, 5 or 10 mg of oral PTH134 formulated with 200 mg 5-CNAC. Subsequently, another 16 subjects were randomized to receive 4 out of 6 different treatments: placebo, teriparatide 20 μg sc, or 2.5 or 5 mg of oral PTH134 formulated with either 100 or 200 mg 5-CNAC. Doses were given ≥6 days apart. All doses of PTH134 were rapidly absorbed, and showed robust blood concentrations in a dose-dependent manner. Interestingly, PTH1-34 disappeared from blood faster after oral than after sc administration. Specifically, 2.5 and 5 mg PTH134 (containing 200 mg 5-CNAC) demonstrated Cmax and AUC0-last values closest to those of sc teriparatide 20 μg (Forsteo®). Mean+/-SD hPTH134 Cmax values were, respectively, 74+/-59, 138+/-101, 717+/-496, and 1624+/-1579 pg/mL for 1, 2.5, 5, and 10 mg doses of this peptide administered with 200 mg 5-CNAC; while mean+/-SD AUC (0-last) values were, respectively, 30+/-40, 62+/-69, 320+/-269, and 627+/-633 h*pg/mL. The corresponding estimates for teriparatide 20 μg sc were 149+/-35 for Cmax and 236+/-58 for AUC (0-last) Ionized calcium remained within normal limits in all treatment groups except for 3 isolated events. Nine subjects withdrew due to treatment-related AEs. Of those, seven were taking PTH134 2.5 or 5 mg: three withdrew for symptomatic hypotension (two of whom were in the 200 mg 5-CNAC group), three because of delayed vomiting (two from the 200 mg 5-CNAC group), one was proactively withdrawn by the investigator for symptomatic hypercalcemia (receiving 2.5 mg/100 mg 5-CNAC) at slightly supra-normal total calcium but normal ionized serum calcium levels. One subject receiving teriparatide and one receiving placebo withdrew for symptomatic hypotension. No serious AEs were reported. In conclusion, the study demonstrated potential therapeutically relevant PTH1-34 systemic exposure levels after oral administration of PTH1-34 formulated with the absorption enhancer 5-CNAC. Doses of 2.5 and 5 mg of oral PTH134 achieved exposure levels closest to those of teriparatide 20 μg sc, with a comparable incidence of AEs in healthy postmenopausal women.
Collapse
Affiliation(s)
- Sibylle P Hämmerle
- Novartis Pharma AG, Integrated Hospital Care Franchise, Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Rachner TD, Hadji P, Hofbauer LC. Novel therapies in benign and malignant bone diseases. Pharmacol Ther 2012; 134:338-44. [PMID: 22401778 DOI: 10.1016/j.pharmthera.2012.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Accepted: 02/15/2012] [Indexed: 01/31/2023]
Abstract
With an ageing population and improving cancer therapies, the two most common benign and malignant bone diseases, osteoporosis and bone metastases, will continue to affect an increasing number of patients. Our expanding knowledge of the molecular processes underlying these conditions has resulted in novel bone targets that are currently being explored in clinical trials. Clearly, the approval of denosumab, a monoclonal antibody directed against RANKL, has just marked the beginning of a new era for bone therapy with several additional new therapies lining up for clinical approval in the coming years. Potential agents targeting the osteoclast include cathepsin K, currently in phase 3 trials, and src inhibitors. Amongst anabolic agents, inhibitors of the Wnt-inhibitor sclerostin and dickkopf-1 are promising in clinical trials. Here, we will provide a comprehensive overview of the most promising agents currently explored for the treatment of bone diseases.
Collapse
Affiliation(s)
- Tilman D Rachner
- Division of Endocrinology, Diabetes, and Bone Diseases (TDR, LCH), Technical University, Dresden, Germany
| | | | | |
Collapse
|
38
|
Fitzpatrick LA, Dabrowski CE, Cicconetti G, Gordon DN, Fuerst T, Engelke K, Genant HK. Ronacaleret, a calcium-sensing receptor antagonist, increases trabecular but not cortical bone in postmenopausal women. J Bone Miner Res 2012; 27:255-62. [PMID: 22052452 DOI: 10.1002/jbmr.554] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Intermittent injections of parathyroid hormone have osteoanabolic effects that increase bone mineral density (BMD). Ronacaleret is an orally administered calcium-sensing receptor antagonist that stimulates endogenous parathyroid hormone release from the parathyroid glands. Our objective was to compare the effects of ronacaleret and teriparatide on volumetric BMD (vBMD) measured by quantitative computed tomography (QCT). We conducted a randomized, placebo-controlled, dose-ranging trial at 45 academic centers with 31 sites participating in the substudy. Patients included 569 postmenopausal women with low bone mineral density; vBMD was assessed at the spine and hip in a subset of 314 women. Patients were treated for up to 12 months with open-label teriparatide 20 µg subcutaneously once daily or randomly assigned in a double-blind manner to ronacaleret 100 mg, 200 mg, 300 mg, or 400 mg once daily, alendronate 70 mg once weekly, or matching placebos. Ronacaleret increased spine integral (0.49% to 3.9%) and trabecular (1.8% to 13.3%) vBMD compared with baseline, although the increments were at least twofold lower than that attained with teriparatide (14.8% and 24.4%, respectively) but similar or superior to that attained with alendronate (5.0% and 4.9%, respectively). There were small non-dose-dependent decreases in integral vBMD of the proximal femur with ronacaleret (-0.1 to -0.8%) compared with increases in the teriparatide (3.9%) and alendronate (2.7%) arms. Parathyroid hormone (PTH) elevations with ronacaleret were prolonged relative to that seen historically with teriparatide. Ronacaleret preferentially increased vBMD of trabecular bone that is counterbalanced by small decreases in BMD at cortical sites. The relative preservation of trabecular bone and loss at cortical sites are consistent with the induction of mild hyperparathyroidism with ronacaleret therapy.
Collapse
|
39
|
Baron R, Hesse E. Update on bone anabolics in osteoporosis treatment: rationale, current status, and perspectives. J Clin Endocrinol Metab 2012; 97:311-25. [PMID: 22238383 PMCID: PMC3275361 DOI: 10.1210/jc.2011-2332] [Citation(s) in RCA: 246] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Osteoporosis is defined as low bone mineral density associated with skeletal fractures secondary to minimal or no trauma, most often involving the spine, the hip, and the forearm. The decrease in bone mineral density is the consequence of an unbalanced bone remodeling process, with higher bone resorption than bone formation. Osteoporosis affects predominantly postmenopausal women, but also older men. This chronic disease represents a considerable medical and socioeconomic burden for modern societies. The therapeutic options for the treatment of osteoporosis have so far comprised mostly antiresorptive drugs, in particular bisphosphonates and more recently denosumab, but also calcitonin and, for women, estrogens or selective estrogen receptor modulators. These drugs have limitations, however, in particular the fact that they lead to a low turnover state where bone formation decreases with the decrease in bone-remodeling activity. In this review, we discuss the alternative class of osteoporosis drugs, i.e. bone anabolics, their biology, and the perspectives they offer for our therapeutic armamentarium. We focus on the two main osteoanabolic pathways identified as of today: PTH, the only anabolic drug currently on the market; and activation of canonical Wnt signaling through inhibition of the endogenous inhibitors sclerostin and dickkopf1. Each approach is based on a different molecular mechanism, but most recent evidence suggests that these two pathways may actually converge, at least in part. Whereas recombinant human PTH treatment is being revisited with different formulations and attempts to regulate endogenous PTH secretion via the calcium-sensing receptor, antibodies to sclerostin and dickkopf1 are currently in clinical trials and may prove to be even more efficient at increasing bone mass, possibly independent of bone turnover. Each of these anabolic approaches has its own limitations and safety issues, but the prospects of effective anabolic therapy for osteoporosis are indeed bright.
Collapse
Affiliation(s)
- Roland Baron
- Department of Medicine, Harvard Medical School, Endocrine Unit, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.
| | | |
Collapse
|
40
|
Melancon BJ, Hopkins CR, Wood MR, Emmitte KA, Niswender CM, Christopoulos A, Conn PJ, Lindsley CW. Allosteric modulation of seven transmembrane spanning receptors: theory, practice, and opportunities for central nervous system drug discovery. J Med Chem 2012; 55:1445-64. [PMID: 22148748 DOI: 10.1021/jm201139r] [Citation(s) in RCA: 183] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Bruce J Melancon
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
The calcium-sensing receptor (CaR) is the key controller of extracellular calcium (Ca(2+)(o)) homeostasis via its regulation of parathyroid hormone (PTH) secretion and renal Ca(2+) reabsorption. The CaR-selective calcimimetic drug Cinacalcet stimulates the CaR to suppress PTH secretion in chronic kidney disease and represents the world's first clinically available receptor positive allosteric modulator (PAM). Negative CaR allosteric modulators (NAMs), known as calcilytics, can increase PTH secretion and are being investigated as possible bone anabolic treatments against age-related osteoporosis. Here we address the current state of development and clinical use of a series of positive and negative CaR modulators. In addition, clinical CaR mutations and transgenic mice carrying tissue-specific CaR deletions have provided a novel understanding of the relative functional importance of CaR in both calciotropic tissues and those elsewhere in the body. The development of CaR-selective modulators and signalling reagents have provided us with a more detailed appreciation of how the CaR signals in vivo. Thus, both of these areas of CaR research will be reviewed.
Collapse
Affiliation(s)
- Donald T Ward
- Faculty of Life Sciences, The University of ManchesterManchester, UK
| | | |
Collapse
|
42
|
Improvement of calcium balance by Fructus Ligustri Lucidi extract in mature female rats was associated with the induction of serum parathyroid hormone levels. Br J Nutr 2011; 108:92-101. [PMID: 22018100 DOI: 10.1017/s0007114511005186] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Fructus Ligustri Lucidi (FLL) is a commonly prescribed herb in many kidney-tonifying Traditional Chinese Medicinal formulae for the treatment of osteoporosis. The present study aimed to identify the active fractions in FLL and to characterise its effects on Ca balance, calciotropic hormone levels as well as bone properties in mature female rats fed diets containing different levels of Ca. In the present study, 4-month-old Sprague-Dawley female rats were treated with either FLL ethanol extract (EE), ethyl acetate-soluble fraction of EE (EAF), water-soluble fraction of EE (WF) or their vehicle for 12 weeks on a medium-Ca diet (MCD, 0·6 % Ca, 0·65 % P). Then, the Sprague-Dawley female rats treated with WF or its vehicle for 12 weeks were fed diets containing different levels of dietary Ca (low-Ca diet (LCD), 0·1 % Ca, 0·65 % P; MCD; high-Ca diet (HCD), 1·2 % Ca, 0·65 % P). The results demonstrated that WF from EE but not EAF exerted a prominent effect on Ca balance by inhibiting urinary and faecal Ca excretion. WF significantly increased Ca balance in rats fed MCD or HCD with an associated increase in serum parathyroid hormone (PTH) levels. WF did not alter bone mineral density or bone mineral content of the tibia in all the rats fed with different levels of dietary Ca. In conclusion, WF was responsible for the positive actions of FLL on Ca absorption and balance. The regulation of Ca balance by WF might involve its action in stimulating PTH production in the mature female rats.
Collapse
|
43
|
Fitzpatrick LA, Smith PL, McBride TA, Fries MA, Hossain M, Dabrowski CE, Gordon DN. Ronacaleret, a calcium-sensing receptor antagonist, has no significant effect on radial fracture healing time: results of a randomized, double-blinded, placebo-controlled Phase II clinical trial. Bone 2011; 49:845-52. [PMID: 21742071 DOI: 10.1016/j.bone.2011.06.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Revised: 06/07/2011] [Accepted: 06/16/2011] [Indexed: 11/28/2022]
Abstract
BACKGROUND Fractures cause significant morbidity, mortality, and use of health care resources. An oral agent that enhances fracture healing could reduce costs and prevent future disabilities. In Phase I studies, ronacaleret, a novel calcium-sensing receptor antagonist, stimulated parathyroid hormone (PTH) release and increased bone formation markers, suggesting that it may act as an effective oral anabolic agent to enhance fracture healing. METHODS This was a randomized, double-blind, placebo-controlled, parallel-group, clinical trial in 85 male and female subjects who had sustained a closed, unilateral, extra-articular fracture of the distal radius and were receiving conservative treatment. Subjects were randomly assigned in a double-blind manner to ronacaleret 200 mg twice daily, ronacaleret 400 mg once daily or matching placebo and followed for 12 weeks. Fracture healing was assessed by radiographs and quantitative computed tomography (CT), and bone turnover markers were measured. The study was terminated early for futility based on the results of an unplanned interim analysis. RESULTS There were no significant differences between treatment groups in time to radiographic fracture healing (74, 65 and 68 days for placebo, 200 mg twice daily and 400 mg once daily dose groups, respectively), cortical bridging, grip strength, pain and swelling, time to cast removal, or range of motion. Markers of bone formation and levels of whole PTH, intact PTH and serum calcium increased following treatment with ronacaleret. CONCLUSION Ronacaleret had no significant effect on duration of healing by radiograph or CT scan, time to cast removal, clinical symptoms, grip strength, or range of motion.
Collapse
|
44
|
Calcilytics: antagonists of the calcium-sensing receptor for the treatment of osteoporosis. Future Med Chem 2011; 3:535-47. [PMID: 21526895 DOI: 10.4155/fmc.11.17] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The only bone anabolic agents currently available on the market are based on the parathyroid hormone (PTH). Secretion of endogenous PTH is controlled by a calcium-sensing receptor at the surface of the parathyroid glands. Antagonists of this receptor (calcilytics) induce the release of the hormone. Provided the effect of the calcilytic is of short duration, a bone anabolic effect should also result. Although the first calcilytic series became known approximately 10 years ago, the number of different structural types is still small today. This article outlines the quest from hits to potent development candidates of all relevant calcilytic series currently known. Even after the front-runners unexpectedly failed in the clinic, the approach for an oral alternative to parenteral PTH remains highly attractive.
Collapse
|
45
|
John MR, Widler L, Gamse R, Buhl T, Seuwen K, Breitenstein W, Bruin GJM, Belleli R, Klickstein LB, Kneissel M. ATF936, a novel oral calcilytic, increases bone mineral density in rats and transiently releases parathyroid hormone in humans. Bone 2011; 49:233-41. [PMID: 21514409 DOI: 10.1016/j.bone.2011.04.007] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2010] [Revised: 04/01/2011] [Accepted: 04/06/2011] [Indexed: 01/28/2023]
Abstract
Parathyroid hormone (PTH), when injected daily as either the intact hormone PTH(1-84) or the active fragment PTH(1-34) (teriparatide), is an efficacious bone anabolic treatment option for osteoporosis patients. Injections lead to rapid and transient spikes in hormone exposure levels, a profile which is a prerequisite to effectively form bone. Oral antagonists of the calcium-sensing receptor (calcilytics) stimulate PTH secretion and represent thus an alternative approach to elevate hormone levels transiently. We report here on ATF936, a novel calcilytic, which triggered rapid, transient spikes in endogenous PTH levels when given orally in single doses of 10 and 30mg/kg to growing rats, and of 1mg/kg to dogs. Eight weeks daily oral application of 30mg/kg of ATF936 to aged female rats induced in the proximal tibia metaphysis increases in bone mineral density, cancellous bone volume and cortical and trabecular thickness as evaluated by computed tomography. In healthy humans, single oral doses of ATF936 produced peak PTH levels in plasma after a median time of 1h and levels returned to normal at 24-h post-dose. The average maximum PTH concentration increase from baseline was 1.9, 3.6, and 6.0-fold at doses of 40, 70, and 140mg. ATF936 was well tolerated. The sharp, transient increase in PTH levels produced by the oral calcilytic ATF936 was comparable to the PTH profile observed after subcutaneous administration of teriparatide. In conclusion, ATF936 might hold potential as an oral bone-forming osteoporosis therapy.
Collapse
|
46
|
Kimura S, Nakagawa T, Matsuo Y, Ishida Y, Okamoto Y, Hayashi M. JTT-305, an orally active calcium-sensing receptor antagonist, stimulates transient parathyroid hormone release and bone formation in ovariectomized rats. Eur J Pharmacol 2011; 668:331-6. [PMID: 21810422 DOI: 10.1016/j.ejphar.2011.07.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Revised: 06/23/2011] [Accepted: 07/07/2011] [Indexed: 10/17/2022]
Abstract
Intermittent administration of parathyroid hormone (PTH) has a potent anabolic effect on bone in humans and animals. Calcium-sensing receptor (CaSR) antagonists stimulate endogenous PTH secretion through CaSR on the surface of parathyroid cells and thereby may be anabolic agents for osteoporosis. JTT-305 is a potent oral short-acting CaSR antagonist and transiently stimulates endogenous PTH secretion. The objective of the present study was to investigate the effects of JTT-305 on PTH secretion and bone in ovariectomized rats. Female rats, immediately after ovariectomy (OVX), were orally administered vehicle or JTT-305 (0.3, 1, or 3 mg/kg) for 12 weeks. The serum PTH concentrations were transiently elevated with increasing doses of JTT-305. In the proximal tibia, JTT-305 prevented OVX-induced decreases in both the cancellous and total bone mineral density (BMD) except for the 0.3mg/kg dose. At the 3mg/kg dose, JTT-305 increased the mineralizing surface and bone formation rate in histomorphometry. The efficacy of JTT-305 at the 3mg/kg dose on the BMD corresponded to that of exogenous rat PTH1-84 injection at doses between 3 and 10 μg/kg. In conclusion, JTT-305 stimulated endogenous transient PTH secretion and bone formation, and consequently prevented bone loss in OVX rats. These results suggest that JTT-305 is orally active and has the potential to be an anabolic agent for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Shuichi Kimura
- Biological/Pharmaceutical Research Laboratories, Central Pharmaceutical Research Institute, Japan Tobacco Inc., Osaka, Japan.
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
Osteoporosis is a common disease characterised by a systemic impairment of bone mass and microarchitecture that results in fragility fractures. With an ageing population, the medical and socioeconomic effect of osteoporosis, particularly postmenopausal osteoporosis, will increase further. A detailed knowledge of bone biology with molecular insights into the communication between bone-forming osteoblasts and bone-resorbing osteoclasts and the orchestrating signalling network has led to the identification of novel therapeutic targets. Novel treatment strategies have been developed that aim to inhibit excessive bone resorption and increase bone formation. The most promising novel treatments include: denosumab, a monoclonal antibody for receptor activator of NF-κB ligand, a key osteoclast cytokine; odanacatib, a specific inhibitor of the osteoclast protease cathepsin K; and antibodies against the proteins sclerostin and dickkopf-1, two endogenous inhibitors of bone formation. This overview discusses these novel therapies and explains their underlying physiology.
Collapse
Affiliation(s)
- Tilman D. Rachner
- Division of Endocrinology, Diabetes, and Bone Diseases, Dresden Technical University Medical Center, Dresden, Germany
| | | | - Lorenz C. Hofbauer
- Division of Endocrinology, Diabetes, and Bone Diseases, Dresden Technical University Medical Center, Dresden, Germany
- Center for Regenerative Therapies Dresden, Germany
| |
Collapse
|
48
|
Abraham AK, Maurer TS, Kalgutkar AS, Gao X, Li M, Healy DR, Petersen DN, Griffith DA, Mager DE. Pharmacodynamic model of parathyroid hormone modulation by a negative allosteric modulator of the calcium-sensing receptor. AAPS JOURNAL 2011; 13:265-73. [PMID: 21437757 DOI: 10.1208/s12248-011-9266-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2010] [Accepted: 02/22/2011] [Indexed: 11/30/2022]
Abstract
In this study, a pharmacodynamic model is developed, based on calcium-parathyroid hormone (PTH) homeostasis, which describes the concentration-effect relationship of a negative allosteric modulator of the calcium-sensing receptor (CaR) in rats. Plasma concentrations of drug and PTH were determined from plasma samples obtained via serial jugular vein sampling following single subcutaneous doses of 1, 5, 45, and 150 mg/kg to male Sprague-Dawley rats (n = 5/dose). Drug pharmacokinetics was described by a one-compartment model with first-order absorption and linear elimination. Concentration-time profiles of PTH were characterized using a model in which the compound allosterically modulates Ca(+2) binding to the CaR that, in turn, modulates PTH through a precursor-pool indirect response model. Additionally, negative feedback was incorporated to account for tolerance observed at higher dose levels. Model fitting and parameter estimation were conducted using the maximum likelihood algorithm. The proposed model well characterized the data and provided compound specific estimates of the K(i) and cooperativity constant (α) of 1.47 ng/mL and 0.406, respectively. In addition, the estimated model parameters for PTH turnover were comparable to that previously reported. The final generalized model is capable of characterizing both PTH-Ca(+2) homeostasis and the pharmacokinetics and pharmacodynamics associated with the negative allosteric CaR modulator. As such, the model provides a simple platform for analysis of drugs targeting the PTH-Ca(+2) system.
Collapse
Affiliation(s)
- Anson K Abraham
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Amherst, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Novel and potent calcium-sensing receptor antagonists: discovery of (5R)-N-[1-ethyl-1-(4-ethylphenyl)propyl]-2,7,7-trimethyl-5-phenyl-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide monotosylate (TAK-075) as an orally active bone anabolic agent. Bioorg Med Chem 2011; 19:1881-94. [PMID: 21353570 DOI: 10.1016/j.bmc.2011.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2011] [Revised: 01/31/2011] [Accepted: 02/01/2011] [Indexed: 11/22/2022]
Abstract
The calcium-sensing receptor antagonist (CaSR) has been recognized as a promising target of anabolic agents for treating osteoporosis. In the course of developing a new drug candidate for osteoporosis, we found tetrahydropyrazolopyrimidine derivative 1 to be an orally active CaSR antagonist that stimulated transient PTH secretion in rats. However, compound 1 showed poor physical and chemical stability. In order to work out this compound's chemical stability and further understand its in vivo efficacy, we focused on modifying the 2-position of the tetrahydropyrazolopyrimidine. As a result of chemical modification, we discovered (5R)-N-[1-ethyl-1-(4-ethylphenyl)propyl]-2,7,7-trimethyl-5-phenyl-4,5,6,7-tetrahydropyrazolo[1,5-a]pyrimidine-3-carboxamide monotosylate 10m (TAK-075), which showed improved solubility, chemical stability, and in vivo efficacy. Furthermore, we describe that evaluating the active metabolite is important during repeated treatment with short-acting CaSR antagonists.
Collapse
|
50
|
Yoshida M, Mori A, Kotani E, Oka M, Makino H, Fujita H, Ban J, Ikeda Y, Kawamoto T, Goto M, Kimura H, Baba A, Yasuma T. Discovery of Novel and Potent Orally Active Calcium-Sensing Receptor Antagonists that Stimulate Pulselike Parathyroid Hormone Secretion: Synthesis and Structure−Activity Relationships of Tetrahydropyrazolopyrimidine Derivatives. J Med Chem 2011; 54:1430-40. [DOI: 10.1021/jm101452x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Masato Yoshida
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 17-85 Jusohonmachi-2-chome, Yodogawa-ku, Osaka 532-8686, Japan
| | - Akira Mori
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 17-85 Jusohonmachi-2-chome, Yodogawa-ku, Osaka 532-8686, Japan
| | - Etsuo Kotani
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 17-85 Jusohonmachi-2-chome, Yodogawa-ku, Osaka 532-8686, Japan
| | - Masahiro Oka
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 17-85 Jusohonmachi-2-chome, Yodogawa-ku, Osaka 532-8686, Japan
| | - Haruhiko Makino
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 17-85 Jusohonmachi-2-chome, Yodogawa-ku, Osaka 532-8686, Japan
| | - Hisashi Fujita
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 17-85 Jusohonmachi-2-chome, Yodogawa-ku, Osaka 532-8686, Japan
| | - Junko Ban
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 17-85 Jusohonmachi-2-chome, Yodogawa-ku, Osaka 532-8686, Japan
| | - Yukihiro Ikeda
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 17-85 Jusohonmachi-2-chome, Yodogawa-ku, Osaka 532-8686, Japan
| | - Tomohiro Kawamoto
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 17-85 Jusohonmachi-2-chome, Yodogawa-ku, Osaka 532-8686, Japan
| | - Mika Goto
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 17-85 Jusohonmachi-2-chome, Yodogawa-ku, Osaka 532-8686, Japan
| | - Hiroyuki Kimura
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 17-85 Jusohonmachi-2-chome, Yodogawa-ku, Osaka 532-8686, Japan
| | - Atsuo Baba
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 17-85 Jusohonmachi-2-chome, Yodogawa-ku, Osaka 532-8686, Japan
| | - Tsuneo Yasuma
- Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, 17-85 Jusohonmachi-2-chome, Yodogawa-ku, Osaka 532-8686, Japan
| |
Collapse
|