1
|
Ishizawa M, Takano M, Kittaka A, Matsumoto T, Makishima M. 2α-Substituted Vitamin D Derivatives Effectively Enhance the Osteoblast Differentiation of Dedifferentiated Fat Cells. Biomolecules 2024; 14:706. [PMID: 38927109 PMCID: PMC11202298 DOI: 10.3390/biom14060706] [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: 03/21/2024] [Revised: 06/08/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
The active form of vitamin D3, 1α,25-dihydroxyvitamin D3 [1,25(OH)2D3], is a principal regulator of calcium homeostasis through activation of the vitamin D receptor (VDR). Previous studies have shown that 2α-(3-hydroxypropyl)-1,25D3 (O1C3) and 2α-(3-hydroxypropoxy)-1,25D3 (O2C3), vitamin D derivatives resistant to inactivation enzymes, can activate VDR, induce leukemic cell differentiation, and increase blood calcium levels in rats more effectively than 1,25(OH)2D3. In this study, to further investigate the usefulness of 2α-substituted vitamin D derivatives, we examined the effects of O2C3, O1C3, and their derivatives on VDR activity in cells and mouse tissues and on osteoblast differentiation of dedifferentiated fat (DFAT) cells, a cell type with potential therapeutic application in regenerative medicine. In cell culture experiments using kidney-derived HEK293 cells, intestinal mucosa-derived CaCO2 cells, and osteoblast-derived MG63 cells, and in mouse experiments, O2C2, O2C3, O1C3, and O1C4 had a weaker effect than or equivalent effect to 1,25(OH)2D3 in VDR transactivation and induction of the VDR target gene CYP24A1, but they enhanced osteoblast differentiation in DFAT cells equally to or more effectively than 1,25(OH)2D3. In long-term treatment with the compound without the medium change (7 days), the derivatives enhanced osteoblast differentiation more effectively than 1,25(OH)2D3. O2C3 and O1C3 were more stable than 1,25(OH)2D3 in DFAT cell culture. These results indicate that 2α-substituted vitamin D derivatives, such as inactivation-resistant O2C3 and O1C3, are more effective than 1,25(OH)2D3 in osteoblast differentiation of DFAT cells, suggesting potential roles in regenerative medicine with DFAT cells and other multipotent cells.
Collapse
Affiliation(s)
- Michiyasu Ishizawa
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, Itabashi-ku, Tokyo 173-8610, Japan
| | - Masashi Takano
- Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan; (M.T.); (A.K.)
| | - Atsushi Kittaka
- Faculty of Pharmaceutical Sciences, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan; (M.T.); (A.K.)
| | - Taro Matsumoto
- Division of Cell Regeneration and Transplantation, Department of Functional Morphology, Nihon University School of Medicine, Tokyo 173-8610, Japan;
| | - Makoto Makishima
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University School of Medicine, Itabashi-ku, Tokyo 173-8610, Japan
| |
Collapse
|
2
|
Xu Z, Yao X, Duan C, Liu H, Xu H. Metabolic changes in kidney stone disease. Front Immunol 2023; 14:1142207. [PMID: 37228601 PMCID: PMC10203412 DOI: 10.3389/fimmu.2023.1142207] [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: 01/11/2023] [Accepted: 03/06/2023] [Indexed: 05/27/2023] Open
Abstract
Kidney stone disease (KSD) is one of the earliest medical diseases known, but the mechanism of its formation and metabolic changes remain unclear. The formation of kidney stones is a extensive and complicated process, which is regulated by metabolic changes in various substances. In this manuscript, we summarized the progress of research on metabolic changes in kidney stone disease and discuss the valuable role of some new potential targets. We reviewed the influence of metabolism of some common substances on stone formation, such as the regulation of oxalate, the release of reactive oxygen species (ROS), macrophage polarization, the levels of hormones, and the alternation of other substances. New insights into changes in substance metabolism changes in kidney stone disease, as well as emerging research techniques, will provide new directions in the treatment of stones. Reviewing the great progress that has been made in this field will help to improve the understanding by urologists, nephrologists, and health care providers of the metabolic changes in kidney stone disease, and contribute to explore new metabolic targets for clinical therapy.
Collapse
Affiliation(s)
- Zhenzhen Xu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xiangyang Yao
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chen Duan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haoran Liu
- Stanford Bio-X, Stanford University, San Francisco, CA, United States
| | - Hua Xu
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, China
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China
- Cancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
3
|
Sundararaman SS, Peters LJF, Jansen Y, Gencer S, Yan Y, Nazir S, Bonnin Marquez A, Kahles F, Lehrke M, Biessen EAL, Jankowski J, Weber C, Döring Y, van der Vorst EPC. Adipocyte calcium sensing receptor is not involved in visceral adipose tissue inflammation or atherosclerosis development in hyperlipidemic Apoe -/- mice. Sci Rep 2021; 11:10409. [PMID: 34001955 PMCID: PMC8128899 DOI: 10.1038/s41598-021-89893-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 05/04/2021] [Indexed: 01/02/2023] Open
Abstract
The calcium sensing receptor (CaSR) is a G-protein coupled receptor that especially plays an important role in the sensing of extracellular calcium to maintain its homeostasis. Several in-vitro studies demonstrated that CaSR plays a role in adipose tissue metabolism and inflammation, resulting in systemic inflammation and contributing to atherosclerosis development. The aim of this study was to investigate whether adipocyte CaSR plays a role in adipose tissue inflammation in-vivo and atherosclerosis development. By using a newly established conditional mature adipocyte specific CaSR deficient mouse on a hyperlipidemic and atherosclerosis prone Apoe−/− background it could be shown that CaSR deficiency in adipocytes does neither contribute to initiation nor to progression of atherosclerotic plaques as judged by the unchanged lesion size or composition. Additionally, CaSR deficiency did not influence gonadal visceral adipose tissue (vAT) inflammation in-vivo, although a small decrease in gonadal visceral adipose cholesterol content could be observed. In conclusion, adipocyte CaSR seems not to be involved in vAT inflammation in-vivo and does not influence atherosclerosis development in hyperlipidemic Apoe−/− mice.
Collapse
Affiliation(s)
- Sai Sahana Sundararaman
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.,Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany.,Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Linsey J F Peters
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.,Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany.,Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Yvonne Jansen
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Selin Gencer
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Yi Yan
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Sumra Nazir
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.,Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany.,Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Andrea Bonnin Marquez
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.,Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany.,Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Florian Kahles
- Department of Internal Medicine I-Cardiology, University Hospital Aachen, Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I-Cardiology, University Hospital Aachen, Aachen, Germany
| | - Erik A L Biessen
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany.,Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany.,Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands.,Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Yvonne Döring
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.,Department of Angiology, Swiss Cardiovascular Center, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Emiel P C van der Vorst
- Interdisciplinary Center for Clinical Research (IZKF), RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany. .,Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, Aachen, Germany. .,Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands. .,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany. .,DZHK (German Centre for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
4
|
Chanpaisaeng K, Teerapornpuntakit J, Wongdee K, Charoenphandhu N. Emerging roles of calcium-sensing receptor in the local regulation of intestinal transport of ions and calcium. Am J Physiol Cell Physiol 2020; 320:C270-C278. [PMID: 33356945 DOI: 10.1152/ajpcell.00485.2020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Whether the intestinal mucosal cells are capable of sensing calcium concentration in the lumen and pericellular interstitium remains enigmatic for decades. Most calcium-regulating organs, such as parathyroid gland, kidney, and bone, are capable of using calcium-sensing receptor (CaSR) to detect plasma calcium and trigger appropriate feedback responses to maintain calcium homeostasis. Although both CaSR transcripts and proteins are abundantly expressed in the crypt and villous enterocytes of the small intestine as well as the surface epithelial cells of the large intestine, the studies of CaSR functions have been limited to amino acid sensing and regulation of epithelial fluid secretion. Interestingly, several lines of recent evidence have indicated that the enterocytes use CaSR to monitor luminal and extracellular calcium levels, thereby reducing the activity of transient receptor potential channel, subfamily V, member 6, and inducing paracrine and endocrine feedback responses to restrict calcium absorption. Recent investigations in zebra fish and rodents have also suggested the role of fibroblast growth factor (FGF)-23 as an endocrine and/or paracrine factor participating in the negative control of intestinal calcium transport. In this review article, besides the CaSR-modulated ion transport, we elaborate the possible roles of CaSR and FGF-23 as well as their crosstalk as parts of a negative feedback loop for counterbalancing the seemingly unopposed calciotropic effect of 1,25-dihydroxyvitamin D3 on the intestinal calcium absorption.
Collapse
Affiliation(s)
- Krittikan Chanpaisaeng
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand.,Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani, Thailand
| | - Jarinthorn Teerapornpuntakit
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand.,Department of Physiology, Faculty of Medical Science, Naresuan University, Phitsanulok, Thailand
| | - Kannikar Wongdee
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand.,Faculty of Allied Health Sciences, Burapha University, Chonburi, Thailand
| | - Narattaphol Charoenphandhu
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand.,Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand.,Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand.,The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| |
Collapse
|
5
|
Tay YKD, Liu M, Bandeira L, Bucovsky M, Lee JA, Silverberg SJ, Walker MD. Occult urolithiasis in asymptomatic primary hyperparathyroidism. Endocr Res 2018; 43:106-115. [PMID: 29400579 PMCID: PMC6042842 DOI: 10.1080/07435800.2018.1431275] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Recent international guidelines suggest renal imaging to detect occult urolithiasis in all patients with asymptomatic primary hyperparathyroidism (PHPT), but data regarding their prevalence and associated risk factors are limited. We evaluated the prevalence and risk factors for occult urolithiasis. METHODS Cross-sectional analysis of 96 asymptomatic PHPT patients from a university hospital in the United States with and without occult nephrolithiasis. RESULTS Occult urolithiasis was identified in 21% of patients. Stone formers had 47% higher 24-hour urinary calcium excretion (p = 0.002). Although available in only a subset of patients (n = 28), activated vitamin D [1,25(OH)2D] was 29% higher (p = 0.02) in stone formers. There was no difference in demographics, BMI, calcium or vitamin D intake, other biochemistries, renal function, BMD, or fractures. Receiver operating characteristic curves indicated that urinary calcium excretion and 1,25(OH)2D had an area under the curve of 0.724 (p = 0.003) and 0.750 (p = 0.04), respectively. A urinary calcium threshold of >211mg/day provided a sensitivity of 84.2% and a specificity of 55.3% while a 1,25(OH)2D threshold of >91pg/mL provided a sensitivity and specificity of 62.5% and 90.0% respectively for the presence of stones. CONCLUSION Occult urolithiasis is present in about one-fifth of patients with asymptomatic PHPT and is associated with higher urinary calcium and 1,25(OH)2D. Given that most patients will not have occult urolithiasis, targeted imaging in those most likely to have occult stones rather than screening all asymptomatic PHPT patients may be useful. The higher sensitivity of urinary calcium versus 1,25(OH)2D suggests screening those with higher urinary calcium may be an appropriate approach.
Collapse
Affiliation(s)
- Yu-Kwang Donovan Tay
- Division of Endocrinology, Department of Medicine, Columbia University, College of Physicians and Surgeons, New York
- Department of Medicine, Sengkang Health, Singapore
- Osteoporosis and Bone Metabolism Unit, Department of Endocrinology, Singapore General Hospital, Singapore
| | - Minghao Liu
- Division of Endocrinology, Department of Medicine, Columbia University, College of Physicians and Surgeons, New York
| | - Leonardo Bandeira
- Division of Endocrinology, Department of Medicine, Columbia University, College of Physicians and Surgeons, New York
| | - Mariana Bucovsky
- Division of Endocrinology, Department of Medicine, Columbia University, College of Physicians and Surgeons, New York
| | - James A. Lee
- Department of Endocrine Surgery, Columbia University, New York, USA
| | - Shonni J. Silverberg
- Division of Endocrinology, Department of Medicine, Columbia University, College of Physicians and Surgeons, New York
| | - Marcella D. Walker
- Division of Endocrinology, Department of Medicine, Columbia University, College of Physicians and Surgeons, New York
| |
Collapse
|
6
|
Schreckenberg R, Schlüter KD. Calcium sensing receptor expression and signalling in cardiovascular physiology and disease. Vascul Pharmacol 2018; 107:S1537-1891(17)30323-3. [PMID: 29514057 DOI: 10.1016/j.vph.2018.02.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/18/2018] [Accepted: 02/21/2018] [Indexed: 12/21/2022]
Abstract
Initially identified in the parathyroidea, the calcium sensing receptor (CaSR) is now recognized as an ubiquitously expressed receptor that exerts specific functions in multiple organs including the cardiovascular system. This review will focus on the role that CaSR plays in vascular and cardiac tissues. In the vasculature, CaSR is expressed in endothelial and smooth muscle cells. CaSR of endothelial cells participates in part to the regulation of local perfusion by linkage of CaSR activation to endothelial hyperpolarization and nitric oxide release. CaSR of smooth muscle cells is involved in the control of proliferation. In the pulmonary vasculature, however, CaSR participates in the onset of pulmonary hypertension, making CaSR antagonism a therapeutic option in this case. In the heart, CaSR is expressed in cardiac fibroblasts and myoyctes, contributing to normal cardiac function and composition of extracellular matrix. More important, activation of CaSR may participate in the cardiac protective effects of ischaemic pre-conditioning. In conclusion, CaSR plays an important physiological role in many regulatory pathways of the cardiovascular system, but due to the complex interaction between various cardiovascular cells and cell-specific effects, use of activators or inhibitors of CaSR for treatment of specific disease forms is yet not on the way.
Collapse
|
7
|
Walker RW, Zhang S, Coleman-Barnett JA, Hamm LL, Hering-Smith KS. Calcium receptor signaling and citrate transport. Urolithiasis 2018; 46:409-418. [PMID: 29383416 DOI: 10.1007/s00240-018-1035-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 01/10/2018] [Indexed: 01/22/2023]
Abstract
The calcium sensing receptor (CaSR) in the distal nephron decreases the propensity for calcium stones. Here we investigate if the apical CaSR in the proximal tubule also prevents stone formation acting via regulation of apical dicarboxylate and citrate transport. Urinary citrate, partially reabsorbed as a dicarboxylate in the proximal tubule lumen, inhibits stone formation by complexing calcium. We previously demonstrated a novel apical calcium-sensitive dicarboxylate transport system in OK proximal tubule cells. This calcium-sensitive process has the potential to modulate the amount of citrate available to complex increased urinary calcium. Using isotope labeled succinate uptake in OK cells along with various pharmacologic tools we examined whether the CaSR alters apical dicarboxylate transport and through which signal transduction pathways this occurs. Our results indicate that in the proximal tubule CaSR adjusts apical dicarboxylate transport, and does so via a CaSR → Gq → PKC signaling pathway. Thus, the CaSR may decrease the propensity for stone formation via actions in both proximal and distal nephron segments.
Collapse
Affiliation(s)
- Ryan W Walker
- Nephrology and Hypertension 8545, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA
| | - Shijia Zhang
- Nephrology and Hypertension 8545, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA
| | - Joycelynn A Coleman-Barnett
- Nephrology and Hypertension 8545, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA
| | - L Lee Hamm
- Nephrology and Hypertension 8545, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA
| | - Kathleen S Hering-Smith
- Nephrology and Hypertension 8545, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70112, USA.
| |
Collapse
|
8
|
Abstract
Calcium is an important ion in cell signaling, hormone regulation, and bone health. Its regulation is complex and intimately connected to that of phosphate homeostasis. Both ions are maintained at appropriate levels to maintain the extracellular to intracellular gradients, allow for mineralization of bone, and to prevent extra skeletal and urinary calcification. The homeostasis involves the target organs intestine, parathyroid glands, kidney, and bone. Multiple hormones converge to regulate the extracellular calcium level: parathyroid hormone, vitamin D (principally 25(OH)D or 1,25(OH)2D), fibroblast growth factor 23, and α-klotho. Fine regulation of calcium homeostasis occurs in the thick ascending limb and collecting tubule segments via actions of the calcium sensing receptor and several channels/transporters. The kidney participates in homeostatic loops with bone, intestine, and parathyroid glands. Initially in the course of progressive kidney disease, the homeostatic response maintains serum levels of calcium and phosphorus in the desired range, and maintains neutral balance. However, once the kidneys are no longer able to appropriately respond to hormones and excrete calcium and phosphate, positive balance ensues leading to adverse cardiac and skeletal abnormalities. © 2016 American Physiological Society. Compr Physiol 6:1781-1800, 2016.
Collapse
Affiliation(s)
- Sharon M Moe
- Division of Nephrology, Indiana University School of Medicine, Roudebush Veterans Administration Medical Center, Indianapolis, Indiana.,Section of Nephrology, Roudebush Veterans Administration Medical Center, Indianapolis, Indiana
| |
Collapse
|
9
|
|
10
|
Moor MB, Bonny O. Ways of calcium reabsorption in the kidney. Am J Physiol Renal Physiol 2016; 310:F1337-50. [PMID: 27009338 DOI: 10.1152/ajprenal.00273.2015] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 03/17/2016] [Indexed: 11/22/2022] Open
Abstract
The role of the kidney in calcium homeostasis has been reshaped from a classic view in which the kidney was regulated by systemic calcitropic hormones such as vitamin D3 or parathyroid hormone to an organ actively taking part in the regulation of calcium handling. With the identification of the intrinsic renal calcium-sensing receptor feedback system, the regulation of paracellular calcium transport involving claudins, and new paracrine regulators such as klotho, the kidney has emerged as a crucial modulator not only of calciuria but also of calcium homeostasis. This review summarizes recent molecular and endocrine contributors to renal calcium handling and highlights the tight link between calcium and sodium reabsorption in the kidney.
Collapse
Affiliation(s)
- Matthias B Moor
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; and
| | - Olivier Bonny
- Department of Pharmacology and Toxicology, University of Lausanne, Lausanne, Switzerland; and Service of Nephrology, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
11
|
Owen JL, Cheng SX, Ge Y, Sahay B, Mohamadzadeh M. The role of the calcium-sensing receptor in gastrointestinal inflammation. Semin Cell Dev Biol 2015; 49:44-51. [PMID: 26709005 DOI: 10.1016/j.semcdb.2015.10.040] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 10/28/2015] [Accepted: 10/29/2015] [Indexed: 12/13/2022]
Abstract
The gastrointestinal (GI) tract must balance the extraction of energy and metabolic end-products from ingested nutrition and resident gut microbes and the maintenance of a symbiotic relationship with this microbiota, with the ability to mount functional immune responses to pathogenic organisms to maintain GI health. The gut epithelium is equipped with bacteria-sensing mechanisms that discriminate between pathogenic and commensal microorganisms and regulate host responses between immunity and tolerance. The epithelium also expresses numerous nutrient-sensing receptors, but their importance in the preservation of the gut microbiota and immune homeostasis remains largely unexplored. Observations that a deficiency in the extracellular calcium-sensing receptor (CaSR) using intestinal epithelium-specific receptor knockout mice resulted in diminished intestinal barrier integrity, altered composition of the gut microbiota, modified expression of intestinal pattern recognition receptors, and a skewing of local and systemic innate responses from regulatory to stimulatory, may change the way that this receptor is considered as a potential immunotherapeutic target in gut homeostasis. These findings suggest that pharmacologic CaSR activators and CaSR-based nutrients such as calcium, polyamines, phenylalanine, tryptophan, and oligo-peptides might be useful in conditioning the gut microenvironment, and thus, in the prevention and treatment of disorders such as inflammatory bowel disease (IBD), infectious enterocolitis, and other inflammatory and secretory diarrheal diseases. Here, we review the emerging roles of the CaSR in intestinal homeostasis and its therapeutic potential for gut pathology.
Collapse
Affiliation(s)
- Jennifer L Owen
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Sam X Cheng
- Division of Gastroenterology, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Yong Ge
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL, USA; Division of Hepatology, Gastroenterology, and Nutrition, University of Florida, Gainesville, FL, USA
| | - Bikash Sahay
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL, USA; Division of Hepatology, Gastroenterology, and Nutrition, University of Florida, Gainesville, FL, USA
| | - Mansour Mohamadzadeh
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL, USA; Division of Hepatology, Gastroenterology, and Nutrition, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW Variations in extracellular calcium level have a large impact on kidney function. Most of the effects seen are attributed to the calcium-sensing receptor (CaSR), a widely expressed G-protein-coupled cell surface protein with an important function in bone mineral homeostasis. The purpose of this review is to recapitulate the novel functional aspects of CaSR. RECENT FINDINGS Results from mouse models demonstrate important functions for CaSR in various tissues. In the kidney, the main role of CaSR is the regulation of calcium reabsorption in the thick ascending limb, independently of its role on parathyroid hormone secretion. CaSR modulates claudin 14, the gatekeeper of paracellular ion transport in the thick ascending limb that is associated with urinary calcium excretion. One intracellular signaling pathway by which CaSR alters tight junction permeability is the calcineurin-NFAT1c-microRNA-claudin14 axis. SUMMARY The main function of CaSR in the kidney is the regulation of calcium excretion in the thick ascending limb, independently of parathyroid hormone. CaSR modulates paracellular cation transport by altering expression of the tight junction protein claudin 14. Still more work is needed to fully understand all functions of CaSR in the kidney. Alternative pathways of calcium 'sensing' in the kidney need to be investigated.
Collapse
Affiliation(s)
- Hakan R Toka
- aDivision of Nephrology, Beth Israel Deaconess Medical Center bDivision of Nephrology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
13
|
Kladnitsky O, Rozenfeld J, Azulay-Debby H, Efrati E, Zelikovic I. The claudin-16 channel gene is transcriptionally inhibited by 1,25-dihydroxyvitamin D. Exp Physiol 2014; 100:79-94. [PMID: 25557732 DOI: 10.1113/expphysiol.2014.083394] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 10/20/2014] [Indexed: 01/12/2023]
Abstract
NEW FINDINGS What is the central question of this study? In the kidney, the bulk of the filtered Mg(2+) is reabsorbed in the thick ascending limb by paracellular conductance, mediated by the tight junction protein, claudin-16, which is encoded by the gene CLDN16. The role of 1,25-dihydroxyvitamin D [1,25(OH)2 VitD] in renal Mg(2+) handling is unclear. We aimed to explore the molecular mechanisms underlying the effect of 1,25(OH)2 VitD on claudin-16-mediated Mg(2+) transport. What is the main finding and its importance? Paracellular, claudin-16-mediated Mg(2+) transport is transcriptionally repressed by 1,25(OH)2 VitD, probably via a Ca(2+)-sensing receptor-dependent mechanism. This renal effect of 1,25(OH)2 VitD may serve as an adaptive mechanism to the 1,25(OH)2 VitD-induced enteric hyperabsorption of dietary Mg(2+). Magnesium is reabsorbed in the thick ascending limb by paracellular conductance, mediated by the CLDN16-encoded tight junction protein, claudin-16. However, the role of 1,25-dihydroxyvitamin D [1,25(OH)2 VitD] in renal Mg(2+) handling is unclear. We have shown that Mg(2+) depletion increases and 1,25(OH)2 VitD inhibits CLDN16 transcription. We have now explored further the molecular mechanisms underlying the effect of 1,25(OH)2 VitD on claudin-16-mediated Mg(2+) transport. Adult mice received parenteral 1,25(OH)2 VitD or 1,25(OH)2 VitD combined with either high-Mg(2+) or low-Mg(2+) diets. Administration of 1,25(OH)2 VitD enhanced urinary excretion of Mg(2+) and Ca(2+). The 1,25(OH)2 VitD also increased renal Ca(2+)-sensing receptor (CaSR) mRNA and decreased renal claudin-16 and claudin-19 mRNA and claudin-16 protein, but did not affect renal claudin-2 mRNA. The 1,25(OH)2 VitD reversed the expected increase in claudin-16 mRNA in Mg(2+)-depleted animals. Comparably treated HEK 293 cells showed similar changes in claudin-16 mRNA, but 1,25(OH)2 VitD did not alter mRNA of the TRPM6 Mg(2+) channel. A luciferase reporter vector containing 2.5 kb of 5'-flanking DNA sequence from human CLDN16 (hCLDN16) was transfected into HEK 293 and OK cells. The hCLDN16 promoter activity was modestly decreased by 1,25(OH)2 VitD, but markedly inhibited in HEK 293 cells coexpressing CaSR. Coexpression in OK cells of dominant-negative CaSR completely abolished inhibition of hCLDN16 promoter activity by 1,25(OH)2 VitD. The 1,25(OH)2 VitD-induced decrease in hCLDN16 promoter activity was attenuated in Mg(2+)-depleted HEK 293 cells. In conclusion, 1,25(OH)2 VitD transcriptionally inhibits claudin-16 expression by a mechanism sensitive to CaSR and Mg(2+). This renal effect of 1,25(OH)2 VitD may serve as an adaptive response to the 1,25(OH)2 VitD-induced increase in intestinal Mg(2+) absorption.
Collapse
Affiliation(s)
- Orly Kladnitsky
- Laboratory of Developmental Nephrology, Department of Physiology and Biophysics, Faculty of Medicine, Technion - Israel Institute of Technology, Haifa, Israel
| | | | | | | | | |
Collapse
|
14
|
Besiroglu H, Sahin S, Otunctemur A, Ozbek E. Calcium-sensing receptor gene polymorphisms in patients with calcium urolithiasis: a systematic review. Ren Fail 2014; 36:1187-92. [PMID: 25019182 DOI: 10.3109/0886022x.2014.937673] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Calcium-sensing receptor gene (CaSR) might be a strong component of the complex genetic background regulating calcium excretion and stone formation in kidney. The polymorphisms in different parts of the gene could be associated with calcium stone formation. OBJECTIVE The individual studies indicate a correlation between CaSR gene polymorphisms and urolithiasis but not sufficient to draw a precise result. We aimed to search literature to come to a reliable conclusion through a systematic review. MATERIALS AND METHODS We have serached individiual studies regarding the issue and high quality reviews. We assessed all the studies to draw a conclusion. CONCLUSION The genetic background of the neprolithiasis is an interesting issue deserving to be disscussed on. Regading genes of which, CaSR gene polymorphisms is the leading one, are studied in many individual studies but they are not sufficient to have a precise conclusion. Well-oriented and documented, phenotypically homogenous large cohort studies are needed for further evaluation.
Collapse
Affiliation(s)
- Huseyin Besiroglu
- Department of Urology, Okmeydani Training and Research Hospital , Istanbul , Turkey
| | | | | | | |
Collapse
|
15
|
Garg MK, Kalra S, Mahalle N. The intestinal calcistat: Determinant of clinical vitamin D deficiency. Indian J Endocrinol Metab 2013; 17:780-783. [PMID: 24083155 PMCID: PMC3784857 DOI: 10.4103/2230-8210.117187] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Affiliation(s)
- M. K. Garg
- Department of Endocrinology, Command Hospital (Southern Command), Pune, Maharashtra, India
| | - Sanjay Kalra
- Department of Endocrinology, Bharti Hospital and BRIDE, Karnal, Haryana, India
| | - Namita Mahalle
- Biochemistry Division, Department of Pathology, Deenanath Mangeshkar Hospital and Research Center, Erandawane, Pune, Maharashtra, India
| |
Collapse
|
16
|
Garg M, Mahalle N. Calcium homeostasis, and clinical or subclinical vitamin D deficiency – Can a hypothesis of “intestinal calcistat” explain it all? Med Hypotheses 2013; 81:253-8. [DOI: 10.1016/j.mehy.2013.04.035] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Accepted: 04/17/2013] [Indexed: 01/25/2023]
|
17
|
MacLeod RJ. Extracellular calcium-sensing receptor/PTH knockout mice colons have increased Wnt/β-catenin signaling, reduced non-canonical Wnt signaling, and increased susceptibility to azoxymethane-induced aberrant crypt foci. J Transl Med 2013; 93:520-7. [PMID: 23545937 DOI: 10.1038/labinvest.2013.51] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Epidemiological evidence suggests increased dietary calcium and dairy products reduce the onset of colon cancer. To understand a role of the colonic extracellular calcium-sensing receptor (CaSR) in calcium-mediated chemoprevention of colon cancer, we induced formation of aberrant crypt foci (ACF) caused by azoxymethane (AOM) injection in 'rescued' CaSR-/PTH- (C-/P-) double knockout colons compared with colons from control CaSR+/PTH+ (C+/P+) mice. C-/P- colonic epithelia had increased Wnt/β-catenin signaling as evidenced by 3-8-fold increases in Wnt3a, CyclinD1, and MMP-7 proteins compared with C+/P+ colonic epithelia. The C-/P- colonic epithelia had reduced Wnt5a and Ror2, and a three-fold increase in TNFR1 compared with C+/P+ epithelia. The C-/P- colons and small intestine had extensive neutrophil infiltration with myeloperoxidase (MPO) levels 18-fold higher then C+/P+ small intestine and colon. Saline-injected C-/P- colons had the same number of ACF/cm(2) as C+/P+ colons, which were injected with AOM. However, there were eight times more ACF/cm(2) in the C-/P- injected with AOM compared with C+/P+ colons, which received AOM. Together our results suggest both inflammation and Wnt/β-catenin signaling are increased in the epithelia of 'rescued' CaSR/PTH double knockout colons, and the capacity for non-canonical Wnt signaling through Wnt5a/Ror2 engagement is reduced. The loss of the colonic CaSR increased the number of ACF/cm(2) in response to AOM injection, suggesting colonic CaSR may mediate the chemoprotective effect of increased dietary calcium against colorectal cancer observed in humans.
Collapse
Affiliation(s)
- R John MacLeod
- Department of Biomedical and Molecular Sciences, GIDRU, Queen's University, Kingston, Ontario, Canada K7L 3N6.
| |
Collapse
|
18
|
Quinn SJ, Thomsen ARB, Egbuna O, Pang J, Baxi K, Goltzman D, Pollak M, Brown EM. CaSR-mediated interactions between calcium and magnesium homeostasis in mice. Am J Physiol Endocrinol Metab 2013; 304:E724-33. [PMID: 23360827 PMCID: PMC3625753 DOI: 10.1152/ajpendo.00557.2012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Accepted: 01/23/2013] [Indexed: 11/22/2022]
Abstract
Calcium (Ca) and magnesium (Mg) homeostasis are interrelated and share common regulatory hormones, including parathyroid hormone (PTH) and vitamin D. However, the role of the calcium-sensing receptor (CaSR) in Mg homeostasis in vivo is not well understood. We sought to investigate the interactions between Mg and Ca homeostasis using genetic mouse models with targeted inactivation of PTH (PTH KO) or both PTH and the calcium-sensing receptor (CaSR) (double knockout, DKO). Serum Mg is lower in PTH KO and DKO mice than in WT mice on standard chow, whereas supplemental dietary Ca leads to equivalent Mg levels for all three genotypes. Mg loading increases serum Mg in all genotypes; however, the increase in serum Mg is most pronounced in the DKO mice. Serum Ca is increased with Mg loading in the PTH KO and DKO mice but not in the WT mice. Here, too, the hypercalcemia is much greater in the DKO mice. Serum and especially urinary phosphate are reduced during Mg loading, which is likely due to intestinal chelation of phosphate by Mg. Mg loading decreases serum PTH in WT mice and increases serum calcitonin in both WT and PTH KO mice but not DKO mice. Furthermore, Mg loading elevates serum 1,25-dihydroxyvitamin D in all genotypes, with greater effects in PTH KO and DKO mice, possibly due to reduced levels of serum phosphorus and FGF23. These hormonal responses to Mg loading and the CaSR's role in regulating renal function may help to explain changes in serum Mg and Ca found during Mg loading.
Collapse
Affiliation(s)
- Stephen J Quinn
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Quinn SJ, Thomsen ARB, Pang JL, Kantham L, Bräuner-Osborne H, Pollak M, Goltzman D, Brown EM. Interactions between calcium and phosphorus in the regulation of the production of fibroblast growth factor 23 in vivo. Am J Physiol Endocrinol Metab 2013; 304:E310-20. [PMID: 23233539 PMCID: PMC3566433 DOI: 10.1152/ajpendo.00460.2012] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 12/04/2012] [Indexed: 11/22/2022]
Abstract
Calcium and phosphorus homeostasis are highly interrelated and share common regulatory hormones, including FGF23. However, little is known about calcium's role in the regulation of FGF23. We sought to investigate the regulatory roles of calcium and phosphorus in FGF23 production using genetic mouse models with targeted inactivation of PTH (PTH KO) or both PTH and the calcium-sensing receptor (CaSR; PTH-CaSR DKO). In wild-type, PTH KO, and PTH-CaSR DKO mice, elevation of either serum calcium or phosphorus by intraperitoneal injection increased serum FGF23 levels. In PTH KO and PTH-CaSR DKO mice, however, increases in serum phosphorus by dietary manipulation were accompanied by severe hypocalcemia, which appeared to blunt stimulation of FGF23 release. Increases in dietary phosphorus in PTH-CaSR DKO mice markedly decreased serum 1,25-dihydroxyvitamin D(3) [1,25(OH)(2)D(3)] despite no change in FGF23, suggesting direct regulation of 1,25(OH)(2)D(3) synthesis by serum phosphorus. Calcium-mediated increases in serum FGF23 required a threshold level of serum phosphorus of about 5 mg/dl. Analogously, phosphorus-elicited increases in FGF23 were markedly blunted if serum calcium was less than 8 mg/dl. The best correlation between calcium and phosphorus and serum FGF23 was found between FGF23 and the calcium × phosphorus product. Since calcium stimulated FGF23 production in the PTH-CaSR DKO mice, this effect cannot be mediated by the full-length CaSR. Thus the regulation of FGF23 by both calcium and phosphorus appears to be fundamentally important in coordinating the serum levels of both mineral ions and ensuring that the calcium × phosphorus product remains within a physiological range.
Collapse
Affiliation(s)
- Stephen J Quinn
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Loupy A, Ramakrishnan SK, Wootla B, Chambrey R, de la Faille R, Bourgeois S, Bruneval P, Mandet C, Christensen EI, Faure H, Cheval L, Laghmani K, Collet C, Eladari D, Dodd RH, Ruat M, Houillier P. PTH-independent regulation of blood calcium concentration by the calcium-sensing receptor. J Clin Invest 2012; 122:3355-3367. [PMID: 22886306 PMCID: PMC3428075 DOI: 10.1172/jci57407] [Citation(s) in RCA: 157] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Accepted: 06/28/2012] [Indexed: 12/27/2022] Open
Abstract
Tight regulation of calcium levels is required for many critical biological functions. The Ca2+-sensing receptor (CaSR) expressed by parathyroid cells controls blood calcium concentration by regulating parathyroid hormone (PTH) secretion. However, CaSR is also expressed in other organs, such as the kidney, but the importance of extraparathyroid CaSR in calcium metabolism remains unknown. Here, we investigated the role of extraparathyroid CaSR using thyroparathyroidectomized, PTH-supplemented rats. Chronic inhibition of CaSR selectively increased renal tubular calcium absorption and blood calcium concentration independent of PTH secretion change and without altering intestinal calcium absorption. CaSR inhibition increased blood calcium concentration in animals pretreated with a bisphosphonate, indicating that the increase did not result from release of bone calcium. Kidney CaSR was expressed primarily in the thick ascending limb of the loop of Henle (TAL). As measured by in vitro microperfusion of cortical TAL, CaSR inhibitors increased calcium reabsorption and paracellular pathway permeability but did not change NaCl reabsorption. We conclude that CaSR is a direct determinant of blood calcium concentration, independent of PTH, and modulates renal tubular calcium transport in the TAL via the permeability of the paracellular pathway. These findings suggest that CaSR inhibitors may provide a new specific treatment for disorders related to impaired PTH secretion, such as primary hypoparathyroidism.
Collapse
Affiliation(s)
- Alexandre Loupy
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Suresh Krishna Ramakrishnan
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Bharath Wootla
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Régine Chambrey
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Renaud de la Faille
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Soline Bourgeois
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Patrick Bruneval
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Chantal Mandet
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Erik Ilso Christensen
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Hélène Faure
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Lydie Cheval
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Kamel Laghmani
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Corinne Collet
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Dominique Eladari
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Robert H. Dodd
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Martial Ruat
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| | - Pascal Houillier
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France.
Université Pierre et Marie Curie, Paris, France.
CNRS ERL7226, Paris, France.
Université Paris Descartes and Necker Hospital Kidney Transplant Department, Assistance Publique–Hôpitaux de Paris, Paris, France.
Hôpital Européen Georges Pompidou, Département de Physiologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
University of Zürich, Institute of Physiology, Zurich, Switzerland.
Hôpital Européen Georges Pompidou, Département de Pathologie, Assistance Publique–Hôpitaux de Paris, Paris, France.
Department of Anatomy, University of Århus, Århus, Denmark.
Signal Transduction and Developmental Neuropharmacology group, CNRS, UPR-3294, N&D Laboratory, Institut Albert Fessard, Gif-sur-Yvette, France.
Hôpital Lariboisière, Biochimie et Biologie Moleculaire, Assistance Publique–Hôpitaux de Paris, Paris, France.
Institut de Chimie des Substances Naturelles, CNRS, Gif-sur-Yvette, France
| |
Collapse
|
21
|
Loupy A, Ramakrishnan SK, Wootla B, Chambrey R, de la Faille R, Bourgeois S, Bruneval P, Mandet C, Christensen EI, Faure H, Cheval L, Laghmani K, Collet C, Eladari D, Dodd RH, Ruat M, Houillier P. PTH-independent regulation of blood calcium concentration by the calcium-sensing receptor. J Clin Invest 2012. [PMID: 22886306 DOI: 10.1172/jci57407.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Tight regulation of calcium levels is required for many critical biological functions. The Ca2+-sensing receptor (CaSR) expressed by parathyroid cells controls blood calcium concentration by regulating parathyroid hormone (PTH) secretion. However, CaSR is also expressed in other organs, such as the kidney, but the importance of extraparathyroid CaSR in calcium metabolism remains unknown. Here, we investigated the role of extraparathyroid CaSR using thyroparathyroidectomized, PTH-supplemented rats. Chronic inhibition of CaSR selectively increased renal tubular calcium absorption and blood calcium concentration independent of PTH secretion change and without altering intestinal calcium absorption. CaSR inhibition increased blood calcium concentration in animals pretreated with a bisphosphonate, indicating that the increase did not result from release of bone calcium. Kidney CaSR was expressed primarily in the thick ascending limb of the loop of Henle (TAL). As measured by in vitro microperfusion of cortical TAL, CaSR inhibitors increased calcium reabsorption and paracellular pathway permeability but did not change NaCl reabsorption. We conclude that CaSR is a direct determinant of blood calcium concentration, independent of PTH, and modulates renal tubular calcium transport in the TAL via the permeability of the paracellular pathway. These findings suggest that CaSR inhibitors may provide a new specific treatment for disorders related to impaired PTH secretion, such as primary hypoparathyroidism.
Collapse
Affiliation(s)
- Alexandre Loupy
- INSERM UMRS 872, Centre de Recherche des Cordeliers, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Battula S, Hao S, Pedraza PL, Stier CT, Ferreri NR. Tumor necrosis factor-alpha induces renal cyclooxygenase-2 expression in response to hypercalcemia. Prostaglandins Other Lipid Mediat 2012; 99:45-50. [PMID: 22800939 DOI: 10.1016/j.prostaglandins.2012.07.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2012] [Revised: 06/29/2012] [Accepted: 07/06/2012] [Indexed: 01/01/2023]
Abstract
The effect of tumor necrosis factor-alpha (TNF) on cyclooxygenase-2 (COX-2) expression in the renal outer medulla (OM) was determined in a model of dihydrotachysterol (DHT)-induced hypercalcemia. Increases in serum calcium and water intake were observed during ingestion of a DHT-containing diet in both wild type (WT) and TNF deficient mice (TNF(-/-)). Polyuria and a decrease in body weight were observed in response to DHT treatment in WT and TNF(-/-) mice. A transient elevation in urinary TNF was observed in WT mice treated with DHT. Moreover, increased urinary levels of prostaglandin E(2) (PGE(2)) and a corresponding increase in COX-2 expression in the OM were observed in WT mice fed DHT. Increased COX-2 expression was not observed in TNF(-/-) mice fed DHT, and the characteristics of PGE(2) synthesis were distinct from those in WT mice. This study demonstrates that COX-2 expression in the OM, secondary to hypercalemia, is TNF-dependent.
Collapse
Affiliation(s)
- Sailaja Battula
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, United States
| | | | | | | | | |
Collapse
|
23
|
The role of the calcium-sensing receptor in human disease. Clin Biochem 2012; 45:943-53. [PMID: 22503956 DOI: 10.1016/j.clinbiochem.2012.03.034] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 03/22/2012] [Accepted: 03/27/2012] [Indexed: 01/18/2023]
Abstract
Following the discovery of the calcium-sensing receptor (CaSR) in 1993, its pivotal role in disorders of calcium homeostasis such as Familial Hypocalciuric Hypercalcemia (FHH) was quickly demonstrated. Since then, it has become clear that the CaSR has immense functional versatility largely through its ability to activate many different signaling pathways in a ligand- and tissue-specific manner. This allows the receptor to play diverse and crucial roles in human physiology and pathophysiology, both in calcium homeostasis and in tissues and biological processes unrelated to calcium balance. This review covers current knowledge of the role of the CaSR in disorders of calcium homeostasis (FHH, neonatal severe hyperparathyroidism, autosomal dominant hypocalcemia, primary and secondary hyperparathyroidism, hypercalcemia of malignancy) as well as unrelated diseases such as breast and colorectal cancer (where the receptor appears to play a tumor suppressor role), Alzheimer's disease, pancreatitis, diabetes mellitus, hypertension and bone and gastrointestinal disorders. In addition, it examines the use or potential use of CaSR agonists or antagonists (calcimimetics and calcilytics) and other drugs mediated through the CaSR, in the management of disorders as diverse as hyperparathyroidism, osteoporosis and gastrointestinal disease.
Collapse
|
24
|
Bozic M, Valdivielso JM. Calcium signaling in renal tubular cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:933-44. [PMID: 22453977 DOI: 10.1007/978-94-007-2888-2_42] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The kidney handles calcium by filtration and reabsorption. About 60% of the plasma calcium is filterable, and 99% is reabsorbed in the tubule. In the proximal tubule, the reabsorption is passive and paracellular, but in the distal tubule is active and transcellular. Thus, renal tubular cells are exposed to very high concentrations of calcium in both, the extracellular and the intracellular compartments. Extracellular calcium signaling is transmitted by the calcium sensing receptor, located both in the luminal and basolateral sides of tubular cells. This receptor is able to control levels of extracellular calcium and acts in consequence to maintain calcium homeostasis. Furthermore, renal tubular cells possess several calcium channels that regulate some of the cell functions. Among those, voltage gated calcium channels, transient receptor potential channels and N-methyl-D-aspartate receptor channels have been reported to control several functions. Those functions include survival, apoptosis, differentiation, epithelial-mesenchymal transition, and active vitamin D and renin synthesis.
Collapse
Affiliation(s)
- Milica Bozic
- Nephrology Research Laboratory, IRB Lleida, University Hospital Arnau de Vilanova, Rovira Roure 80, Planta 1, 25198 Lleida, Spain.
| | | |
Collapse
|
25
|
Messa P, Alfieri C, Brezzi B. Clinical utilization of cinacalcet in hypercalcemic conditions. Expert Opin Drug Metab Toxicol 2011; 7:517-28. [PMID: 21361849 DOI: 10.1517/17425255.2011.562196] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Cinacalcet has recently been introduced as a treatment for secondary hyperparathyroidism in dialysis patients and for parathyroid carcinoma. However, there has been an increasing interest in finding out whether cinacalcet can be used as a treatment for other parathyroid hormone (PTH)-dependent hypercalcemic conditions also. AREAS COVERED The article reports the most relevant recent contributions dealing with calcium sensing receptor (CaSR) physiology as well as cinacalcet pharmacokinetics and pharmacodynamics. It also looks at the different hypercalcemic conditions where the use of cinacalcet has been proposed. This article was researched using clinical trials, case reports and outstanding basic research published in the last 3 years (MEDLINE database up to 31 November 2010). It provides the reader with an insight into the many unaddressed issues regarding cinacalcet that need to be resolved before it can be used in newly proposed fields. EXPERT OPINION Since cinacalcet may not only have an effect on parathyroid CaSR but also on CaSR expressed at bone and renal levels, it can currently only be considered a good alternative to parathyroidectomy in PTH-dependent hypercalcemic conditions when surgical intervention is burdened by a high failure rate or when it can be considered a risky procedure. At present, cinacalcet cannot be considered the first choice treatment in asymptomatic primary hyperparathyroidism or in mild-to-moderate forms of familial hypocalciuric hypocalcemia.
Collapse
Affiliation(s)
- Piergiorgio Messa
- Division of Nephrology, Dialysis, and Renal Transplant, Fondazione Ca' Granda-IRCCS, OspedaleMaggiore-Policlinico, v. Commenda 15, 20122 Milano, Italy.
| | | | | |
Collapse
|
26
|
Riccardi D, Brown EM. Physiology and pathophysiology of the calcium-sensing receptor in the kidney. Am J Physiol Renal Physiol 2010; 298:F485-99. [PMID: 19923405 PMCID: PMC2838589 DOI: 10.1152/ajprenal.00608.2009] [Citation(s) in RCA: 235] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Accepted: 11/13/2009] [Indexed: 12/21/2022] Open
Abstract
The extracellular calcium-sensing receptor (CaSR) plays a major role in the maintenance of a physiological serum ionized calcium (Ca2+) concentration by regulating the circulating levels of parathyroid hormone. It was molecularly identified in 1993 by Brown et al. in the laboratory of Dr. Steven Hebert with an expression cloning strategy. Subsequent studies have demonstrated that the CaSR is highly expressed in the kidney, where it is capable of integrating signals deriving from the tubular fluid and/or the interstitial plasma. Additional studies elucidating inherited and acquired mutations in the CaSR gene, the existence of activating and inactivating autoantibodies, and genetic polymorphisms of the CaSR have greatly enhanced our understanding of the role of the CaSR in mineral ion metabolism. Allosteric modulators of the CaSR are the first drugs in their class to become available for clinical use and have been shown to treat successfully hyperparathyroidism secondary to advanced renal failure. In addition, preclinical and clinical studies suggest the possibility of using such compounds in various forms of hypercalcemic hyperparathyroidism, such as primary and lithium-induced hyperparathyroidism and that occurring after renal transplantation. This review addresses the role of the CaSR in kidney physiology and pathophysiology as well as current and in-the-pipeline treatments utilizing CaSR-based therapeutics.
Collapse
Affiliation(s)
- Daniela Riccardi
- School of Biosciences, Cardiff University, Cardiff, United Kingdom.
| | | |
Collapse
|