1
|
Albert AP, Greenberg HZ. Role of the calcium-sensing receptor in regulating vascular function. J Cell Commun Signal 2025; 19:e70004. [PMID: 39912052 PMCID: PMC11798592 DOI: 10.1002/ccs3.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/26/2024] [Accepted: 01/20/2025] [Indexed: 02/07/2025] Open
Abstract
Functional expression of the calcium-sensing receptor (CaSR) in calcitropic tissues, for example, parathyroid glands and kidneys, is important for maintaining Ca2+ homeostasis. It is also established that the CaSR is present in tissues previously thought to be noncalcitropic and this review discusses the role of the CaSR in vascular function, focusing mainly on contractility but also outlining its role in cell proliferation and calcification. Stimulation of the CaSR by extracellular Ca2+ concentration ([Ca2+]o) on perivascular sensory nerves and vascular endothelial cells is associated with vasodilatation through the release of vasoactive substances and stimulation of IKCa channels and nitric oxide synthesis, respectively, which mediate endothelium-derived hyperpolarizations and activation of BKCa channels and KATP channels in vascular smooth muscle cells (VSMCs). CaSR-induced vasoconstrictions are mediated by the CaSR expressed in VSMCs, which are coupled to the Gq/11 protein-coupled pathway. In addition, the CaSR expressed on VSMCs also regulates proliferation and calcification. Consequently, the CaSR has been implicated in regulating systemic and pulmonary blood pressure and calcimimetics and calcilytics are potential therapeutic targets for cardiovascular diseases, such as hypertension, pulmonary artery hypertension, and atherosclerosis.
Collapse
Affiliation(s)
- Anthony P. Albert
- Vascular Biology SectionCardiovascular & Genomics Research InstituteSt. George's, University of LondonLondonUK
- Norwich Medical SchoolUniversity of East AngliaNorfolkUK
| | - Harry Z.E. Greenberg
- Vascular Biology SectionCardiovascular & Genomics Research InstituteSt. George's, University of LondonLondonUK
| |
Collapse
|
2
|
The Calcilytic Drug Calhex-231 Ameliorates Vascular Hyporesponsiveness in Traumatic Hemorrhagic Shock by Inhibiting Oxidative Stress and miR-208a-Mediated Mitochondrial Fission. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4132785. [PMID: 33343806 PMCID: PMC7732383 DOI: 10.1155/2020/4132785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/27/2020] [Accepted: 11/26/2020] [Indexed: 12/29/2022]
Abstract
Background The calcium-sensing receptor (CaSR) plays a fundamental role in extracellular calcium homeostasis in humans. Surprisingly, CaSR is also expressed in nonhomeostatic tissues and is involved in regulating diverse cellular functions. The objective of this study was to determine if Calhex-231 (Cal), a negative modulator of CaSR, may be beneficial in the treatment of traumatic hemorrhagic shock (THS) by improving cardiovascular function and investigated the mechanisms. Methods Rats that had been subjected to THS and hypoxia-treated vascular smooth muscle cells (VSMCs) were used in this study. The effects of Cal on cardiovascular function, animal survival, hemodynamics, and vital organ function in THS rats and the relationship to oxidative stress, mitochondrial fusion-fission, and microRNA (miR-208a) were investigated. Results Cal significantly improved hemodynamics, elevated blood pressure, increased vital organ blood perfusion and local oxygen supply, and markedly improved the survival outcomes of THS rats. Furthermore, Cal significantly improved vascular reactivity after THS in vivo and in vitro. Cal also restored the THS-induced decrease in myosin light chain (MLC) phosphorylation (the key element for VSMC contraction). Inhibition of MLC phosphorylation antagonized the Cal-induced restoration of vascular reactivity following THS. Cal suppressed oxidative stress in THS rats and hypoxic-VSMCs. Meanwhile, THS induced expression of mitochondrial fission proteins Drp1 and Fis1 and decreased expression of mitochondrial fusion protein Mfn1 in vascular tissues. Cal reduced expression of Drp1 and Fis1. In hypoxic-VSMCs, Cal inhibited mitochondrial fragmentation and preserved mitochondrial morphology. In addition, miR-208a mimic decreased Fis1 expression, and miR-208a inhibitor prevented Cal-induced Fis1 downregulation in hypoxic-VSMCs. Conclusion Calhex-231 exhibits outstanding potential for effective therapy of traumatic hemorrhagic shock, and the beneficial effects result from its protection of vascular function via inhibition of oxidative stress and miR-208a-mediated mitochondrial fission.
Collapse
|
3
|
Poulin A, Bellemare PL, Fortier C, Mac-Way F, Desmeules S, Marquis K, Gaudreault V, Lebel M, Agharazii M. Acute effects of cinacalcet on arterial stiffness and ventricular function in hemodialysis patients: A randomized double-blinded crossover study. Medicine (Baltimore) 2017; 96:e6912. [PMID: 28538380 PMCID: PMC5457860 DOI: 10.1097/md.0000000000006912] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Serum calcium concentration (Ca) plays an essential role in a vascular muscle tone and myocardial contractility. Previously, we showed that acutely lowering Ca by hemodialysis reduced arterial stiffness. Cinacalcet is a calcimimetic that lowers Ca and parathyroid hormone (PTH). The aim of the present study was to examine whether acute lowering of Ca by cinacalcet improves vascular stiffness and myocardial diastolic dysfunction. METHOD This is a double-blinded randomized placebo-controlled crossover study that included 21 adult patients with end-stage kidney disease undergoing chronic hemodialysis. Subjects were assigned to placebo-cinacalcet (30 mg) or cinacalcet-placebo sequence. After each treatment period (7 days), aortic, brachial, and carotid stiffness were determined by examining carotid-femoral pulse wave velocity (cf-PWV), carotid-radial PWV (cr-PWV), and carotid distension. A central pulse wave profile was determined by radial artery tonometry and cardiac function was evaluated by echocardiography. RESULTS Cinacalcet reduced PTH (483 [337-748] to 201 [71-498] ng/L, P < .001) and ionized Ca (1.11 [1.08-1.15] to 1.05 [1.00-1.10] mmol/L, P = .04). Cinacalcet did not reduced cf-PWV significantly (12.2 [10.4-15.4] to 12.2 [11.0-14.6] m/s, P = .16). After adjustments for mean blood pressure, sequence, carryover, and treatment effects, cf-PWV was not significantly lowered by cinacalcet (-0.35 m/s, P = .139). There were no significant changes in central blood pressures, brachial and carotid stiffness, and echocardiographic parameters. CONCLUSION In this study, 30 mg daily cinacalcet for 1 week did not have any significant impact on peripheral and central blood pressures, arterial stiffness parameters, or cardiac function (NCT01250405).
Collapse
Affiliation(s)
- Aurélie Poulin
- CHU de Québec Research Center, L’Hôtel-Dieu de Québec Hospital
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Pierre-Luc Bellemare
- CHU de Québec Research Center, L’Hôtel-Dieu de Québec Hospital
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Catherine Fortier
- CHU de Québec Research Center, L’Hôtel-Dieu de Québec Hospital
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Fabrice Mac-Way
- CHU de Québec Research Center, L’Hôtel-Dieu de Québec Hospital
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Simon Desmeules
- CHU de Québec Research Center, L’Hôtel-Dieu de Québec Hospital
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Karine Marquis
- CHU de Québec Research Center, L’Hôtel-Dieu de Québec Hospital
| | - Valérie Gaudreault
- CHU de Québec Research Center, L’Hôtel-Dieu de Québec Hospital
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Marcel Lebel
- CHU de Québec Research Center, L’Hôtel-Dieu de Québec Hospital
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Mohsen Agharazii
- CHU de Québec Research Center, L’Hôtel-Dieu de Québec Hospital
- Department of Medicine, Faculty of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
4
|
Billington EO, Bristow SM, Gamble GD, de Kwant JA, Stewart A, Mihov BV, Horne AM, Reid IR. Acute effects of calcium supplements on blood pressure: randomised, crossover trial in postmenopausal women. Osteoporos Int 2017; 28:119-125. [PMID: 27543500 DOI: 10.1007/s00198-016-3744-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/12/2016] [Indexed: 12/17/2022]
Abstract
UNLABELLED Calcium supplements appear to increase cardiovascular risk, but the mechanism is unknown. We investigated the acute effects of calcium supplements on blood pressure in postmenopausal women. The reduction in systolic blood pressure was smaller after calcium compared with the placebo in the hours following dosing. INTRODUCTION Calcium supplements appear to be associated with increased cardiovascular risk; however, the mechanism of this is uncertain. We previously reported that blood pressure declined over a day in older women, and that this reduction was smaller following a calcium supplement. To confirm this finding, we investigated the acute effects of calcium supplements on blood pressure. METHODS This was a randomised controlled crossover trial in 40 healthy postmenopausal women (mean age 71 years and BMI 27.2 kg/m2). Women attended on two occasions, with visits separated by ≥7 days. At each visit, they received either 1 g of calcium as citrate, or placebo. Blood pressure and serum calcium concentrations were measured immediately before, and 2, 4 and 6 h after each intervention. RESULTS Ionised and total calcium concentrations increased after calcium (p < 0.0001 versus placebo). Systolic blood pressure decreased after both calcium and placebo, but significantly less so after calcium (p = 0.02). The reduction in systolic blood pressure from baseline was smaller after calcium compared with placebo by 6 mmHg at 4 h (p = 0.036) and by 9 mmHg at 6 h (p = 0.002). The reduction in diastolic blood pressure was similar after calcium and placebo. CONCLUSIONS These findings are consistent with those of our previous trial and indicate that the use of calcium supplements in postmenopausal women attenuates the post-breakfast reduction in systolic blood pressure by around 6-9 mmHg. Whether these changes in blood pressure influence cardiovascular risk requires further study.
Collapse
Affiliation(s)
- E O Billington
- Department of Medicine, The University of Auckland, Private Bag 92019, Auckland Mail Centre 1142, Auckland, New Zealand
- Department of Medicine, University of Calgary, Calgary, Canada
| | - S M Bristow
- Department of Medicine, The University of Auckland, Private Bag 92019, Auckland Mail Centre 1142, Auckland, New Zealand.
| | - G D Gamble
- Department of Medicine, The University of Auckland, Private Bag 92019, Auckland Mail Centre 1142, Auckland, New Zealand
| | - J A de Kwant
- Department of Medicine, The University of Auckland, Private Bag 92019, Auckland Mail Centre 1142, Auckland, New Zealand
| | - A Stewart
- Department of Medicine, The University of Auckland, Private Bag 92019, Auckland Mail Centre 1142, Auckland, New Zealand
| | - B V Mihov
- Department of Medicine, The University of Auckland, Private Bag 92019, Auckland Mail Centre 1142, Auckland, New Zealand
| | - A M Horne
- Department of Medicine, The University of Auckland, Private Bag 92019, Auckland Mail Centre 1142, Auckland, New Zealand
| | - I R Reid
- Department of Medicine, The University of Auckland, Private Bag 92019, Auckland Mail Centre 1142, Auckland, New Zealand
| |
Collapse
|
5
|
Reid IR, Bristow SM, Bolland MJ. Cardiovascular complications of calcium supplements. J Cell Biochem 2016; 116:494-501. [PMID: 25491763 DOI: 10.1002/jcb.25028] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 12/04/2014] [Indexed: 12/26/2022]
Abstract
There is longstanding concern that calcium supplements might increase cardiovascular risk in patients with renal impairment. The Auckland Calcium Study suggested that the same problem occurs in older people taking these supplements for prevention of osteoporosis. Our subsequent meta-analyses, (which followed protocols finalized before the data was available) confirmed that calcium supplements, with or without vitamin D, adversely affected risk of myocardial infarction and, possibly, stroke. Several groups have re-visited these data, consistently finding an adverse effect of calcium on myocardial infarction, not always statistically significant because some meta-analyses have been under-powered. Whether or not an adverse effect of calcium plus vitamin D on myocardial infarction is found depends on whether two specific groups of subjects are included-those in the Women's Health Initiative who were already taking calcium at the time of randomization, and subjects from an open, cluster-randomized study in which baseline cardiovascular risk was different between groups. Vitamin D alone does not affect vascular risk, so it is unlikely that differences between calcium alone and calcium plus vitamin D are real, and they are more likely to result from the inclusion of studies at high risk of bias. The mechanisms of the adverse cardiovascular effects are uncertain but may be mediated by the increase in serum calcium following supplement ingestion, and the effects of this on vascular function and coagulation. Available evidence suggests the risks of calcium supplements outweigh any small benefits on fracture incidence, so the case for their use is weak.
Collapse
Affiliation(s)
- Ian R Reid
- University of Auckland, Auckland, New Zealand; Department of Endocrinology, Auckland District Health Board, New Zealand
| | | | | |
Collapse
|
6
|
Acute effects of calcium supplements on blood pressure and blood coagulation: secondary analysis of a randomised controlled trial in post-menopausal women. Br J Nutr 2015; 114:1868-74. [DOI: 10.1017/s0007114515003694] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
AbstractRecent evidence suggests that Ca supplements increase the risk of cardiovascular events, but the mechanism(s) by which this occurs is uncertain. In a study primarily assessing the effects of various Ca supplements on blood Ca levels, we also investigated the effects of Ca supplements on blood pressure and their acute effects on blood coagulation. We randomised 100 post-menopausal women to 1 g/d of Ca or a placebo containing no Ca. Blood pressure was measured at baseline and every 2 h up to 8 h after their first dose and after 3 months of supplementation. Blood coagulation was measured by thromboelastography (TEG) in a subgroup of participants (n 40) up to 8 h only. Blood pressure declined over 8 h in both the groups, consistent with its normal diurnal rhythm. The reduction in systolic blood pressure was smaller in the Ca group compared with the control group by >5 mmHg between 2 and 6 h (P≤0·02), and the reduction in diastolic blood pressure was smaller at 2 h (between-groups difference 4·5 mmHg, P=0·004). Blood coagulability, assessed by TEG, increased from baseline over 8 h in the calcium citrate and control groups. At 4 h, the increase in the coagulation index was greater in the calcium citrate group compared with the control group (P=0·03), which appeared to be due to a greater reduction in the time to clot initiation. These data suggest that Ca supplements may acutely influence blood pressure and blood coagulation. Further investigation of this possibility is required.
Collapse
|
7
|
Yamamura A, Ohara N, Tsukamoto K. Inhibition of Excessive Cell Proliferation by Calcilytics in Idiopathic Pulmonary Arterial Hypertension. PLoS One 2015; 10:e0138384. [PMID: 26375676 PMCID: PMC4574199 DOI: 10.1371/journal.pone.0138384] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 08/28/2015] [Indexed: 11/19/2022] Open
Abstract
Idiopathic pulmonary arterial hypertension (IPAH) is a rare and progressive disease of unknown pathogenesis. Vascular remodeling due to excessive proliferation of pulmonary arterial smooth muscle cells (PASMCs) is a critical pathogenic event that leads to early morbidity and mortality. The excessive cell proliferation is closely linked to the augmented Ca2+ signaling in PASMCs. More recently, we have shown by an siRNA knockdown method that the Ca2+-sensing receptor (CaSR) is upregulated in PASMCs from IPAH patients, involved in the enhanced Ca2+ response and subsequent excessive cell proliferation. In this study, we examined whether pharmacological blockade of CaSR attenuated the excessive proliferation of PASMCs from IPAH patients by MTT assay. The proliferation rate of PASMCs from IPAH patients was much higher (~1.5-fold) than that of PASMCs from normal subjects and patients with chronic thromboembolic pulmonary hypertension (CTEPH). Treatment with NPS2143, an antagonist of CaSR or calcilytic, clearly suppressed the cell proliferation in a concentration-dependent manner (IC50 = 2.64 μM) in IPAH-PASMCs, but not in normal and CTEPH PASMCs. Another calcilytic, Calhex 231, which is structurally unrelated to NPS2143, also concentration-dependently inhibited the excessive proliferation of IPAH-PASMCs (IC50 = 1.89 μM). In contrast, R568, an activator of CaSR or calcimimetic, significantly facilitated the proliferation of IPAH-PASMCs (EC50 = 0.33 μM). Similar results were obtained by BrdU incorporation assay. These results reveal that the excessive PASMC proliferation was modulated by pharmacological tools of CaSR, showing us that calcilytics are useful for a novel therapeutic approach for pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Aya Yamamura
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan
- * E-mail:
| | - Naoki Ohara
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan
| | - Kikuo Tsukamoto
- Department of Pharmacy, College of Pharmacy, Kinjo Gakuin University, Nagoya, Japan
| |
Collapse
|
8
|
Martínez-Miguel P, Medrano-Andrés D, Lopes-Martín V, Arribas-Gómez I, Rodríguez-Puyol M, Rodríguez-Puyol D, López-Ongil S. Regulation of endothelin-converting enzyme-1 (ECE-1) by the calcimimetic R-568. Pharmacol Res 2013; 76:106-18. [DOI: 10.1016/j.phrs.2013.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 07/17/2013] [Accepted: 07/24/2013] [Indexed: 11/28/2022]
|
9
|
Sikjaer T, Amstrup AK, Rolighed L, Kjaer SG, Mosekilde L, Rejnmark L. PTH(1-84) replacement therapy in hypoparathyroidism: a randomized controlled trial on pharmacokinetic and dynamic effects after 6 months of treatment. J Bone Miner Res 2013; 28:2232-43. [PMID: 23649554 DOI: 10.1002/jbmr.1964] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 04/04/2013] [Accepted: 04/10/2013] [Indexed: 12/27/2022]
Abstract
Untreated, hypoparathyroidism (hypoPT) is a state of hypocalcemia with inappropriately low plasma parathyroid hormone (PTH) levels and hyperphosphatemia. PTH administration normalizes plasma calcium and phosphate levels and reduces the doses of calcium and active vitamin D analogues needed. To develop an evidence-based clinical algorithm to monitor hypoPT patients treated with recombinant human PTH (rhPTH[1-84]) injected subcutaneously in the thigh, we performed a 24-hour monitoring study of pharmacokinetic and pharmacodynamic effects in a group of 38 patients who had completed a 6-month randomized study on effects of treatment with a fixed rhPTH(1-84) dose of 100 µg/d or similar placebo as an add-on to conventional treatment. PTH levels rose immediately, reaching a median peak level of 26.5 (interquartile range [IQR], 20.1-42.5) pmol/L 15 minutes following injection. Thereafter, levels gradually decreased until reaching predosing levels after 16 hours, with a plasma half-life of 2.2 (1.7-2.5) hours. rhPTH(1-84) changed the diurnal rhythms of ionized calcium levels and 1,25-dihydroxyvitamin D (1,25[OH]2 D) levels, with rising levels following injection. Ionized calcium peaked after 7.0 (5.0-10.0) hours. Asymptomatic hypercalcemia was present in 71% of the rhPTH(1-84)-treated patients. Compared with placebo, 24-hour urinary calcium, phosphate, and magnesium did not change, although the diurnal variation in renal excretion rates changed significantly in response to treatment. In conclusion, as a safety precaution, we recommend occasionally measuring calcium levels at approximately 7 hours after administration in order to reveal episodes of hypercalcemia. A 100-µg daily dose of rhPTH(1-84) appears to be too high in some patients, suggesting a need for a device allowing for individual dose adjustments.
Collapse
Affiliation(s)
- Tanja Sikjaer
- Department of Endocrinology and Internal Medicine, MEA, Aarhus University Hospital, Aarhus, Denmark
| | | | | | | | | | | |
Collapse
|
10
|
Moor MB, Kruse A, Uehlinger DE, Eisenberger U. Arterial Stiffness Depends on Serum Ionized Calcium Levels During Dialysis With Regional Citrate Anticoagulation. Artif Organs 2013; 37:467-74. [DOI: 10.1111/aor.12037] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Matthias B. Moor
- Department of Nephrology and Hypertension; University Hospital of Bern; Bern; Switzerland
| | - Anja Kruse
- Department of Nephrology and Hypertension; University Hospital of Bern; Bern; Switzerland
| | - Dominik E. Uehlinger
- Department of Nephrology and Hypertension; University Hospital of Bern; Bern; Switzerland
| | | |
Collapse
|
11
|
Yamamura A, Yamamura H, Guo Q, Zimnicka AM, Wan J, Ko EA, Smith KA, Pohl NM, Song S, Zeifman A, Makino A, Yuan JXJ. Dihydropyridine Ca(2+) channel blockers increase cytosolic [Ca(2+)] by activating Ca(2+)-sensing receptors in pulmonary arterial smooth muscle cells. Circ Res 2013; 112:640-50. [PMID: 23300272 DOI: 10.1161/circresaha.113.300897] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE An increase in cytosolic free Ca(2+) concentration ([Ca(2+)](cyt)) in pulmonary arterial smooth muscle cells (PASMC) is a major trigger for pulmonary vasoconstriction and an important stimulus for PASMC proliferation and pulmonary vascular remodeling. The dihydropyridine Ca(2+) channel blockers, such as nifedipine, have been used for treatment of idiopathic pulmonary arterial hypertension (IPAH). OBJECTIVE Our previous study demonstrated that the Ca(2+)-sensing receptor (CaSR) was upregulated and the extracellular Ca(2+)-induced increase in [Ca(2+)](cyt) was enhanced in PASMC from patients with IPAH and animals with experimental pulmonary hypertension. Here, we report that the dihydropyridines (eg, nifedipine) increase [Ca(2+)](cyt) by activating CaSR in PASMC from IPAH patients (in which CaSR is upregulated), but not in normal PASMC. METHODS AND RESULTS The nifedipine-mediated increase in [Ca(2+)](cyt) in IPAH-PASMC was concentration dependent with a half maximal effective concentration of 0.20 µmol/L. Knockdown of CaSR with siRNA in IPAH-PASMC significantly inhibited the nifedipine-induced increase in [Ca(2+)](cyt), whereas overexpression of CaSR in normal PASMC conferred the nifedipine-induced rise in [Ca(2+)](cyt). Other dihydropyridines, nicardipine and Bay K8644, had similar augmenting effects on the CaSR-mediated increase in [Ca(2+)](cyt) in IPAH-PASMC; however, the nondihydropyridine blockers, such as diltiazem and verapamil, had no effect on the CaSR-mediated rise in [Ca(2+)](cyt). CONCLUSIONS The dihydropyridine derivatives increase [Ca(2+)](cyt) by potentiating the activity of CaSR in PASMC independently of their blocking (or activating) effect on Ca(2+) channels; therefore, it is possible that the use of dihydropyridine Ca(2+) channel blockers (eg, nifedipine) to treat IPAH patients with upregulated CaSR in PASMC may exacerbate pulmonary hypertension.
Collapse
Affiliation(s)
- Aya Yamamura
- Department of Medicine, Section of Pulmonary, Critical Care, Sleep, and Allergy Medicine and Department of Pharmacology, Institute for Personalized Respiratory Medicine, Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Yamamura A, Guo Q, Yamamura H, Zimnicka AM, Pohl NM, Smith KA, Fernandez RA, Zeifman A, Makino A, Dong H, Yuan JXJ. Enhanced Ca(2+)-sensing receptor function in idiopathic pulmonary arterial hypertension. Circ Res 2012; 111:469-81. [PMID: 22730443 DOI: 10.1161/circresaha.112.266361] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
RATIONALE A rise in cytosolic Ca(2+) concentration ([Ca(2+)](cyt)) in pulmonary arterial smooth muscle cells (PASMC) is an important stimulus for pulmonary vasoconstriction and vascular remodeling. Increased resting [Ca(2+)](cyt) and enhanced Ca(2+) influx have been implicated in PASMC from patients with idiopathic pulmonary arterial hypertension (IPAH). OBJECTIVE We examined whether the extracellular Ca(2+)-sensing receptor (CaSR) is involved in the enhanced Ca(2+) influx and proliferation in IPAH-PASMC and whether blockade of CaSR inhibits experimental pulmonary hypertension. METHODS AND RESULTS In normal PASMC superfused with Ca(2+)-free solution, addition of 2.2 mmol/L Ca(2+) to the perfusate had little effect on [Ca(2+)](cyt). In IPAH-PASMC, however, restoration of extracellular Ca(2+) induced a significant increase in [Ca(2+)](cyt). Extracellular application of spermine also markedly raised [Ca(2+)](cyt) in IPAH-PASMC but not in normal PASMC. The calcimimetic R568 enhanced, whereas the calcilytic NPS 2143 attenuated, the extracellular Ca(2+)-induced [Ca(2+)](cyt) rise in IPAH-PASMC. Furthermore, the protein expression level of CaSR in IPAH-PASMC was greater than in normal PASMC; knockdown of CaSR in IPAH-PASMC with siRNA attenuated the extracellular Ca(2+)-mediated [Ca(2+)](cyt) increase and inhibited IPAH-PASMC proliferation. Using animal models of pulmonary hypertension, our data showed that CaSR expression and function were both enhanced in PASMC, whereas intraperitoneal injection of the calcilytic NPS 2143 prevented the development of pulmonary hypertension and right ventricular hypertrophy in rats injected with monocrotaline and mice exposed to hypoxia. CONCLUSIONS The extracellular Ca(2+)-induced increase in [Ca(2+)](cyt) due to upregulated CaSR is a novel pathogenic mechanism contributing to the augmented Ca(2+) influx and excessive PASMC proliferation in patients and animals with pulmonary arterial hypertension.
Collapse
Affiliation(s)
- Aya Yamamura
- Kinjo Gakuin University School of Pharmacy, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Torres PAU, De Broe M. Calcium-sensing receptor, calcimimetics, and cardiovascular calcifications in chronic kidney disease. Kidney Int 2012; 82:19-25. [PMID: 22437409 DOI: 10.1038/ki.2012.69] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Renal function impairment goes along with a disturbed calcium, phosphate, and vitamin D metabolism, resulting in secondary hyperparathyroidism (sHPT). These mineral metabolism disturbances are associated with soft tissue calcifications, particularly arteries, cardiac valves, and myocardium, ultimately associated with increased risk of mortality in patients with chronic kidney disease (CKD). sHPT may lead to cardiovascular calcifications by other mechanisms including an impaired effect of parathyroid hormone (PTH), and a decreased calcium-sensing receptor (CaR) expression on cardiovascular structures. PTH may play a direct role on vascular calcifications through activation of a receptor, the type-1 PTH/PTHrP receptor, normally attributed to PTH-related peptide (PTHrP). The CaR in vascular cells may also play a role on vascular mineralization as suggested by its extremely reduced expression in atherosclerotic calcified human arteries. Calcimimetic compounds increasing the CaR sensitivity to extracellular calcium efficiently reduce serum PTH, calcium, and phosphate in dialysis patients with sHPT. They upregulate the CaR in vascular cells and attenuate vascular mineralization in uremic states. In this article, the pathophysiological mechanisms associated with cardiovascular calcifications in case of sHPT, the impact of medical and surgical correction of sHPT, the biology of the CaR in vascular structures and its function in CKD state, and finally the role played by the CaR and its modulation by the calcimimetics on uremic-related cardiovascular calcifications are reviewed.
Collapse
|
14
|
Smajilovic S, Yano S, Jabbari R, Tfelt-Hansen J. The calcium-sensing receptor and calcimimetics in blood pressure modulation. Br J Pharmacol 2012; 164:884-93. [PMID: 21410453 DOI: 10.1111/j.1476-5381.2011.01317.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Calcium is a crucial second messenger in the cardiovascular system. However, calcium may also be an extracellular first messenger through a G-protein-coupled receptor that senses extracellular concentration (Ca(2+)(o)), the calcium-sensing receptor (CaR). The most prominent physiological function of the CaR is to maintain the extracellular Ca(2+) level in a very tight range by regulating the circulating levels of parathyroid hormone (PTH). This control over PTH and Ca(2+) levels is partially lost in patients suffering from primary and secondary hyperparathyroidism. Allosteric modulators of the CaR (calcimimetics) are the first drugs in their class to become available for clinical use and have been shown to successfully treat certain forms of primary and secondary hyperparathyroidism. In addition, several studies suggest beneficial effects of calcimimetics on cardiovascular risk factors associated with hyperparathyroidism. Although a plethora of studies demonstrated the CaR in heart and blood vessels, exact roles of the receptor in the cardiovascular system still remain to be elucidated. However, several studies point toward a possibility that the CaR might be involved in the regulation of vascular tone. This review will summarize the current knowledge on the possible functions of the CaR and calcimimetics on blood pressure regulation.
Collapse
Affiliation(s)
- Sanela Smajilovic
- Laboratory of Molecular Cardiology, Department of Cardiology, Copenhagen University Hospital, Denmark
| | | | | | | |
Collapse
|
15
|
Karohl C, Raggi P. Cinacalcet: will it play a role in reducing cardiovascular events? Future Cardiol 2012; 8:357-70. [PMID: 22420327 DOI: 10.2217/fca.11.82] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Secondary hyperparathyroidism is a common complication of chronic kidney disease and it is associated with high morbidity and mortality. It is characterized by high parathyroid hormone levels and bone turnover leading to bone pain, deformity and fragility. Furthermore, secondary hyperparathyroidism adversely affects the cardiovascular system and has been associated with cardiovascular calcification and cardiomyopathy. Cinacalcet, a type II calcimimetic, is an effective and well-tolerated oral therapy for the management of secondary hyperparathyroidism. It is an allosteric activator of the calcium-sensing receptor enhancing sensitivity of parathyroid cells to extracellular calcium, which leads to inhibition of parathyroid hormone secretion. The calcium-sensing receptor expression in cardiomyocytes, endothelial cells and vascular smooth muscle cells raises the possibility that this receptor may be implicated in the pathophysiology of cardiovascular disease and constitute a potential therapeutic target. This article reviews the role of the calcimimetic agent cinacalcet in the prevention and progression of cardiovascular calcification and uremic cardiomyopathy in the chronic kidney disease setting.
Collapse
Affiliation(s)
- Cristina Karohl
- Division of Cardiology & Department of Medicine, Emory University, 1365 Clifton Rd NE, AT-504, Atlanta, GA 30322, USA
| | | |
Collapse
|
16
|
Huang Y, Cavanaugh A, Breitwieser GE. Regulation of stability and trafficking of calcium-sensing receptors by pharmacologic chaperones. ADVANCES IN PHARMACOLOGY 2012; 62:143-73. [PMID: 21907909 DOI: 10.1016/b978-0-12-385952-5.00007-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Gain- or loss-of-function mutations and polymorphisms of the calcium-sensing receptor (CaSR) cause Ca(2+) handling diseases. Altered expression and/or signaling of wild-type CaSR can also contribute to pathology. Recent studies have demonstrated that a significant proportion of mutations cause altered targeting and/or trafficking of CaSR to the plasma membrane. Pharmacological approaches to rescue of CaSR function include treatment with allosteric modulators, which potentiate the effects of the orthosteric agonist Ca(2+). Dissection of the mechanism(s) contributing to allosteric agonist-mediated rescue of loss-of-function CaSR mutants has demonstrated pharmacologic chaperone actions coincident with CaSR biosynthesis. The distinctive responses to the allosteric agonist (NPS R-568), which promotes CaSR stability, and the allosteric antagonist (NPS 2143), which promotes CaSR degradation, have led to a model for a conformational checkpoint during CaSR biosynthesis. The conformational checkpoint would "tune" CaSR biosynthesis to cellular signaling state. Navigation of a distinct checkpoint for endoplasmic release can also be augmented by pharmacologic chaperones. The diverse, post-endoplasmic reticulum quality control site(s) for pharmacologic chaperone modulation of CaSR stability and trafficking redefines the role(s) of allosteric modulators in regulation of overall GPCR function.
Collapse
Affiliation(s)
- Ying Huang
- Cancer Drug Research Laboratory, McGill University, Royal Victoria Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
17
|
Weston AH, Geraghty A, Egner I, Edwards G. The vascular extracellular calcium-sensing receptor: an update. Acta Physiol (Oxf) 2011; 203:127-37. [PMID: 21199402 DOI: 10.1111/j.1748-1716.2010.02249.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The extracellular calcium-sensing receptor (CaR) was first described in the parathyroid gland. Recent studies have shown that the CaR is also expressed in blood vessels, especially in the endothelial and adventitial layers but its physiological function is still not clear. However, an understanding of its possible role(s) in the vasculature (perivascular-neurones, heart and blood vessels) is important because of the use of synthetic positive allosteric CaR modulators in hyperparathyroidism and the potential importance of negative modulators in the treatment of osteoporosis. In this review, the effects of CaR activation and inhibition are detailed and the possible role of the CaR as both an amplifier and attenuator of myo-endothelial coupling in the vasculature is described.
Collapse
Affiliation(s)
- A H Weston
- Faculty of Life Sciences, University of Manchester, UK
| | | | | | | |
Collapse
|
18
|
Thakore P, Ho WSV. Vascular actions of calcimimetics: role of Ca²(+) -sensing receptors versus Ca²(+) influx through L-type Ca²(+) channels. Br J Pharmacol 2011; 162:749-62. [PMID: 20958288 DOI: 10.1111/j.1476-5381.2010.01079.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND PURPOSE The calcimimetic, (R)-N-(3-(3-(trifluoromethyl)phenyl)propyl)-1-(1-napthyl)ethylamine hydrochloride (cinacalcet), which activates Ca²(+) -sensing receptors (CaR) in parathyroid glands, is used to treat hyperparathyroidism. Interestingly, CaR in perivascular nerves or endothelial cells is also thought to modulate vascular tone. This study aims to characterize the vascular actions of calcimimetics. EXPERIMENTAL APPROACH In rat isolated small mesenteric arteries, the relaxant responses to the calcimimetics, cinacalcet and (R)-2-[[[1-(1-naphthyl)ethyl]amino]methyl]-1H-indole hydrochloride (calindol) were characterized, with particular emphasis on the role of CaR, endothelium, perivascular nerves, K(+) channels and Ca²(+) channels. Effects of L-ornithine, which activates a Ca(2+) -sensitive receptor related to CaR (GPRC6A), were also tested. KEY RESULTS Cinacalcet induced endothelium-independent relaxation (pEC₅₀ 5.58 ± 0.07, E(max) 97 ± 6%) that was insensitive to sensory nerve desensitization by capsaicin or blockade of large-conductance Ca²(+) -activated K(+) channels by iberiotoxin. Calindol, another calcimimetic, caused more potent relaxation (pEC₅₀ 6.10 ± 0.10, E(max) 101 ± 6%), which was attenuated by endothelial removal or capsaicin, but not iberiotoxin. The negative modulator of CaR, calhex 231 or changes in [Ca²(+) ](o) had negligible effect on relaxation to both calcimimetics. The calcimimetics relaxed vessels precontracted with high [K(+) ](o) and inhibited Ca²(+) influx in endothelium-denuded vessels stimulated by methoxamine, but not ionomycin. They also inhibited contractions to the L-type Ca²(+) channel activator, BayK8644. L-ornithine induced small relaxation alone and had no effect on the responses to calcimimetics. CONCLUSION AND IMPLICATIONS Cinacalcet and calindol are potent arterial relaxants. Under the experimental conditions used, they predominantly act by inhibiting Ca²(+) influx through L-type Ca²(+) channels into vascular smooth muscle, whereas Ca²(+) -sensitive receptors (CaR or GPRC6A) play a minor role.
Collapse
Affiliation(s)
- Pratish Thakore
- Division of Basic Medical Sciences, St George's University of London, Cranmer Terrace, London, UK
| | | |
Collapse
|
19
|
Chow JYC, Estrema C, Orneles T, Dong X, Barrett KE, Dong H. Calcium-sensing receptor modulates extracellular Ca(2+) entry via TRPC-encoded receptor-operated channels in human aortic smooth muscle cells. Am J Physiol Cell Physiol 2011; 301:C461-8. [PMID: 21562303 DOI: 10.1152/ajpcell.00389.2010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Ca-sensing receptor (CaSR), a member of the G protein-coupled receptor family, regulates the synthesis of parathyroid hormone in response to changes in serum Ca(2+) concentrations. The functions of CaSR in human vascular smooth muscle cells are largely unknown. Here we sought to study CaSR activation and the underlying molecular mechanisms in human aortic smooth muscle cells (HASMC). Extracellular Ca(2+) ([Ca(2+)](o)) dose-dependently increased free cytosolic Ca(2+) ([Ca(2+)](cyt)) in HASMC, with a half-maximal response (EC(50)) of 0.52 mM and a Hill coefficient of 5.50. CaSR was expressed in HASMC, and the [Ca(2+)](o)-induced [Ca(2+)](cyt) rise was abolished by dominant negative mutants of CaSR. The CaSR-mediated increase in [Ca(2+)](cyt) was also significantly inhibited by pertussis toxin, the phospholipase C inhibitor U-73122, or the general protein kinase C (PKC) inhibitor chelerythrine, but not by the conventional PKC inhibitor, Gö6976. Depletion of membrane cholesterol by pretreatment with methyl-β-cyclodextrin markedly decreased CaSR-induced increase in [Ca(2+)](cyt). Blockade of TRPC channels with 2-aminoethoxydiphenyl borate, SKF-96365, or La(3) significantly inhibited [Ca(2+)](o) entry, whereas activation of TRPC6 channels with flufenamic acid potentiated [Ca(2+)](o) entry. Neither cyclopiazonic acid nor caffeine or ionomycin had any effect on [Ca(2+)](cyt) in [Ca(2+)](o)-free solutions. TRPC6 and PKCε mRNA and proteins were detected in HASMC, and [Ca(2+)](o) induced PKCε phosphorylation, which could be prevented by chelerythrine. Our data suggest that CaSR activation mediates [Ca(2+)](o) entry, likely through TRPC6-encoded receptor-operated channels that are regulated by a PLC/PKCε cascade. Our study therefore provides evidence not only for functional expression of CaSR, but also for a novel pathway whereby it regulates [Ca(2+)](o) entry in HASMC.
Collapse
Affiliation(s)
- Jimmy Y C Chow
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | | | | | | | | | | |
Collapse
|
20
|
Koh J, Dar M, Untch BR, Dixit D, Shi Y, Yang Z, Adam MA, Dressman H, Wang X, Gesty-Palmer D, Marks JR, Spurney R, Druey KM, Olson JA. Regulator of G protein signaling 5 is highly expressed in parathyroid tumors and inhibits signaling by the calcium-sensing receptor. Mol Endocrinol 2011; 25:867-76. [PMID: 21393447 DOI: 10.1210/me.2010-0277] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The molecular mechanisms responsible for aberrant calcium signaling in parathyroid disease are poorly understood. The loss of appropriate calcium-responsive modulation of PTH secretion observed in parathyroid disease is commonly attributed to decreased expression of the calcium-sensing receptor (CaSR), a G protein-coupled receptor. However, CaSR expression is highly variable in parathyroid adenomas, and the lack of correlation between CaSR abundance and calcium-responsive PTH kinetics indicates that mechanisms independent of CaSR expression may contribute to aberrant calcium sensing in parathyroid disease. To gain a better understanding of parathyroid tumors and the molecular determinants that drive parathyroid adenoma development, we performed gene expression profiling on a panel of 64 normal and neoplastic parathyroid tissues. The microarray data revealed high-level expression of genes known to be involved in parathyroid biology (PTH, VDR, CGA, CaSR, and GCM2). Moreover, our screen identified regulator of G protein signaling 5 (RGS5) as a candidate inhibitor of CaSR signaling. We confirmed RGS5 to be highly expressed in parathyroid adenomas relative to matched-pair normal glands. Transient expression of RGS5 in cells stably expressing CaSR resulted in dose-dependent abrogation of calcium-stimulated inositol trisphosphate production and ERK1/2 phosphorylation. Furthermore, we found that RGS5-nullizygous mice display reduced plasma PTH levels, an outcome consistent with attenuated opposition to CaSR activity. Collectively, these data suggest that RGS5 can act as a physiological regulator of calcium sensing by CaSR in the parathyroid gland. The abnormally elevated expression of RGS5 observed in parathyroid adenomas could thus represent a novel mechanism of CaSR desensitization in patients with primary hyperparathyroidism.
Collapse
Affiliation(s)
- James Koh
- Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Atchison DK, Ortiz-Capisano MC, Beierwaltes WH. Acute activation of the calcium-sensing receptor inhibits plasma renin activity in vivo. Am J Physiol Regul Integr Comp Physiol 2010; 299:R1020-6. [PMID: 20660105 PMCID: PMC2957381 DOI: 10.1152/ajpregu.00238.2010] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Accepted: 07/19/2010] [Indexed: 01/17/2023]
Abstract
In vitro, the renin-secreting juxtaglomerular cells express the calcium-sensing receptor, and its activation with the calcimimetic cinacalcet inhibits renin release. To test whether the activation of calcium-sensing receptor similarly inhibits plasma renin activity (PRA) in vivo, we hypothesized that the calcium-sensing receptor is expressed in juxtaglomerular cells in vivo, and acutely administered cinacalcet would inhibit renin activity in anesthetized rats. Since cinacalcet inhibits parathyroid hormone, which may stimulate renin activity, we sought to determine whether cinacalcet inhibits renin activity by decreasing parathyroid hormone. Lastly, we hypothesized that chronically administered cinacalcet would inhibit basal and stimulated renin in conscious rats. Calcium-sensing receptors and renin were localized in the same juxtaglomerular cells using immunofluorescence in rat cortical slices fixed in vivo. Cinacalcet was administered acutely via intravenous bolus in anesthetized rats and chronically in conscious rats by oral gavage. Acute administration of cinacalcet decreased basal renin activity from 13.6 ± 2.4 to 6.1 ± 1.1 ng ANG I·ml(-1)·h(-1) (P < 0.001). Likewise, cinacalcet decreased furosemide-stimulated renin from 30.6 ± 2.3 to 21.3 ± 2.3 ng ANG I·ml(-1)·h(-1) (P < 0.001). In parathyroidectomized rats, cinacalcet decreased renin activity from 9.3 ± 1.3 to 5.2 ± 0.5 ng ANG I·ml(-1)·h(-1) (P < 0.05) similar to sham-operated controls (13.5 ± 2.2 to 6.6 ± 0.8 ng ANG I·ml(-1)·h(-1), P < 0.05). Chronic administration of cinacalcet over 7 days had no significant effect on PRA under basal or stimulated conditions. In conclusion, calcium-sensing receptors are expressed in juxtaglomerular cells in vivo, and acute activation of these receptors with cinacalcet inhibits PRA in anesthetized rats, independent of parathyroid hormone.
Collapse
Affiliation(s)
- Douglas K Atchison
- Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan 48202, USA
| | | | | |
Collapse
|
22
|
Riccardi D, Finney BA, Wilkinson WJ, Kemp PJ. Novel regulatory aspects of the extracellular Ca2+-sensing receptor, CaR. Pflugers Arch 2009; 458:1007-22. [PMID: 19484257 DOI: 10.1007/s00424-009-0681-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2009] [Revised: 04/30/2009] [Accepted: 05/05/2009] [Indexed: 01/15/2023]
Abstract
The capacity to sense and adapt to changes in environmental cues is of paramount importance for every living organism. From yeast to man, cells must be able to match cellular activities to growth environment and nutrient availability. Key to this process is the development of membrane-bound systems that can detect modifications in the extracellular environment and to translate these into biological responses. Evidence gathered over the last 15 years has demonstrated that many of these cell surface "sensors" belong to the G protein-coupled receptor superfamily. Crucial to our understanding of nutrient sensing in mammalian species has been the identification of the extracellular Ca(2+)/cation-sensing receptor, CaR. CaR was the first ion-sensing molecule identified in man and genetic studies in humans have revealed the importance of the CaR in mineral ion metabolism. Latter, it has become apparent that the CaR also plays an important role outside the Ca(2+) homeostatic system, as an integrator of multiple environmental signals for the regulation of many vital cellular processes, from cell-to-cell communication to secretion and cell survival/cell death. Recently, novel aspects of receptor function reveal an unexpected role for the CaR in the regulation of growth and development in utero.
Collapse
|
23
|
Meola M, Petrucci I, Barsotti G. Long-term treatment with cinacalcet and conventional therapy reduces parathyroid hyperplasia in severe secondary hyperparathyroidism. Nephrol Dial Transplant 2009; 24:982-9. [PMID: 19181759 PMCID: PMC2644631 DOI: 10.1093/ndt/gfn654] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Accepted: 10/31/2008] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND The effect of cinacalcet on the structural pattern of hyperplastic parathyroid glands was evaluated, using high-resolution colour Doppler (CD) sonography, in haemodialysis patients with severe, inadequately controlled, secondary hyperparathyroidism (sHPT). METHODS Nine patients (6 males, 3 females; mean age +/- SD, 55.5 +/- 12.6 years) received cinacalcet, with adaptation of existing concomitant therapies. Biochemical parameters and the morphology and vascular pattern of hyperplastic parathyroid glands were measured at baseline and every 6 months thereafter, for a follow-up period of 24-30 months. RESULTS At baseline, 28 hyperplastic glands were identified. Cinacalcet led to a reduction in glandular volume during the course of the study: 68% in glands with a baseline volume <500 mm(3) and 54% in glands with a baseline volume >or=500 mm(3). The mean volume +/- SD of glands <500 mm(3) changed significantly from the baseline (233 +/- 115 mm(3)) to the end of follow-up (102 +/- 132 mm(3), P = 0.007). Levels of mean serum phosphorus, calcium and calcium-phosphorus product decreased, but not significantly, whereas there were significant decreases in mean parathyroid hormone +/- SD levels (1196 +/- 381 pg/ml versus 256 +/- 160 pg/ml; P < 0.0001) and alkaline phosphatase +/- SD levels (428 +/- 294 versus 223 +/- 88 IU/l; P = 0.04), accompanied by an improvement in a subjective clinical score. CONCLUSIONS Cinacalcet, in combination with conventional treatments, led to an improvement in biochemical and clinical parameters of sHPT and reduced glandular volume in patients with severe sHPT. Volume reduction was more evident in smaller glands. Longer term, larger, randomized clinical trials are needed to confirm these preliminary findings and to further define a more systematic approach in the treatment of sHPT.
Collapse
Affiliation(s)
- Mario Meola
- Department of Internal Medicine, S Anna School of Advanced Studies, Nephrology and Dialysis Unit, University of Pisa, Italy.
| | | | | |
Collapse
|
24
|
Touyz RM, Montezano AC. Vascular smooth muscle cells sense calcium: a new paradigm in vascular calcification. Cardiovasc Res 2008; 81:237-9. [PMID: 19088081 DOI: 10.1093/cvr/cvn345] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
25
|
Smajilovic S, Tfelt-Hansen J. Novel Role of the Calcium-Sensing Receptor in Blood Pressure Modulation. Hypertension 2008; 52:994-1000. [DOI: 10.1161/hypertensionaha.108.117689] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Sanela Smajilovic
- From the Laboratory of Molecular Cardiology (S.S., J.T-H.), Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Denmark; and the Danish National Research Foundation Centre for Cardiac Arrhythmia (S.S., J.T-H.), Copenhagen, Denmark
| | - Jacob Tfelt-Hansen
- From the Laboratory of Molecular Cardiology (S.S., J.T-H.), Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Denmark; and the Danish National Research Foundation Centre for Cardiac Arrhythmia (S.S., J.T-H.), Copenhagen, Denmark
| |
Collapse
|