1
|
Pereira RS, Kumar R, Cais A, Paulini L, Kahler A, Bravo J, Minciacchi VR, Krack T, Kowarz E, Zanetti C, Godavarthy PS, Hoeller F, Llavona P, Stark T, Tascher G, Nowak D, Meduri E, Huntly BJP, Münch C, Pampaloni F, Marschalek R, Krause DS. Distinct and targetable role of calcium-sensing receptor in leukaemia. Nat Commun 2023; 14:6242. [PMID: 37802982 PMCID: PMC10558580 DOI: 10.1038/s41467-023-41770-0] [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: 09/30/2022] [Accepted: 09/12/2023] [Indexed: 10/08/2023] Open
Abstract
Haematopoietic stem cells (HSC) reside in the bone marrow microenvironment (BMM), where they respond to extracellular calcium [eCa2+] via the G-protein coupled calcium-sensing receptor (CaSR). Here we show that a calcium gradient exists in this BMM, and that [eCa2+] and response to [eCa2+] differ between leukaemias. CaSR influences the location of MLL-AF9+ acute myeloid leukaemia (AML) cells within this niche and differentially impacts MLL-AF9+ AML versus BCR-ABL1+ leukaemias. Deficiency of CaSR reduces AML leukaemic stem cells (LSC) 6.5-fold. CaSR interacts with filamin A, a crosslinker of actin filaments, affects stemness-associated factors and modulates pERK, β-catenin and c-MYC signaling and intracellular levels of [Ca2+] in MLL-AF9+ AML cells. Combination treatment of cytarabine plus CaSR-inhibition in various models may be superior to cytarabine alone. Our studies suggest CaSR to be a differential and targetable factor in leukaemia progression influencing self-renewal of AML LSC via [eCa2+] cues from the BMM.
Collapse
Affiliation(s)
- Raquel S Pereira
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Rahul Kumar
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Alessia Cais
- Pediatric Neurooncology, Hopp Children's Cancer Center Heidelberg (KiTZ) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lara Paulini
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Alisa Kahler
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Jimena Bravo
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Valentina R Minciacchi
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Theresa Krack
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Eric Kowarz
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Costanza Zanetti
- University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
| | | | - Fabian Hoeller
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Pablo Llavona
- Institute of Molecular Biology gGmbH (IMB), Mainz, Germany
| | - Tabea Stark
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Georg Tascher
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Daniel Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Eshwar Meduri
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Brian J P Huntly
- Department of Haematology, University of Cambridge, Cambridge, United Kingdom
| | - Christian Münch
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany
| | - Francesco Pampaloni
- Buchmann Institute for Molecular Life Sciences (BMLS, CEF-MC), Goethe University, Frankfurt am Main, Germany
| | - Rolf Marschalek
- Institute of Pharmaceutical Biology, Goethe University, Frankfurt am Main, Germany
| | - Daniela S Krause
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany.
- Institute of Biochemistry II, Faculty of Medicine, Goethe University, Frankfurt am Main, Germany.
- Institute of General Pharmacology and Toxicology, Goethe-University, Frankfurt am Main, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
- Frankfurt Cancer Institute, Frankfurt, Germany.
| |
Collapse
|
2
|
Calcium-Sensing Receptor (CaSR)-Mediated Intracellular Communication in Cardiovascular Diseases. Cells 2022; 11:cells11193075. [PMID: 36231037 PMCID: PMC9562006 DOI: 10.3390/cells11193075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/31/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
The calcium-sensing receptor (CaSR), a G-protein-coupled receptor (GPCR), is a cell-surface-located receptor that can induce highly diffusible messengers (IP3, Ca2+, cAMP) in the cytoplasm to activate various cellular responses. Recently, it has also been suggested that the CaSR mediates the intracellular communications between the endoplasmic reticulum (ER), mitochondria, nucleus, protease/proteasome, and autophagy-lysosome, which are involved in related cardiovascular diseases. The complex intracellular signaling of this receptor challenges it as a valuable therapeutic target. It is, therefore, necessary to understand the mechanisms behind the signaling characteristics of this receptor in intracellular communication. This review provides an overview of the recent research progress on the various regulatory mechanisms of the CaSR in related cardiovascular diseases and the heart-kidney interaction; the associated common causes are also discussed.
Collapse
|
3
|
Tuffour A, Kosiba AA, Zhang Y, Peprah FA, Gu J, Shi H. Role of the calcium-sensing receptor (CaSR) in cancer metastasis to bone: Identifying a potential therapeutic target. Biochim Biophys Acta Rev Cancer 2021; 1875:188528. [PMID: 33640382 DOI: 10.1016/j.bbcan.2021.188528] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 02/16/2021] [Accepted: 02/20/2021] [Indexed: 12/20/2022]
Abstract
Cancer is a major cause of morbidity and mortality worldwide due to its ability to evade immune surveillance and metastasize from its origin to a secondary point of contact. Though several treatment techniques have been developed to suppress or manage cancer spread, a strategy for total control over the disease continues to evade researchers. In considering ways to control or prevent cancer from metastasizing to the bone, we analyze the impact of the calcium-sensing receptor (CaSR), whose primary role is to maintain calcium (Ca2+) homeostasis in cellular and systemic physiological processes. CaSR is a pleiotropic receptor capable of enhancing the proliferation of some cancers such as breast, lung, prostate and kidney cancers at its primary site(s) and stimulating bone metastasis, while exerting a suppressive effect in others such as colon cancer. The activity of CaSR not only increases cancer cell proliferation, migration and suppression of apoptosis in the organs indicated, but also increases the secretion of parathyroid hormone-related protein (PTHrP) and epiregulin, which induce osteolytic activity and osteoblastic suppression. In addition, released cytokines and Ca2+ from bone resorption are critical factors that further promote cancer proliferation. In this review, we seek to highlight previous viewpoints on CaSR, discuss its role in a new context, and consider its potential clinical application in cancer treatment.
Collapse
Affiliation(s)
- Alex Tuffour
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| | | | - Yao Zhang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Frank Addai Peprah
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Jie Gu
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Haifeng Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China.
| |
Collapse
|
4
|
Leach K, Hannan FM, Josephs TM, Keller AN, Møller TC, Ward DT, Kallay E, Mason RS, Thakker RV, Riccardi D, Conigrave AD, Bräuner-Osborne H. International Union of Basic and Clinical Pharmacology. CVIII. Calcium-Sensing Receptor Nomenclature, Pharmacology, and Function. Pharmacol Rev 2020; 72:558-604. [PMID: 32467152 PMCID: PMC7116503 DOI: 10.1124/pr.119.018531] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The calcium-sensing receptor (CaSR) is a class C G protein-coupled receptor that responds to multiple endogenous agonists and allosteric modulators, including divalent and trivalent cations, L-amino acids, γ-glutamyl peptides, polyamines, polycationic peptides, and protons. The CaSR plays a critical role in extracellular calcium (Ca2+ o) homeostasis, as demonstrated by the many naturally occurring mutations in the CaSR or its signaling partners that cause Ca2+ o homeostasis disorders. However, CaSR tissue expression in mammals is broad and includes tissues unrelated to Ca2+ o homeostasis, in which it, for example, regulates the secretion of digestive hormones, airway constriction, cardiovascular effects, cellular differentiation, and proliferation. Thus, although the CaSR is targeted clinically by the positive allosteric modulators (PAMs) cinacalcet, evocalcet, and etelcalcetide in hyperparathyroidism, it is also a putative therapeutic target in diabetes, asthma, cardiovascular disease, and cancer. The CaSR is somewhat unique in possessing multiple ligand binding sites, including at least five putative sites for the "orthosteric" agonist Ca2+ o, an allosteric site for endogenous L-amino acids, two further allosteric sites for small molecules and the peptide PAM, etelcalcetide, and additional sites for other cations and anions. The CaSR is promiscuous in its G protein-coupling preferences, and signals via Gq/11, Gi/o, potentially G12/13, and even Gs in some cell types. Not surprisingly, the CaSR is subject to biased agonism, in which distinct ligands preferentially stimulate a subset of the CaSR's possible signaling responses, to the exclusion of others. The CaSR thus serves as a model receptor to study natural bias and allostery. SIGNIFICANCE STATEMENT: The calcium-sensing receptor (CaSR) is a complex G protein-coupled receptor that possesses multiple orthosteric and allosteric binding sites, is subject to biased signaling via several different G proteins, and has numerous (patho)physiological roles. Understanding the complexities of CaSR structure, function, and biology will aid future drug discovery efforts seeking to target this receptor for a diversity of diseases. This review summarizes what is known to date regarding key structural, pharmacological, and physiological features of the CaSR.
Collapse
Affiliation(s)
- Katie Leach
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Fadil M Hannan
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Tracy M Josephs
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Andrew N Keller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Thor C Møller
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Donald T Ward
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Enikö Kallay
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Rebecca S Mason
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Rajesh V Thakker
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Daniela Riccardi
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Arthur D Conigrave
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| | - Hans Bräuner-Osborne
- Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Australia (K.L., T.M.J., A.N.K.); Nuffield Department of Women's & Reproductive Health (F.M.H.) and Academic Endocrine Unit, Radcliffe Department of Clinical Medicine (F.M.H., R.V.T.), University of Oxford, Oxford, United Kingdom; Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark (T.C.M., H.B.-O.); Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom (D.T.W.); Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria (E.K.); Physiology, School of Medical Sciences and Bosch Institute (R.S.M.) and School of Life & Environmental Sciences, Charles Perkins Centre (A.D.C.), University of Sydney, Sydney, Australia; and School of Biosciences, Cardiff University, Cardiff, United Kingdom (D.R.)
| |
Collapse
|
5
|
Li J, Liao P, Wang K, Miao Z, Xiao R, Zhu L, Hu Q. Calcium Sensing Receptor Inhibits Growth of Human Lung Adenocarcinoma Possibly via the GSK3β/Cyclin D1 Pathway. Front Cell Dev Biol 2020; 8:446. [PMID: 32671062 PMCID: PMC7330125 DOI: 10.3389/fcell.2020.00446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/13/2020] [Indexed: 12/12/2022] Open
Abstract
The effect of calcium sensing receptor (CaSR) on tumor cell proliferation has been studied in several human cancers, and great discrepancies were found in different tumors. However, the role of CaSR in lung adenocarcinomas (LUADs) is not clear. Therefore, we investigated the function of CaSR on regulating the growth of human LUAD and its possible mechanism. The expression of CaSR protein and its relationship with pathological parameters were examined in paraffin sections from 51 LUAD patients, by immunohistochemistry. The results showed that CasR expression was negatively correlated with the Ki-67 index as well as the grade of malignancy in LUAD. Further, CaSR demonstrated an in vitro inhibitory effect on the proliferation of human LUAD A549 cells by regulating CaSR activity with agonist cinacalcet, antagonist NPS2143, or shRNA-CaSR transfection. Tumor xenograft models also verified the in vivo proliferation-inhibiting role of CaSR by subcutaneous injecting A549 cells into nude mice with or without changes of CaSR activity. Molecularly, Western blotting showed that CaSR positively regulated the activity of glycogen synthase kinase 3β (GSK3β), followed by the downregulation of Cyclin D1. We used the dominant negative mutant and the constitutively active mutant plasmid of GSK3β to alter GSK3β activity. Our functional experiments showed that the proliferation-inhibition of CaSR was suppressed by the inactivation of GSK3β and enhanced by the activation of GSK3β. These results suggested that CaSR played a proliferation-inhibiting role in LUAD, at least partially by regulating the GSK3β/Cyclin D1 pathway.
Collapse
Affiliation(s)
- Jiansha Li
- Institute of Pathology, Tongji Hospital, Wuhan, China
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Wuhan, China
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pu Liao
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Wuhan, China
- Department of Pathology, Union Hospital, Wuhan, China
| | - Kun Wang
- Department of Nephrology, Tongji Hospital, Wuhan, China
| | | | - Rui Xiao
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Liping Zhu
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| | - Qinghua Hu
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Wuhan, China
- Department of Pathophysiology, School of Basic Medicine, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Yang X, Lou J, Shan W, Hu Y, Du Q, Liao Q, Xie R, Xu J. Pathogenic roles of altered calcium channels and transporters in colon tumorogenesis. Life Sci 2019; 239:116909. [DOI: 10.1016/j.lfs.2019.116909] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 09/12/2019] [Accepted: 09/23/2019] [Indexed: 01/26/2023]
|
7
|
Wang W, Yu S, Huang S, Deng R, Ding Y, Wu Y, Li X, Wang A, Wang S, Chen W, Lu Y. A Complex Role for Calcium Signaling in Colorectal Cancer Development and Progression. Mol Cancer Res 2019; 17:2145-2153. [PMID: 31366605 DOI: 10.1158/1541-7786.mcr-19-0429] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/27/2019] [Accepted: 07/29/2019] [Indexed: 11/16/2022]
Abstract
Clinical data suggest that many malignant cancers are associated with hypercalcemia. Hypercalcemia can facilitate the proliferation and metastasis of gastric and colon tumors, and has been considered a hallmark of end-stage disease. However, it has also been reported that dietary calcium or vitamin D supplementation could reduce the risk of many types of cancers. In particular, the intestines can absorb considerable amounts of calcium via Ca2+-permeable ion channels, and hypercalcemia is common in patients with colorectal cancer. Thus, this review considers the role of calcium signaling in the context of colorectal cancer and summarizes the functions of specific regulators of cellular calcium levels in the proliferation, invasion, metastasis, cell death, and drug resistance of colorectal cancer cells. The data reveal that even a slight upregulation of intracellular Ca2+ signaling can facilitate the onset and progression of colorectal cancer, while continuous Ca2+ influx and Ca2+ overload may cause tumor cell death. This dual function of Ca2+ signaling adds nuance to the debate over the hallmarks of colorectal cancer, and may even provide new directions and strategies for clinical interventions.
Collapse
Affiliation(s)
- Wei Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Suyun Yu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Shuai Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Rui Deng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Yushi Ding
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Yuanyuan Wu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P.R. China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P.R. China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P.R. China.,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Shijun Wang
- Shandong Co-Innovation Center of TCM Formula, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Shandong, P.R. China
| | - Wenxing Chen
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P.R. China. .,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, P.R. China. .,Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| |
Collapse
|
8
|
Amable G, Martínez-León E, Picco ME, Di Siervi N, Davio C, Rozengurt E, Rey O. Metformin inhibits β-catenin phosphorylation on Ser-552 through an AMPK/PI3K/Akt pathway in colorectal cancer cells. Int J Biochem Cell Biol 2019; 112:88-94. [PMID: 31082618 DOI: 10.1016/j.biocel.2019.05.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 02/08/2023]
Abstract
Several epidemiologic studies have revealed strong inverse associations between metformin use and risk of colorectal cancer development. Nevertheless, the underlying mechanisms are still uncertain. The Wnt/β-catenin pathway, which plays a central role in intestinal homeostasis and sporadic colorectal cancer development, is regulated by phosphorylation cascades that are dependent and independent of Wnt. Here we report that a non-canonical Ser552 phosphorylation in β-catenin, which promotes its nuclear accumulation and transcriptional activity, is blocked by metformin via AMPK-mediated PI3K/Akt signaling inhibition.
Collapse
Affiliation(s)
- Gastón Amable
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín", Caba, 1120, Argentina
| | - Eduardo Martínez-León
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín", Caba, 1120, Argentina
| | - María Elisa Picco
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín", Caba, 1120, Argentina
| | - Nicolas Di Siervi
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Investigaciones Farmacológicas, Facultad de Farmacia y Bioquímica, Argentina
| | - Carlos Davio
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Investigaciones Farmacológicas, Facultad de Farmacia y Bioquímica, Argentina; Departamento de Farmacología, Caba, 1113, Argentina
| | - Enrique Rozengurt
- Unit of Signal Transduction and Gastrointestinal Cancer, Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, Molecular Biology Institute and Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, CA, 90095-1786, USA
| | - Osvaldo Rey
- Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad de Buenos Aires, Argentina; Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín", Caba, 1120, Argentina.
| |
Collapse
|
9
|
Martínez-León E, Amable G, Jácamo R, Picco ME, Anaya L, Rozengurt E, Rey O. Protein kinase D1 inhibition interferes with mitosis progression. J Cell Physiol 2019; 234:20510-20519. [PMID: 30997696 DOI: 10.1002/jcp.28651] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/20/2019] [Accepted: 03/25/2019] [Indexed: 12/23/2022]
Abstract
Protein kinase D1 (PKD1) plays a vital role in signal transduction, cell proliferation, membrane trafficking, and cancer; however, the majority of the studies up to date had centered primarily on PKD1 functions in interphase, very little is known about its role during cell division. We previously demonstrated that during mitosis PKD1 is activated and associated with centrosomes, spindles, and midbodies. However, these observations did not address whether PKD1 was associated with mitosis regulation. Accordingly, we used rapidly acting PKD-specific inhibitors to examine the contribution of PKD1 the sequence of events in mitosis. We found that although PKD1 overexpression did not affect mitosis progression, suppression of its catalytic activity by two structurally unrelated inhibitors (kb NB 142-70 and CRT 0066101) induced a significant delay in metaphase to anaphase transition time. PKD1 inhibition during mitosis also produced the appearance of abnormal spindles, defects in chromosome alignment, and segregation as well as apoptosis. Thus, these observations indicate that PKD1 activity is associated with mitosis regulation.
Collapse
Affiliation(s)
- Eduardo Martínez-León
- Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín," Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA), Buenos Aires, Argentina
| | - Gastón Amable
- Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín," Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA), Buenos Aires, Argentina
| | - Rodrigo Jácamo
- Section of Molecular Hematology and Therapy, Department of Stem Cell Transplantation and Cellular Therapy, M.D. Anderson Cancer Center, University of Texas, Houston, TX
| | - María Elisa Picco
- Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín," Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA), Buenos Aires, Argentina
| | - Laura Anaya
- División de Hematología, Hospital de Clínicas "José de San Martín," CABA, Buenos Aires, Argentina
| | - Enrique Rozengurt
- Unit of Signal Transduction and Gastrointestinal Cancer, Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, Molecular Biology Institute and Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA
| | - Osvaldo Rey
- Consejo Nacional de Investigaciones Científicas y Técnicas, Instituto de Inmunología, Genética y Metabolismo, Facultad de Farmacia y Bioquímica, Hospital de Clínicas "José de San Martín," Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires (CABA), Buenos Aires, Argentina
| |
Collapse
|
10
|
Hannan FM, Kallay E, Chang W, Brandi ML, Thakker RV. The calcium-sensing receptor in physiology and in calcitropic and noncalcitropic diseases. Nat Rev Endocrinol 2018; 15:33-51. [PMID: 30443043 PMCID: PMC6535143 DOI: 10.1038/s41574-018-0115-0] [Citation(s) in RCA: 199] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The Ca2+-sensing receptor (CaSR) is a dimeric family C G protein-coupled receptor that is expressed in calcitropic tissues such as the parathyroid glands and the kidneys and signals via G proteins and β-arrestin. The CaSR has a pivotal role in bone and mineral metabolism, as it regulates parathyroid hormone secretion, urinary Ca2+ excretion, skeletal development and lactation. The importance of the CaSR for these calcitropic processes is highlighted by loss-of-function and gain-of-function CaSR mutations that cause familial hypocalciuric hypercalcaemia and autosomal dominant hypocalcaemia, respectively, and also by the fact that alterations in parathyroid CaSR expression contribute to the pathogenesis of primary and secondary hyperparathyroidism. Moreover, the CaSR is an established therapeutic target for hyperparathyroid disorders. The CaSR is also expressed in organs not involved in Ca2+ homeostasis: it has noncalcitropic roles in lung and neuronal development, vascular tone, gastrointestinal nutrient sensing, wound healing and secretion of insulin and enteroendocrine hormones. Furthermore, the abnormal expression or function of the CaSR is implicated in cardiovascular and neurological diseases, as well as in asthma, and the CaSR is reported to protect against colorectal cancer and neuroblastoma but increase the malignant potential of prostate and breast cancers.
Collapse
Affiliation(s)
- Fadil M Hannan
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Enikö Kallay
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Wenhan Chang
- Endocrine Research Unit, Veterans Affairs Medical Center, University of California, San Francisco, San Francisco, CA, USA
| | - Maria Luisa Brandi
- Metabolic Bone Diseases Unit, Department of Surgery and Translational Medicine, University of Florence, Florence, Italy.
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
11
|
Iamartino L, Elajnaf T, Kallay E, Schepelmann M. Calcium-sensing receptor in colorectal inflammation and cancer: Current insights and future perspectives. World J Gastroenterol 2018; 24:4119-4131. [PMID: 30271078 PMCID: PMC6158479 DOI: 10.3748/wjg.v24.i36.4119] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/11/2018] [Accepted: 08/01/2018] [Indexed: 02/06/2023] Open
Abstract
The extracellular calcium-sensing receptor (CaSR) is best known for its action in the parathyroid gland and kidneys where it controls body calcium homeostasis. However, the CaSR has different roles in the gastrointestinal tract, where it is ubiquitously expressed. In the colon, the CaSR is involved in controlling multiple mechanisms, including fluid transport, inflammation, cell proliferation and differentiation. Although the expression pattern and functions of the CaSR in the colonic microenvironment are far from being completely understood, evidence has been accumulating that the CaSR might play a protective role against both colonic inflammation and colorectal cancer. For example, CaSR agonists such as dipeptides have been suggested to reduce colonic inflammation, while dietary calcium was shown to reduce the risk of colorectal cancer. CaSR expression is lost in colonic malignancies, indicating that the CaSR is a biomarker for colonic cancer progression. This dual anti-inflammatory and anti-tumourigenic role of the CaSR makes it especially interesting in colitis-associated colorectal cancer. In this review, we describe the clinical and experimental evidence for the role of the CaSR in colonic inflammation and colorectal cancer, the intracellular signalling pathways which are putatively involved in these actions, and the possibilities to exploit these actions of the CaSR for future therapies of colonic inflammation and cancer.
Collapse
Affiliation(s)
- Luca Iamartino
- Institute of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna 1090, Austria
| | - Taha Elajnaf
- Institute of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna 1090, Austria
| | - Enikö Kallay
- Institute of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna 1090, Austria
| | - Martin Schepelmann
- Institute of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna 1090, Austria
| |
Collapse
|
12
|
Calcium-sensing receptor in nutrient sensing: an insight into the modulation of intestinal homoeostasis. Br J Nutr 2018; 120:881-890. [DOI: 10.1017/s0007114518002088] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
AbstractThe animal gut effectively prevents the entry of hazardous substances and microbes while permitting the transfer of nutrients, such as water, electrolytes, vitamins, proteins, lipids, carbohydrates, minerals and microbial metabolites, which are intimately associated with intestinal homoeostasis. The gut maintains biological functions through its nutrient-sensing receptors, including the Ca-sensing receptor (CaSR), which activates a variety of signalling pathways, depending on cellular context. CaSR coordinates food digestion and nutrient absorption, promotes cell proliferation and differentiation, regulates energy metabolism and immune response, stimulates hormone secretion, mitigates secretory diarrhoea and enhances intestinal barrier function. Thus, CaSR is crucial to the maintenance of gut homoeostasis and protection of intestinal health. In this review, we focused on the emerging roles of CaSR in the modulation of intestinal homoeostasis including related underlying mechanisms. By elucidating the relationship between CaSR and animal gut homoeostasis, effective and inexpensive methods for treating intestinal health imbalance through nutritional manipulation can be developed. This article is expected to provide experimental data of the effects of CaSR on animal or human health.
Collapse
|
13
|
Tang L, Jiang L, McIntyre ME, Petrova E, Cheng SX. Calcimimetic acts on enteric neuronal CaSR to reverse cholera toxin-induced intestinal electrolyte secretion. Sci Rep 2018; 8:7851. [PMID: 29777154 PMCID: PMC5959902 DOI: 10.1038/s41598-018-26171-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 05/08/2018] [Indexed: 01/19/2023] Open
Abstract
Treatment of acute secretory diarrheal illnesses remains a global challenge. Enterotoxins produce secretion through direct epithelial action and indirectly by activating enteric nervous system (ENS). Using a microperfused colonic crypt technique, we have previously shown that R568, a calcimimetic that activates the calcium-sensing receptor (CaSR), can act on intestinal epithelium and reverse cholera toxin-induced fluid secretion. In the present study, using the Ussing chamber technique in conjunction with a tissue-specific knockout approach, we show that the effects of cholera toxin and CaSR agonists on electrolyte secretion by the intestine can also be attributed to opposing actions of the toxin and CaSR on the activity of the ENS. Our results suggest that targeting intestinal CaSR might represent a previously undescribed new approach for treating secretory diarrheal diseases and other conditions with ENS over-activation.
Collapse
Affiliation(s)
- Lieqi Tang
- Department of Pediatrics, University of Florida, Gainesville, FL, 32610, USA
| | - Lingli Jiang
- Department of Pediatrics, University of Florida, Gainesville, FL, 32610, USA
| | - Megan E McIntyre
- Department of Pediatrics, University of Florida, Gainesville, FL, 32610, USA
| | - Ekaterina Petrova
- Department of Pediatrics, University of Florida, Gainesville, FL, 32610, USA
| | - Sam X Cheng
- Department of Pediatrics, University of Florida, Gainesville, FL, 32610, USA. .,Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, University of Florida, Gainesville, FL, 32610, USA.
| |
Collapse
|
14
|
Sun X, Tang L, Winesett S, Chang W, Cheng SX. Calcimimetic R568 inhibits tetrodotoxin-sensitive colonic electrolyte secretion and reduces c-fos expression in myenteric neurons. Life Sci 2017; 194:49-58. [PMID: 29247746 DOI: 10.1016/j.lfs.2017.12.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Revised: 12/08/2017] [Accepted: 12/12/2017] [Indexed: 12/13/2022]
Abstract
AIMS Calcium-sensing receptor (CaSR) is expressed on neurons of both submucosal and myenteric plexuses of the enteric nervous system (ENS) and the CaSR agonist R568 inhibited Cl- secretion in intestine. The purpose of this study was to localize the primary site of action of R568 in the ENS and to explore how CaSR regulates secretion through the ENS. MATERIALS AND METHODS Two preparations of rat proximal and distal colon were used. The full-thickness preparation contained both the submucosal and myenteric plexuses, whereas for the "stripped" preparation the myenteric plexus with the muscle layers was removed. Both preparations were mounted onto Ussing chambers and Cl- secretory responses were compared by measuring changes in short circuit current (Isc). Two tissue-specific CaSR knockouts (i.e., neuron-specific vs. enterocyte-specific) were generated to compare the effect of R568 on expression of c-fos protein in myenteric neurons by immunocytochemistry. KEY FINDINGS In full-thickness colons, tetrodotoxin (TTX) inhibited Isc, both in proximal and distal colons. A nearly identical inhibition was produced by R568. However, in stripped preparations, while the effect of TTX on Isc largely remained, the effect of R568 was nearly completely eliminated. In keeping with this, R568 reduced c-fos protein expression only in myenteric neurons of wild type mice and mutant mice that contained CaSR in neurons (i.e., villinCre/Casrflox/flox mice), but not in myenteric neurons of nestinCre/Casrflox/flox mice in which neuronal cell CaSR was eliminated. SIGNIFICANCE These results indicate that R568 exerts its anti-secretory effects predominantly via CaSR-mediated inhibition of neuronal activity in the myenteric plexus.
Collapse
Affiliation(s)
- Xiangrong Sun
- Department of Physiology and Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, China; Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Lieqi Tang
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Steven Winesett
- Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Wenhan Chang
- Endocrine Research, VA Medical Center, University of California at San Francisco, San Francisco, CA, USA
| | - Sam Xianjun Cheng
- Division of Gastroenterology, Nutrition and Hepatology, Department of Pediatrics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
15
|
Xie R, Xu J, Xiao Y, Wu J, Wan H, Tang B, Liu J, Fan Y, Wang S, Wu Y, Dong TX, Zhu MX, Carethers JM, Dong H, Yang S. Calcium Promotes Human Gastric Cancer via a Novel Coupling of Calcium-Sensing Receptor and TRPV4 Channel. Cancer Res 2017; 77:6499-6512. [PMID: 28951460 DOI: 10.1158/0008-5472.can-17-0360] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 06/19/2017] [Accepted: 09/21/2017] [Indexed: 12/18/2022]
Abstract
Although dietary calcium intake has long been recommended for disease prevention, the influence of calcium in development of cancer in the upper gastrointestinal tract has not been explored. Here, we assess the roles of calcium and calcium-sensing receptor (CaSR) in gastric cancer development. CaSR expression was enhanced in gastric cancer specimens, which positively correlated with serum calcium concentrations, tumor progression, poor survival, and male gender in gastric cancer patients. CaSR and transient receptor potential cation channel subfamily V member 4 (TRPV4) were colocalized in gastric cancer cells, and CaSR activation evoked TRPV4-mediated Ca2+ entry. Both CaSR and TRPV4 were involved in Ca2+-induced proliferation, migration, and invasion of gastric cancer cells through a Ca2+/AKT/β-catenin relay, which occurred only in gastric cancer cells or normal cells overexpressing CaSR. Tumor growth and metastasis of gastric cancer depended on CaSR in nude mice. Overall, our findings indicate that calcium may enhance expression and function of CaSR to potentially promote gastric cancer, and that targeting the novel CaSR/TRPV4/Ca2+ pathway might serve as preventive or therapeutic strategies for gastric cancer. Cancer Res; 77(23); 6499-512. ©2017 AACR.
Collapse
Affiliation(s)
- Rui Xie
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.,Department of Gastroenterology, Affiliated Hospital to Zunyi Medical College, Zunyi, China
| | - Jingyu Xu
- Department of Gastroenterology, Affiliated Hospital to Zunyi Medical College, Zunyi, China
| | - Yufeng Xiao
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jilin Wu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Hanxing Wan
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jingjing Liu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yahan Fan
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Suming Wang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yuyun Wu
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Tobias Xiao Dong
- Department of Medicine, University of California, San Diego, California
| | - Michael X Zhu
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, Texas
| | - John M Carethers
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Hui Dong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China. .,Department of Medicine, University of California, San Diego, California
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
16
|
Momen-Heravi F, Masugi Y, Qian ZR, Nishihara R, Liu L, Smith-Warner SA, Keum N, Zhang L, Tchrakian N, Nowak JA, Yang W, Ma Y, Bowden M, da Silva A, Wang M, Fuchs CS, Meyerhardt JA, Ng K, Wu K, Giovannucci E, Ogino S, Zhang X. Tumor expression of calcium sensing receptor and colorectal cancer survival: Results from the nurses' health study and health professionals follow-up study. Int J Cancer 2017; 141:2471-2479. [PMID: 28856682 DOI: 10.1002/ijc.31021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 07/16/2017] [Accepted: 08/01/2017] [Indexed: 12/11/2022]
Abstract
Although experimental evidence suggests calcium-sensing receptor (CASR) as a tumor-suppressor, the prognostic role of tumor CASR expression in colorectal carcinoma remains unclear. We hypothesized that higher tumor CASR expression might be associated with improved survival among colorectal cancer patients. We evaluated tumor expression levels of CASR by immunohistochemistry in 809 incident colorectal cancer patients within the Nurses' Health Study and the Health Professionals Follow-up Study. We used Cox proportional hazards regression models to estimate multivariable hazard ratio (HR) for the association of tumor CASR expression with colorectal cancer-specific and all-cause mortality. We adjusted for potential confounders including tumor biomarkers such as microsatellite instability, CpG island methylator phenotype, LINE-1 methylation level, expressions of PTGS2, VDR and CTNNB1 and mutations of KRAS, BRAF and PIK3CA. There were 240 colorectal cancer-specific deaths and 427 all-cause deaths. The median follow-up of censored patients was 10.8 years (interquartile range: 7.2, 15.1). Compared with patients with no or weak expression of CASR, the multivariable HRs for colorectal cancer-specific mortality were 0.80 [95% confidence interval (CI): 0.55-1.16] in patients with moderate CASR expression and 0.50 (95% CI: 0.32-0.79) in patients with intense CASR expression (p-trend = 0.003). The corresponding HRs for overall mortality were 0.85 (0.64-1.13) and 0.81 (0.58-1.12), respectively. Higher tumor CASR expression was associated with a lower risk of colorectal cancer-specific mortality. This finding needs further confirmation and if confirmed, may lead to better understanding of the role of CASR in colorectal cancer progression.
Collapse
Affiliation(s)
- Fatemeh Momen-Heravi
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.,Division of Periodontics, Section of Oral, Diagnostic and Rehabilitation Sciences, College of Dental Medicine, Columbia University, New York, NY
| | - Yohei Masugi
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Zhi Rong Qian
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Reiko Nishihara
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Food Science and Biotechnology, Dongguk University, Goyang, South Korea.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Pathology, Program in MPE Molecular Pathological Epidemiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Li Liu
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Food Science and Biotechnology, Dongguk University, Goyang, South Korea.,Department of Epidemiology and Biostatistics, and the Ministry of Education Key Lab of Environment and Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, People's Republic of China
| | - Stephanie A Smith-Warner
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Food Science and Biotechnology, Dongguk University, Goyang, South Korea.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - NaNa Keum
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Food Science and Biotechnology, Dongguk University, Goyang, South Korea
| | - Lanjing Zhang
- Department of Pathology, University Medical Center of Princeton, Plainsboro, NJ.,Department of Biological Sciences, Rutgers University, Newark, NJ.,Clinical Investigations and Precision Therapeutics Research Program, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ.,Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ
| | - Nairi Tchrakian
- Department of Pathology, St James's Hospital, Dublin, Ireland
| | - Jonathan A Nowak
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA.,Department of Pathology, Program in MPE Molecular Pathological Epidemiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Wanshui Yang
- Department of Social Science and Public Health, School of Basic Medical Science, Jiujiang University, Jiujiang, People's Republic of China
| | - Yanan Ma
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.,Department of Biostatistics and Epidemiology, School of Public Health, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Michaela Bowden
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Annacarolina da Silva
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA.,Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Molin Wang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Charles S Fuchs
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.,Division of Periodontics, Section of Oral, Diagnostic and Rehabilitation Sciences, College of Dental Medicine, Columbia University, New York, NY.,Yale Cancer Center & Smilow Cancer Hospital, New Haven, CT.,Department of Medicine, Yale School of Medicine, New Haven, CT
| | - Jeffrey A Meyerhardt
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Kimmie Ng
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Food Science and Biotechnology, Dongguk University, Goyang, South Korea
| | - Edward Giovannucci
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Food Science and Biotechnology, Dongguk University, Goyang, South Korea
| | - Shuji Ogino
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA.,Department of Pathology, Program in MPE Molecular Pathological Epidemiology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.,Department of Oncologic Pathology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Xuehong Zhang
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA.,Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
17
|
Fatty acid and mineral receptors as drug targets for gastrointestinal disorders. Future Med Chem 2017; 9:315-334. [DOI: 10.4155/fmc-2016-0205] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nutrient-sensing receptors, including fatty acid receptors (FFA1–FFA4), Ca2+-sensing receptors and Zn2+-sensing receptors, are involved in several biological processes. These receptors are abundantly expressed in the GI tract, where they have been shown to play crucial roles in regulating GI function. This review provides an overview of the GI functions of fatty acid and mineral receptors, including the regulation of gastric and enteroendocrine functions, GI motility, ion transport and cell growth. Recently, several lines of evidence have implicated these receptors as promising therapeutic targets for the treatment of GI disorders, for example, inflammatory bowel disease, colorectal cancer, metabolic syndrome and diarrheal diseases. A future perspective on drug discovery research targeting these receptors is discussed.
Collapse
|
18
|
Aggarwal A, Kállay E. Cross Talk between the Calcium-Sensing Receptor and the Vitamin D System in Prevention of Cancer. Front Physiol 2016; 7:451. [PMID: 27803671 PMCID: PMC5067519 DOI: 10.3389/fphys.2016.00451] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/21/2016] [Indexed: 12/25/2022] Open
Abstract
There is epidemiological evidence for the cancer preventive effect of dietary calcium (Ca2+) and vitamin D. This effect is strongest in colorectal cancer (CRC). The active vitamin D metabolite, 1,25-dihydroxyvitamin D3 (1,25D3), bound to its receptor, the vitamin D receptor (VDR) regulates the expression of hundreds of different genes in a cell- and tissue-specific manner. While Ca2+ acts through multiple mechanisms and pathways, some of its effects are mediated by the calcium-sensing receptor (CaSR). The joint action of Ca2+ and 1,25D3 is due to the fact that both regulate some of the main processes involved in the development of various cancers, such as proliferation, differentiation, apoptosis, migration, and inflammation. Moreover, 1,25D3, bound to VDR can induce translation of the CaSR, while the amount and activity of the CaSR affects 1,25D3 signaling. However, the complexity of the cross-talk between the CaSR and the vitamin D system goes beyond regulating similar pathways and affecting each other's expression. Our aim was to review some of the mechanisms that drive the cross-talk between the vitamin D system and the CaSR with a special focus on the interaction in CRC cells. We evaluated the molecular evidence that supports the epidemiological observation that both vitamin D and calcium are needed for protection against malignant transformation of the colon and that their effect is modulated by the presence of a functional CaSR.
Collapse
Affiliation(s)
- Abhishek Aggarwal
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of ViennaVienna, Austria; Department of Pediatrics/Endocrinology, School of Medicine, Stanford UniversityStanford, CA, USA
| | - Enikö Kállay
- Department of Pathophysiology and Allergy Research, Center of Pathophysiology, Infectiology and Immunology, Medical University of Vienna Vienna, Austria
| |
Collapse
|
19
|
|
20
|
Bikle DD, Jiang Y, Nguyen T, Oda Y, Tu CL. Disruption of Vitamin D and Calcium Signaling in Keratinocytes Predisposes to Skin Cancer. Front Physiol 2016; 7:296. [PMID: 27462278 PMCID: PMC4940389 DOI: 10.3389/fphys.2016.00296] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 06/27/2016] [Indexed: 12/13/2022] Open
Abstract
1,25 dihydroxyvitamin D (1,25(OH)2D), the active metabolite of vitamin D, and calcium regulate epidermal differentiation. 1,25(OH)2D exerts its effects through the vitamin D receptor (VDR), a transcription factor in the nuclear hormone receptor family, whereas calcium acts through the calcium sensing receptor (Casr), a membrane bound member of the G protein coupled receptor family. We have developed mouse models in which the Vdr and Casr have been deleted in the epidermis (epidVdr−∕− and epidCasr−∕−). Both genotypes show abnormalities in calcium induced epidermal differentiation in vivo and in vitro, associated with altered hedgehog (HH) and β–catenin signaling that when abnormally expressed lead to basal cell carcinomas (BCC) and trichofolliculomas, respectively. The Vdr−∕− mice are susceptible to tumor formation following UVB or chemical carcinogen exposure. More recently we found that the keratinocytes from these mice over express long non-coding RNA (lncRNA) oncogenes such as H19 and under express lncRNA tumor suppressors such as lincRNA-21. Spontaneous tumors have not been observed in either the epidVdr−∕− or epidCasr−∕−. But in mice with epidermal specific deletion of both Vdr and Casr (epidVdr−∕−/epidCasr−∕− [DKO]) tumor formation occurs spontaneously when the DKO mice are placed on a low calcium diet. These results demonstrate important interactions between vitamin D and calcium signaling through their respective receptors that lead to cancer when these signals are disrupted. The roles of the β–catenin, hedgehog, and lncRNA pathways in predisposing the epidermis to tumor formation when vitamin D and calcium signaling are disrupted will be discussed.
Collapse
Affiliation(s)
- Daniel D Bikle
- Departments of Medicine and Dermatology, VA Medical Center and University of California, San Francisco San Francisco, CA, USA
| | - Yan Jiang
- Departments of Medicine and Dermatology, VA Medical Center and University of California, San Francisco San Francisco, CA, USA
| | - Thai Nguyen
- Departments of Medicine and Dermatology, VA Medical Center and University of California, San Francisco San Francisco, CA, USA
| | - Yuko Oda
- Departments of Medicine and Dermatology, VA Medical Center and University of California, San Francisco San Francisco, CA, USA
| | - Chia-Ling Tu
- Departments of Medicine and Dermatology, VA Medical Center and University of California, San Francisco San Francisco, CA, USA
| |
Collapse
|
21
|
Wang J, Sinnett-Smith J, Stevens JV, Young SH, Rozengurt E. Biphasic Regulation of Yes-associated Protein (YAP) Cellular Localization, Phosphorylation, and Activity by G Protein-coupled Receptor Agonists in Intestinal Epithelial Cells: A NOVEL ROLE FOR PROTEIN KINASE D (PKD). J Biol Chem 2016; 291:17988-8005. [PMID: 27369082 DOI: 10.1074/jbc.m115.711275] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Indexed: 12/14/2022] Open
Abstract
We examined the regulation of Yes-associated protein (YAP) localization, phosphorylation, and transcriptional activity in intestinal epithelial cells. Our results show that stimulation of intestinal epithelial IEC-18 cells with the G protein-coupled receptor (GPCR) agonist angiotensin II, a potent mitogen for these cells, induced rapid translocation of YAP from the nucleus to the cytoplasm (within 15 min) and a concomitant increase in YAP phosphorylation at Ser(127) and Ser(397) Angiotensin II elicited YAP phosphorylation and cytoplasmic accumulation in a dose-dependent manner (ED50 = 0.3 nm). Similar YAP responses were provoked by stimulation with vasopressin or serum. Treatment of the cells with the protein kinase D (PKD) family inhibitors CRT0066101 and kb NB 142-70 prevented the increase in YAP phosphorylation on Ser(127) and Ser(397) via Lats2, YAP cytoplasmic accumulation, and increase in the mRNA levels of YAP/TEAD-regulated genes (Ctgf and Areg). Furthermore, siRNA-mediated knockdown of PKD1, PKD2, and PKD3 markedly attenuated YAP nuclear-cytoplasmic shuttling, phosphorylation at Ser(127), and induction of Ctgf and Areg expression in response to GPCR activation. These results identify a novel role for the PKD family in the control of biphasic localization, phosphorylation, and transcriptional activity of YAP in intestinal epithelial cells. In turn, YAP and TAZ are necessary for the stimulation of the proliferative response of intestinal epithelial cells to GPCR agonists that act via PKD. The discovery of interaction between YAP and PKD pathways identifies a novel cross-talk in signal transduction and demonstrates, for the first time, that the PKDs feed into the YAP pathway.
Collapse
Affiliation(s)
- Jia Wang
- From the Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine
| | - James Sinnett-Smith
- From the Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, CURE: Digestive Diseases Research Center, and the Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California 90073
| | - Jan V Stevens
- From the Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine
| | - Steven H Young
- From the Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, the Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California 90073
| | - Enrique Rozengurt
- From the Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, CURE: Digestive Diseases Research Center, and the Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California 90073 Molecular Biology Institute, UCLA, Los Angeles, California 90095 and
| |
Collapse
|
22
|
Tang L, Cheng CY, Sun X, Pedicone AJ, Mohamadzadeh M, Cheng SX. The Extracellular Calcium-Sensing Receptor in the Intestine: Evidence for Regulation of Colonic Absorption, Secretion, Motility, and Immunity. Front Physiol 2016; 7:245. [PMID: 27458380 PMCID: PMC4914593 DOI: 10.3389/fphys.2016.00245] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 06/03/2016] [Indexed: 12/14/2022] Open
Abstract
Different from other epithelia, the intestinal epithelium has the complex task of providing a barrier impeding the entry of toxins, food antigens, and microbes, while at the same time allowing for the transfer of nutrients, electrolytes, water, and microbial metabolites. These molecules/organisms are transported either transcellularly, crossing the apical and basolateral membranes of enterocytes, or paracellularly, passing through the space between enterocytes. Accordingly, the intestinal epithelium can affect energy metabolism, fluid balance, as well as immune response and tolerance. To help accomplish these complex tasks, the intestinal epithelium has evolved many sensing receptor mechanisms. Yet, their roles and functions are only now beginning to be elucidated. This article explores one such sensing receptor mechanism, carried out by the extracellular calcium-sensing receptor (CaSR). In addition to its established function as a nutrient sensor, coordinating food digestion, nutrient absorption, and regulating energy metabolism, we present evidence for the emerging role of CaSR in the control of intestinal fluid homeostasis and immune balance. An additional role in the modulation of the enteric nerve activity and motility is also discussed. Clearly, CaSR has profound effects on many aspects of intestinal function. Nevertheless, more work is needed to fully understand all functions of CaSR in the intestine, including detailed mechanisms of action and specific pathways involved. Considering the essential roles CaSR plays in gastrointestinal physiology and immunology, research may lead to a translational opportunity for the development of novel therapies that are based on CaSR's unique property of using simple nutrients such as calcium, polyamines, and certain amino acids/oligopeptides as activators. It is possible that, through targeting of intestinal CaSR with a combination of specific nutrients, oral solutions that are both inexpensive and practical may be developed to help in conditioning the gut microenvironment and in maintaining digestive health.
Collapse
Affiliation(s)
- Lieqi Tang
- Department of Pediatrics, Gastroenterology, Hepatology, and Nutrition, University of Florida Gainesville, FL, USA
| | - Catherine Y Cheng
- Department of Pediatrics, Gastroenterology, Hepatology, and Nutrition, University of Florida Gainesville, FL, USA
| | - Xiangrong Sun
- Department of Pediatrics, Gastroenterology, Hepatology, and Nutrition, University of Florida Gainesville, FL, USA
| | - Alexandra J Pedicone
- Department of Pediatrics, Gastroenterology, Hepatology, and Nutrition, University of Florida Gainesville, FL, USA
| | - Mansour Mohamadzadeh
- Department of Medicine, Center for Inflammation and Mucosal Immunology, University of Florida Gainesville, FL, USA
| | - Sam X Cheng
- Department of Pediatrics, Gastroenterology, Hepatology, and Nutrition, University of Florida Gainesville, FL, USA
| |
Collapse
|
23
|
The calcium-sensing receptor and the hallmarks of cancer. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1398-407. [DOI: 10.1016/j.bbamcr.2015.11.017] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/17/2015] [Accepted: 11/18/2015] [Indexed: 02/07/2023]
|
24
|
Tang B, Chow JYC, Dong TX, Yang SM, Lu DS, Carethers JM, Dong H. Calcium sensing receptor suppresses human pancreatic tumorigenesis through a novel NCX1/Ca(2+)/β-catenin signaling pathway. Cancer Lett 2016; 377:44-54. [PMID: 27108064 DOI: 10.1016/j.canlet.2016.04.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 03/09/2016] [Accepted: 04/17/2016] [Indexed: 02/07/2023]
Abstract
The calcium sensing receptor (CaSR) is functionally expressed in normal human pancreases, but its pathological role in pancreatic tumorigenesis is currently unknown. We sought to investigate the role of CaSR in pancreatic cancer (PC) and the underlying molecular mechanisms. We revealed that the expression of CaSR was consistently downregulated in the primary cancer tissues from PC patients, which was correlated with tumor size, differentiation and poor survival of the patients. CaSR activation markedly suppressed pancreatic tumorigenesis in vitro and in vivo likely through the Ca(2+) entry mode of Na(+)/Ca(2+) exchanger 1 (NCX1) to induce Ca(2+) entry into PC cells. Moreover, NCX1-mediated Ca(2+) entry resulted in Ca(2+)-dependent inhibition of β-catenin signaling in PC cells, eventually leading to the inhibition of pancreatic tumorigenesis. Collectively, we demonstrate for the first time that CaSR exerts a suppressive function in pancreatic tumorigenesis through a novel NCX1/Ca(2+)/β-catenin signaling pathway. Targeting this specific signaling pathway could be a potential therapeutic strategy for PC.
Collapse
Affiliation(s)
- Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Jimmy Y C Chow
- Department of Medicine, University of California, San Diego, CA, USA
| | - Tobias Xiao Dong
- Department of Medicine, University of California, San Diego, CA, USA
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - De-Sheng Lu
- Cancer Research Center, Shenzhen University, Shenzhen, China
| | - John M Carethers
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
| | - Hui Dong
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University, Chongqing, China; Department of Medicine, University of California, San Diego, CA, USA.
| |
Collapse
|
25
|
Cheng SX. Calcium-sensing receptor: A new target for therapy of diarrhea. World J Gastroenterol 2016; 22:2711-2724. [PMID: 26973410 PMCID: PMC4777994 DOI: 10.3748/wjg.v22.i9.2711] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/18/2015] [Accepted: 12/08/2015] [Indexed: 02/06/2023] Open
Abstract
Management of acute diarrhea remains a global challenge, particularly in resource-limiting countries. Oral rehydration solution (ORS), a passive rehydrating therapy developed approximately 40 years ago, remains the mainstay treatment. Although ORS is effective for hydration, since it does not inhibit enterotoxin-mediated excessive secretion, reduced absorption and compromised barrier function - the primary mechanisms of diarrhea, ORS does not offer a rapid relief of diarrhea symptom. There are a few alternative therapies available, yet the use of these drugs is limited by their expense, lack of availability and/or safety concerns. Novel anti-diarrheal therapeutic approaches, particularly those simple affordable therapies, are needed. This article explores intestinal calcium-sensing receptor (CaSR), a newly uncovered target for therapy of diarrhea. Unlike others, targeting this host antidiarrheal receptor system appears “all-inclusive”: it is anti-secretory, pro-absorptive, anti-motility, and anti-inflammatory. Thus, activating CaSR reverses changes of both secretory and inflammatory diarrheas. Considering its unique property of using simple nutrients such as calcium, polyamines, and certain amino acids/oligopeptides as activators, it is possible that through targeting of CaSR with a combination of specific nutrients, novel oral rehydrating solutions that are inexpensive and practical to use in all countries may be developed.
Collapse
|
26
|
Wang J, Han L, Sinnett-Smith J, Han LL, Stevens JV, Rozengurt N, Young SH, Rozengurt E. Positive cross talk between protein kinase D and β-catenin in intestinal epithelial cells: impact on β-catenin nuclear localization and phosphorylation at Ser552. Am J Physiol Cell Physiol 2016; 310:C542-57. [PMID: 26739494 DOI: 10.1152/ajpcell.00302.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/05/2016] [Indexed: 12/17/2022]
Abstract
Given the fundamental role of β-catenin signaling in intestinal epithelial cell proliferation and the growth-promoting function of protein kinase D1 (PKD1) in these cells, we hypothesized that PKDs mediate cross talk with β-catenin signaling. The results presented here provide several lines of evidence supporting this hypothesis. We found that stimulation of intestinal epithelial IEC-18 cells with the G protein-coupled receptor (GPCR) agonist angiotensin II (ANG II), a potent inducer of PKD activation, promoted endogenous β-catenin nuclear localization in a time-dependent manner. A significant increase was evident within 1 h of ANG II stimulation (P< 0.01), peaked at 4 h (P< 0.001), and declined afterwards. GPCR stimulation also induced a marked increase in β-catenin-regulated genes and phosphorylation at Ser(552) in intestinal epithelial cells. Exposure to preferential inhibitors of the PKD family (CRT006610 or kb NB 142-70) or knockdown of the isoforms of the PKD family prevented the increase in β-catenin nuclear localization and phosphorylation at Ser(552) in response to ANG II. GPCR stimulation also induced the formation of a complex between PKD1 and β-catenin, as shown by coimmunoprecipitation that depended on PKD1 catalytic activation, as it was abrogated by cell treatment with PKD family inhibitors. Using transgenic mice that express elevated PKD1 protein in the intestinal epithelium, we detected a marked increase in the localization of β-catenin in the nucleus of crypt epithelial cells in the ileum of PKD1 transgenic mice, compared with nontransgenic littermates. Collectively, our results identify a novel cross talk between PKD and β-catenin in intestinal epithelial cells, both in vitro and in vivo.
Collapse
Affiliation(s)
- Jia Wang
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California
| | - Liang Han
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California
| | - James Sinnett-Smith
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California; CURE, Digestive Diseases Research Center, Los Angeles, California; Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California
| | - Li-Li Han
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California
| | - Jan V Stevens
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California
| | - Nora Rozengurt
- CURE, Digestive Diseases Research Center, Los Angeles, California;
| | - Steven H Young
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California; Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California
| | - Enrique Rozengurt
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, Los Angeles, California; CURE, Digestive Diseases Research Center, Los Angeles, California; Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California; and Veterans Affairs Greater Los Angeles Health Care System, Los Angeles, California
| |
Collapse
|
27
|
Aggarwal A, Höbaus J, Tennakoon S, Prinz-Wohlgenannt M, Graça J, Price SA, Heffeter P, Berger W, Baumgartner-Parzer S, Kállay E. Active vitamin D potentiates the anti-neoplastic effects of calcium in the colon: A cross talk through the calcium-sensing receptor. J Steroid Biochem Mol Biol 2016; 155:231-8. [PMID: 25758239 DOI: 10.1016/j.jsbmb.2015.02.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 02/09/2015] [Accepted: 02/11/2015] [Indexed: 02/06/2023]
Abstract
Epidemiological studies suggest an inverse correlation between dietary calcium (Ca(2+)) and vitamin D intake and the risk of colorectal cancer (CRC). It has been shown in vitro that the active vitamin D metabolite, 1,25-dihydroxyvitamin D3 (1,25-D3) can upregulate expression of the calcium-sensing receptor (CaSR). In the colon, CaSR has been suggested to regulate proliferation of colonocytes. However, during tumorigenesis colonic CaSR expression is downregulated and we hypothesized that the loss of CaSR could influence the anti-tumorigenic effects of Ca(2+) and vitamin D. Our aim was to assess the impact of CaSR expression and function on the anti-neoplastic effects of 1,25-D3 in colon cancer cell lines. We demonstrated that in the healthy colon of mice, high vitamin D diet (2500 IU/kg diet) increased expression of differentiation and apoptosis markers, decreased expression of proliferation markers and significantly upregulated CaSR mRNA expression, compared with low vitamin D diet (100 IU/kg diet). To determine the role of CaSR in this process, we transfected Caco2-15 and HT29 CRC cells with wild type CaSR (CaSR-WT) or a dominant negative CaSR mutant (CaSR-DN) and treated them with 1,25-D3 alone, or in combination with CaSR activators (Ca(2+) and NPS R-568). 1,25-D3 enhanced the anti-proliferative effects of Ca(2+) and induced differentiation and apoptosis only in cells with a functional CaSR, which were further enhanced in the presence of NPS R-568, a positive allosteric modulator of CaSR. The mutant CaSR inhibited the anti-tumorigenic effects of 1,25-D3 suggesting that the anti-neoplastic effects of 1,25-D3 are, at least in part, mediated by the CaSR. Taken together, our data provides molecular evidence to support the epidemiological observation that both, vitamin D and calcium are needed for protection against malignant transformation of the colon and that their effect is modulated by the presence of a functional CaSR. This article is part of a Special Issue entitled '17th Vitamin D Workshop'.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/pathology
- Adenocarcinoma/prevention & control
- Aniline Compounds/pharmacology
- Animals
- Caco-2 Cells
- Calcium/metabolism
- Calcium/pharmacology
- Cell Differentiation/drug effects
- Cell Proliferation/drug effects
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Colon/drug effects
- Colon/metabolism
- Colon/pathology
- Colonic Neoplasms/genetics
- Colonic Neoplasms/metabolism
- Colonic Neoplasms/pathology
- Colonic Neoplasms/prevention & control
- Dietary Supplements
- Gene Expression Regulation, Neoplastic
- HT29 Cells
- Humans
- Male
- Mice
- Mice, Transgenic
- Mutation
- Phenethylamines
- Propylamines
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Calcium-Sensing
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Signal Transduction
- Transfection
- Vitamin D/analogs & derivatives
- Vitamin D/pharmacology
Collapse
Affiliation(s)
- Abhishek Aggarwal
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Julia Höbaus
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Samawansha Tennakoon
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | | | - João Graça
- Safety Assessment, AstraZeneca, Macclesfield, UK
| | | | - Petra Heffeter
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Research Platform 'Translational Cancer Therapy Research', Vienna, Austria
| | - Walter Berger
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria; Research Platform 'Translational Cancer Therapy Research', Vienna, Austria
| | | | - Enikö Kállay
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
28
|
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: 30] [Impact Index Per Article: 3.3] [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
|
29
|
Young SH, Rey O, Rozengurt E. Intracellular Ca(2+) oscillations generated via the extracellular Ca(2+)-sensing receptor (CaSR) in response to extracellular Ca(2+) or L-phenylalanine: Impact of the highly conservative mutation Ser170Thr. Biochem Biophys Res Commun 2015; 467:1-6. [PMID: 26431875 DOI: 10.1016/j.bbrc.2015.09.144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 09/26/2015] [Indexed: 12/21/2022]
Abstract
The extracellular Ca(2+)-sensing receptor (CaSR) is an allosteric protein that responds to changes in the extracellular concentration of Ca(2+) ([Ca(2+)]e) and aromatic amino acids with the production of different patterns of oscillations in intracellular Ca(2+) concentration ([Ca(2+)]i). An increase in [Ca(2+)]e stimulates sinusoidal oscillations in [Ca(2+)]i whereas aromatic amino acid-induced CaR activation in the presence of a threshold [Ca(2+)]e promotes transient oscillations in [Ca(2+)]i. Here, we examined spontaneous and ligand-evoked [Ca(2+)]i oscillations in single HEK-293 cells transfected with the wild type CaSR or with a mutant CaSR in which Ser170 was converted to Thr (CaSRS170T). Our analysis demonstrates that cells expressing CaSRS170T display [Ca(2+)]i oscillations in the presence of low concentrations of extracellular Ca(2+) and respond to L-Phe with robust transient [Ca(2+)]i oscillations. Our results indicate that the S170T mutation induces a marked increase in CaSR sensitivity to [Ca(2+)]e and imply that the allosteric regulation of the CaSR by aromatic amino acids is not only mediated by an heterotropic positive effect on Ca(2+) binding cooperativity but, as biased agonists, aromatic amino acids stabilize a CaSR conformation that couples to a different signaling pathway leading to transient [Ca(2+)]i oscillations.
Collapse
Affiliation(s)
- Steven H Young
- Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, David Geffen School of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Osvaldo Rey
- Institute of Immunology, Genetics and Metabolism, Consejo Nacional de Investigaciones Cientificas y Tecnicas, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Enrique Rozengurt
- Division of Digestive Diseases, Department of Medicine, CURE: Digestive Diseases Research Center, David Geffen School of Medicine and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, CA 90095, USA; Veterans Affairs West Los Angeles Healthcare System, Los Angeles, CA, USA.
| |
Collapse
|
30
|
Aggarwal A, Prinz-Wohlgenannt M, Tennakoon S, Höbaus J, Boudot C, Mentaverri R, Brown EM, Baumgartner-Parzer S, Kállay E. The calcium-sensing receptor: A promising target for prevention of colorectal cancer. BIOCHIMICA ET BIOPHYSICA ACTA 2015; 1853:2158-67. [PMID: 25701758 PMCID: PMC4549785 DOI: 10.1016/j.bbamcr.2015.02.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Revised: 02/07/2015] [Accepted: 02/09/2015] [Indexed: 02/07/2023]
Abstract
The inverse correlation between dietary calcium intake and the risk of colorectal cancer (CRC) is well known, but poorly understood. Expression of the calcium-sensing receptor (CaSR), a calcium-binding G protein-coupled receptor is downregulated in CRC leading us to hypothesize that the CaSR has tumor suppressive roles in the colon. The aim of this study was to understand whether restoration of CaSR expression could reduce the malignant phenotype in CRC. In human colorectal tumors, expression of the CaSR negatively correlated with proliferation markers whereas loss of CaSR correlated with poor tumor differentiation and reduced apoptotic potential. In vivo, dearth of CaSR significantly increased expression of proliferation markers and decreased levels of differentiation and apoptotic markers in the colons of CaSR/PTH double knock-out mice confirming the tumor suppressive functions of CaSR. In vitro CRC cells stably overexpressing wild-type CaSR showed significant reduction in proliferation, as well as increased differentiation and apoptotic potential. The positive allosteric modulator of CaSR, NPS R-568 further enhanced these effects, whereas treatment with the negative allosteric modulator, NPS 2143 inhibited these functions. Interestingly, the dominant-negative mutant (R185Q) was able to abrogate these effects. Our results demonstrate a critical tumor suppressive role of CaSR in the colon. Restoration of CaSR expression and function is linked to regulation of the balance between proliferation, differentiation, and apoptosis and provides a rationale for novel strategies in CRC therapy.
Collapse
MESH Headings
- Amino Acid Substitution
- Aniline Compounds/pharmacology
- Animals
- Apoptosis/drug effects
- Apoptosis/genetics
- Caco-2 Cells
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Cell Proliferation/drug effects
- Cell Proliferation/genetics
- Colorectal Neoplasms/genetics
- Colorectal Neoplasms/metabolism
- Colorectal Neoplasms/pathology
- Colorectal Neoplasms/prevention & control
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Gene Expression Regulation, Neoplastic/genetics
- Humans
- Male
- Mice
- Mice, Knockout
- Mutation, Missense
- Naphthalenes/pharmacology
- Phenethylamines
- Propylamines
- Receptors, Calcium-Sensing/antagonists & inhibitors
- Receptors, Calcium-Sensing/genetics
- Receptors, Calcium-Sensing/metabolism
- Receptors, G-Protein-Coupled/antagonists & inhibitors
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Tumor Suppressor Proteins/antagonists & inhibitors
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
Collapse
Affiliation(s)
- Abhishek Aggarwal
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | | | - Samawansha Tennakoon
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Julia Höbaus
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Cedric Boudot
- INSERM U1088, University of Picardie Jules Verne, Amiens, France
| | | | - Edward M Brown
- Division of Endocrinology, Diabetes and Hypertension, Department of Medicine, Brigham and Women's Hospital, Boston, USA
| | | | - Enikö Kállay
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
31
|
Tang L, Peng M, Liu L, Chang W, Binder HJ, Cheng SX. Calcium-sensing receptor stimulates Cl(-)- and SCFA-dependent but inhibits cAMP-dependent HCO3(-) secretion in colon. Am J Physiol Gastrointest Liver Physiol 2015; 308:G874-83. [PMID: 25792563 PMCID: PMC4437021 DOI: 10.1152/ajpgi.00341.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 03/10/2015] [Indexed: 02/07/2023]
Abstract
Colonic bicarbonate (HCO3(-)) secretion is a well-established physiological process that is closely linked to overall fluid and electrolyte movement in the mammalian colon. These present studies show that extracellular calcium-sensing receptor (CaSR), a fundamental mechanism for sensing and regulating ionic and nutrient compositions of extracellular milieu in the small and large intestine, regulates HCO3(-) secretion. Basal and induced HCO3(-) secretory responses to CaSR agonists were determined by pH stat techniques used in conjunction with short-circuit current measurements in mucosa from rat distal colon mounted in Ussing chambers. R568, a specific CaSR activator, stimulated lumen Cl(-)- and short-chain fatty acid (SCFA)-dependent HCO3(-) secretion but inhibited cyclic nucleotide-activated HCO3(-) secretion. Consequently, at physiological conditions (either at basal or during lumen acid challenge) when electroneutral Cl(-)/HCO3(-) and SCFA/HCO3(-) exchangers dominate, CaSR stimulates HCO3(-) secretion; in contrast, in experimental conditions that stimulate fluid and HCO3(-) secretion, e.g., when forskolin activates electrogenic cystic fibrosis transmembrane conductance regulator-mediated HCO3(-) conductance, CaSR activation inhibits HCO3(-) secretion. Corresponding changes in JHCO3 (μeq·h(-1)·cm(-2), absence vs. presence of R568) were 0.18 ± 0.03 vs. 0.31 ± 0.08 under basal nonstimulated conditions and 1.85 ± 0.23 vs. 0.45 ± 0.06 under forskolin-stimulated conditions. Similarly, activation of CaSR by R568 stimulated Cl(-)- and SCFA-dependent HCO3(-) secretion and inhibited cAMP-dependent HCO3(-) secretion in colon mucosa of wild-type mice; such effects were abolished in CaSR-null mice. These results suggest a new paradigm for regulation of intestinal ion transport in which HCO3(-) secretion may be fine-tuned by CaSR in accordance with nutrient availability and state of digestion and absorption. The ability of CaSR agonists to inhibit secretagogue-induced intestinal HCO3(-) secretion suggests that modulation of CaSR activity may provide a new therapeutic approach to correct HCO3(-) deficit and metabolic acidosis, a primary cause of morbidity and mortality in acute infectious diarrheal illnesses.
Collapse
Affiliation(s)
- Lieqi Tang
- 1Division of Gastroenterology, Nutrition and Hepatology, Department of Pediatrics, University of Florida, Gainesville, Florida;
| | - Minzhi Peng
- 1Division of Gastroenterology, Nutrition and Hepatology, Department of Pediatrics, University of Florida, Gainesville, Florida;
| | - Li Liu
- 2Department of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong, People's Republic of China;
| | - Wenhan Chang
- 3Endocrine Research, VA Medical Center, University of California at San Francisco, San Francisco, California;
| | - Henry J. Binder
- 4Department of Internal Medicine, Yale University, New Haven, Connecticut
| | - Sam X. Cheng
- 1Division of Gastroenterology, Nutrition and Hepatology, Department of Pediatrics, University of Florida, Gainesville, Florida;
| |
Collapse
|
32
|
Aggarwal A, Prinz-Wohlgenannt M, Gröschel C, Tennakoon S, Meshcheryakova A, Chang W, Brown EM, Mechtcheriakova D, Kállay E. The calcium-sensing receptor suppresses epithelial-to-mesenchymal transition and stem cell- like phenotype in the colon. Mol Cancer 2015; 14:61. [PMID: 25879211 PMCID: PMC4405849 DOI: 10.1186/s12943-015-0330-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 03/03/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The calcium sensing receptor (CaSR), a calcium-binding G protein-coupled receptor is expressed also in tissues not directly involved in calcium homeostasis like the colon. We have previously reported that CaSR expression is down-regulated in colorectal cancer (CRC) and that loss of CaSR provides growth advantage to transformed cells. However, detailed mechanisms underlying these processes are largely unknown. METHODS AND RESULTS In a cohort of 111 CRC patients, we found significant inverse correlation between CaSR expression and markers of epithelial-to-mesenchymal transition (EMT), a process involved in tumor development in CRC. The colon of CaSR/PTH double-knockout, as well as the intestine-specific CaSR knockout mice showed significantly increased expression of markers involved in the EMT process. In vitro, stable expression of the CaSR (HT29(CaSR)) gave a more epithelial-like morphology to HT29 colon cancer cells with increased levels of E-Cadherin compared with control cells (HT29(EMP)). The HT29(CaSR) cells had reduced invasive potential, which was attributed to the inhibition of the Wnt/β-catenin pathway as measured by a decrease in nuclear translocation of β-catenin and transcriptional regulation of genes like GSK-3β and Cyclin D1. Expression of a spectrum of different mesenchymal markers was significantly down-regulated in HT29(CaSR) cells. The CaSR was able to block upregulation of mesenchymal markers even in an EMT-inducing environment. Moreover, overexpression of the CaSR led to down-regulation of stem cell-like phenotype. CONCLUSIONS The results from this study demonstrate that the CaSR inhibits epithelial-to-mesenchymal transition and the acquisition of a stem cell-like phenotype in the colon of mice lacking the CaSR as well as colorectal cancer cells, identifying the CaSR as a key molecule in preventing tumor progression. Our results support the rationale to develop new strategies either preventing CaSR loss or reversing its silencing.
Collapse
Affiliation(s)
- Abhishek Aggarwal
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria.
| | - Maximilian Prinz-Wohlgenannt
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria.
| | - Charlotte Gröschel
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria.
| | - Samawansha Tennakoon
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria.
| | - Anastasia Meshcheryakova
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria.
| | - Wenhan Chang
- Endocrine Research Unit, Department of Veteran Affairs Medical Center, University of California, San Francisco, CA, USA.
| | - Edward M Brown
- Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, MA, USA.
| | - Diana Mechtcheriakova
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria.
| | - Enikö Kállay
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria.
| |
Collapse
|
33
|
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
|
34
|
Chang KW, Huang NA, Liu IH, Wang YH, Wu P, Tseng YT, Hughes MW, Jiang TX, Tsai MH, Chen CY, Oyang YJ, Lin EC, Chuong CM, Lin SP. Emergence of differentially regulated pathways associated with the development of regional specificity in chicken skin. BMC Genomics 2015; 16:22. [PMID: 25612663 PMCID: PMC4326372 DOI: 10.1186/s12864-014-1202-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 12/22/2014] [Indexed: 01/17/2023] Open
Abstract
Background Regional specificity allows different skin regions to exhibit different characteristics, enabling complementary functions to make effective use of the integumentary surface. Chickens exhibit a high degree of regional specificity in the skin and can serve as a good model for when and how these regional differences begin to emerge. Results We used developing feather and scale regions in embryonic chickens as a model to gauge the differences in their molecular pathways. We employed cosine similarity analysis to identify the differentially regulated and co-regulated genes. We applied low cell techniques for expression validation and chromatin immunoprecipitation (ChIP)-based enhancer identification to overcome limited cell availabilities from embryonic chicken skin. We identified a specific set of genes demonstrating a high correlation as being differentially expressed during feather and scale development and maturation. Some members of the WNT, TGF-beta/BMP, and Notch family known to be involved in feathering skin differentiation were found to be differentially regulated. Interestingly, we also found genes along calcium channel pathways that are differentially regulated. From the analysis of differentially regulated pathways, we used calcium signaling pathways as an example for further verification. Some voltage-gated calcium channel subunits, particularly CACNA1D, are expressed spatio-temporally in the skin epithelium. These calcium signaling pathway members may be involved in developmental decisions, morphogenesis, or epithelial maturation. We further characterized enhancers associated with histone modifications, including H3K4me1, H3K27ac, and H3K27me3, near calcium channel-related genes and identified signature intensive hotspots that may be correlated with certain voltage-gated calcium channel genes. Conclusion We demonstrated the applicability of cosine similarity analysis for identifying novel regulatory pathways that are differentially regulated during development. Our study concerning the effects of signaling pathways and histone signatures on enhancers suggests that voltage-gated calcium signaling may be involved in early skin development. This work lays the foundation for studying the roles of these gene pathways and their genomic regulation during the establishment of skin regional specificity. Electronic supplementary material The online version of this article (doi:10.1186/s12864-014-1202-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kai-Wei Chang
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan. .,Genome and Systems Biology Degree Program, Academia Sinica, Taipei, Taiwan.
| | - Nancy A Huang
- Department of Computer Science and Information Engineering, National Taiwan University, Taipei, Taiwan.
| | - I-Hsuan Liu
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan. .,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan.
| | - Yi-Hui Wang
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan.
| | - Ping Wu
- Department of Pathology, School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Yen-Tzu Tseng
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan.
| | - Michael W Hughes
- Department of Pathology, School of Medicine, University of Southern California, Los Angeles, CA, USA. .,International Research Center for Wound Repair and Regeneration, National Cheng-Kung University, Tainan, Taiwan.
| | - Ting Xin Jiang
- Department of Pathology, School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Mong-Hsun Tsai
- Institute of Biotechnology, National Taiwan University, Taipei, Taiwan. .,Agricultural Biotechnology Research Centre, Academia Sinica, Taipei, Taiwan. .,Center for Systems Biology, National Taiwan University, Taipei, Taiwan.
| | - Chien-Yu Chen
- Department of Bio-Industrial Mechatronics Engineering, National Taiwan University, Taipei, Taiwan. .,Center for Systems Biology, National Taiwan University, Taipei, Taiwan.
| | - Yen-Jen Oyang
- Department of Computer Science and Information Engineering, National Taiwan University, Taipei, Taiwan. .,Center for Systems Biology, National Taiwan University, Taipei, Taiwan.
| | - En-Chung Lin
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan.
| | - Cheng-Ming Chuong
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan. .,Department of Pathology, School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Shau-Ping Lin
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan. .,Institute of Biotechnology, National Taiwan University, Taipei, Taiwan. .,Agricultural Biotechnology Research Centre, Academia Sinica, Taipei, Taiwan. .,Center for Systems Biology, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
35
|
Hernández-Bedolla MA, Carretero-Ortega J, Valadez-Sánchez M, Vázquez-Prado J, Reyes-Cruz G. Chemotactic and proangiogenic role of calcium sensing receptor is linked to secretion of multiple cytokines and growth factors in breast cancer MDA-MB-231 cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:166-82. [DOI: 10.1016/j.bbamcr.2014.10.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 10/11/2014] [Accepted: 10/15/2014] [Indexed: 12/18/2022]
|
36
|
Abstract
Vitamin D and calcium are well-established regulators of keratinocyte proliferation and differentiation. Therefore, it was not a great surprise that deletion of the vitamin D receptor (VDR) should predispose the skin to tumor formation, and that the combination of deleting both the VDR and calcium sensing receptor (CaSR) should be especially pro-oncogenic. In this review I have examined 4 mechanisms that appear to underlie the means by which VDR acts as a tumor suppressor in skin. First, DNA damage repair is curtailed in the absence of the VDR, allowing mutations in DNA to accumulate. Second and third involve the increased activation of the hedgehog and β-catenin pathways in the epidermis in the absence of the VDR, leading to poorly regulated proliferation with reduced differentiation. Finally, VDR deletion leads to a shift in the expression of long noncoding RNAs toward a more oncogenic profile. How these different mechanisms interact and their relative importance in the predisposition of the VDR null epidermis to tumor formation remain under active investigation.
Collapse
Affiliation(s)
- Daniel D Bikle
- VA Medical Center and University of California San Francisco, 1700 Owens Street, Room 373, San Francisco, CA 94158, USA
| |
Collapse
|
37
|
Cheng SX, Lightfoot YL, Yang T, Zadeh M, Tang L, Sahay B, Wang GP, Owen JL, Mohamadzadeh M. Epithelial CaSR deficiency alters intestinal integrity and promotes proinflammatory immune responses. FEBS Lett 2014; 588:4158-66. [PMID: 24842610 PMCID: PMC4234694 DOI: 10.1016/j.febslet.2014.05.007] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 04/30/2014] [Accepted: 05/02/2014] [Indexed: 12/22/2022]
Abstract
The intestinal epithelium is equipped with sensing receptor mechanisms that interact with luminal microorganisms and nutrients to regulate barrier function and gut immune responses, thereby maintaining intestinal homeostasis. Herein, we clarify the role of the extracellular calcium-sensing receptor (CaSR) using intestinal epithelium-specific Casr(-/-) mice. Epithelial CaSR deficiency diminished intestinal barrier function, altered microbiota composition, and skewed immune responses towards proinflammatory. Consequently, Casr(-/-) mice were significantly more prone to chemically induced intestinal inflammation resulting in colitis. Accordingly, CaSR represents a potential therapeutic target for autoinflammatory disorders, including inflammatory bowel diseases.
Collapse
Affiliation(s)
- Sam X Cheng
- Division of Gastroenterology, Department of Pediatrics, University of Florida, Gainesville, FL 32607, USA
| | - Yaíma L Lightfoot
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32608, USA; Division of Gastroenterology, Hepatology & Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Tao Yang
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32608, USA; Division of Gastroenterology, Hepatology & Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mojgan Zadeh
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32608, USA; Division of Gastroenterology, Hepatology & Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Lieqi Tang
- Division of Gastroenterology, Department of Pediatrics, University of Florida, Gainesville, FL 32607, USA
| | - Bikash Sahay
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32608, USA; Division of Gastroenterology, Hepatology & Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Gary P Wang
- Division of Infectious Diseases and Global Medicine, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Jennifer L Owen
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mansour Mohamadzadeh
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32608, USA; Division of Gastroenterology, Hepatology & Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
38
|
Fetahu IS, Höbaus J, Aggarwal A, Hummel DM, Tennakoon S, Mesteri I, Baumgartner-Parzer S, Kállay E. Calcium-sensing receptor silencing in colorectal cancer is associated with promoter hypermethylation and loss of acetylation on histone 3. Int J Cancer 2014; 135:2014-23. [PMID: 24691920 PMCID: PMC4282356 DOI: 10.1002/ijc.28856] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2013] [Accepted: 03/06/2014] [Indexed: 12/25/2022]
Abstract
The calcium-sensing receptor (CaSR) is suggested to mediate the antiproliferative effects of calcium in colon. However, in colorectal cancer (CRC) the expression of the CaSR is silenced and the underlying mechanisms leading to its loss are poorly understood. We investigated whether loss of the CaSR expression in colorectal tumors is caused by DNA hypermethylation and imbalance of transcriptionally permissive/repressive histone alterations. We observed significantly lower CaSR mRNA expression (n = 65, p < 0.001) in colorectal tumors compared with the adjacent mucosa from the same patient. Immunofluorescence staining confirmed downregulation of the CaSR protein also. The CaSR promoter was methylated to a greater extent in tumors compared with adjacent mucosa as determined by bisulfite sequencing (n = 20, p < 0.01) and by pyrosequencing (n = 45, p < 0.001), and methylation correlated inversely with mRNA expression (n = 20, ρ = -0.310, p < 0.05 and n = 45, ρ = -0.588, p < 0.001). Treatments with 5-aza-2'-deoxycytidine (DAC), a DNA methyltransferase inhibitor and/or with two different histone deacetylase inhibitors, trichostatin A (TSA) or suberoylanilide hydroxamic acid (SAHA) restored the expression of CaSR in colon cancer cells. Restored CaSR expression in Coga1A and HT29 cells was functional. Inhibition of lysine-specific demethylase 1 (LSD1) to prevent demethylation of mono- and dimethylated H3K4, increased CaSR expression only marginally. Our data show that hypermethylation of the CaSR promoter and H3K9 deacetylation, but not H3K4me2 demethylation are important factors that cause silencing of the CaSR in colorectal cancer.
Collapse
Affiliation(s)
- Irfete S Fetahu
- Department of Pathophysiology and Allergy Research, Medical University of ViennaVienna, Austria
| | - Julia Höbaus
- Department of Pathophysiology and Allergy Research, Medical University of ViennaVienna, Austria
| | - Abhishek Aggarwal
- Department of Pathophysiology and Allergy Research, Medical University of ViennaVienna, Austria
| | - Doris M Hummel
- Department of Pathophysiology and Allergy Research, Medical University of ViennaVienna, Austria
| | - Samawansha Tennakoon
- Department of Pathophysiology and Allergy Research, Medical University of ViennaVienna, Austria
| | - Ildiko Mesteri
- Department of Pathology, Medical University of ViennaVienna, Austria
| | | | - Enikő Kállay
- Department of Pathophysiology and Allergy Research, Medical University of ViennaVienna, Austria
| |
Collapse
|
39
|
Emerging roles of the extracellular calcium-sensing receptor in nutrient sensing: control of taste modulation and intestinal hormone secretion. Br J Nutr 2014; 111 Suppl 1:S16-22. [PMID: 24382107 DOI: 10.1017/s0007114513002250] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The extracellular Ca-sensing receptor (CaSR) is a sensor for a number of key nutrients within the body, including Ca ions (Ca²⁺) and L-amino acids. The CaSR is expressed in a number of specialised cells within the gastrointestinal (GI) tract, and much work has been done to examine CaSR's role as a nutrient sensor in this system. This review article examines two emerging roles for the CaSR within the GI tract--as a mediator of kokumi taste modulation in taste cells and as a regulator of dietary hormone release in response to L-amino acids in the intestine.
Collapse
|
40
|
Singh N, Aslam MN, Varani J, Chakrabarty S. Induction of calcium sensing receptor in human colon cancer cells by calcium, vitamin D and aquamin: Promotion of a more differentiated, less malignant and indolent phenotype. Mol Carcinog 2013; 54:543-53. [PMID: 26076051 DOI: 10.1002/mc.22123] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 11/26/2013] [Indexed: 12/22/2022]
Abstract
The calcium sensing receptor (CaSR) is a robust promoter of differentiation in colonic epithelial cells and functions as a tumor suppressor. Cancer cells that do not express CaSR (termed CaSR null) are highly malignant while acquisition of CaSR expression in these cells circumvents the malignant phenotype. We hypothesize that chemopreventive agents mediate their action through the induction of CaSR. Here, we compare the effectiveness of Ca(2+), vitamin D, and Aquamin (a marine algae product containing Ca(2+), magnesium and detectable levels of 72 additional minerals) on the induction of CaSR in the CBS and HCT116 human colon carcinoma cell lines and the corresponding CaSR null cells isolated from these lines. All three agonists induced CaSR mRNA and protein expression and inhibited cellular proliferation in the parental and CaSR null cells. Aquamin was found to be most potent in this regard. Induction of CaSR expression by these agonists resulted in demethylation of the CaSR gene promoter with a concurrent increase in CaSR promoter reporter activity. However, demethylation per se did not induce CaSR transcription. Induction of CaSR expression resulted in a down-regulated expression of tumor inducers and up-regulated expression of tumor suppressors. Again, Aquamin was found to be most potent in these biologic effects. This study provides a rationale for the use of a multi-mineral approach in the chemoprevention of colon cancer and suggests that induction of CaSR may be a measure of the effectiveness of chemopreventive agents.
Collapse
Affiliation(s)
- Navneet Singh
- Department of Microbiology, Immunology and Cell Biology Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IIllinois
| | - Muhammad N Aslam
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - James Varani
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan
| | - Subhas Chakrabarty
- Department of Microbiology, Immunology and Cell Biology Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, IIllinois
| |
Collapse
|
41
|
Young SH, Rey O, Sinnett-Smith J, Rozengurt E. Intracellular Ca2+ oscillations generated via the Ca2+-sensing receptor are mediated by negative feedback by PKCα at Thr888. Am J Physiol Cell Physiol 2013; 306:C298-306. [PMID: 24336654 DOI: 10.1152/ajpcell.00194.2013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
To clarify the mechanism(s) underlying intracellular Ca(2+) concentration ([Ca(2+)]i) oscillations induced by an elevation in extracellular Ca(2+) concentration ([Ca(2+)]e) via the extracellular Ca(2+)-sensing receptor (CaR), we analyzed the pattern of [Ca(2+)]i response in multiple (2,303) individual HEK-293 cells transfected with the human CaR. An increase in the [Ca(2+)]e from 1.5 to 3 mM produced oscillatory fluctuations in [Ca(2+)]i in 70% of the cell population. To determine the role of PKC in the generation of [Ca(2+)]i oscillations, cells were exposed to increasing concentrations (0.5-5 μM) of the preferential PKC inhibitor Ro-31-8220 before stimulation by extracellular Ca(2+). Ro-31-8220 at 3-5 μM completely eliminated the [Ca(2+)]e-evoked [Ca(2+)]i oscillations and transformed the pattern to a peak and sustained plateau response. Treatment with other broad PKC inhibitors, including GFI or Gö6983, produced an identical response. Similarly, treatment with Ro-31-8220 or GFI eliminated [Ca(2+)]e-evoked [Ca(2+)]i oscillations in colon-derived SW-480 cells expressing the CaR. Treatment with inhibitors targeting classic PKCs, including Gö6976 and Ro-32-0432 as well as small interfering RNA-mediated knockdown of PKCα, strikingly reduced the proportion of cell displaying [Ca(2+)]e-evoked [Ca(2+)]i oscillations. Furthermore, none of the cells analyzed expressing a CaR mutant in which the major PKC phosphorylation site Thr(888) was converted to alanine (CaRT888A) showed [Ca(2+)]i oscillations after CaR activation. Our results show that [Ca(2+)]i oscillations induced by activation of the CaR in response to an increase in extracellular Ca(2+) or exposure to the calcimimetic R-568 result from negative feedback involving PKCα-mediated phosphorylation of the CaR at Thr(888).
Collapse
Affiliation(s)
- Steven H Young
- Division of Digestive Diseases, Department of Medicine, Center for Ulcer Research and Education: Digestive Diseases Research Center David Geffen School of Medicine and Molecular Biology Institute, University of California, Los Angeles, California; and
| | | | | | | |
Collapse
|
42
|
Singh N, Chakrabarty S. Induction of CaSR expression circumvents the molecular features of malignant CaSR null colon cancer cells. Int J Cancer 2013; 133:2307-14. [PMID: 23674327 DOI: 10.1002/ijc.28270] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Accepted: 04/29/2013] [Indexed: 12/16/2022]
Abstract
We recently reported on the isolation and characterization of calcium sensing receptor (CaSR) null human colon cancer cells (Singh et al., Int J Cancer 2013; 132: 1996-2005). CaSR null cells possess a myriad of molecular features that are linked to a highly malignant and drug resistant phenotype of colon cancer. The CaSR null phenotype can be maintained in defined human embryonic stem cell culture medium. We now show that the CaSR null cells can be induced to differentiate in conventional culture medium, regained the expression of CaSR with a concurrent reversal of the cellular and molecular features associated with the null phenotype. These features include cellular morphology, expression of colon cancer stem cell markers, expression of survivin and thymidylate synthase and sensitivity to fluorouracil. Other features include the expression of epithelial mesenchymal transition linked molecules and transcription factors, oncogenic miRNAs and tumor suppressive molecule and miRNA. With the exception of cancer stem cell markers, the reversal of molecular features, upon the induction of CaSR expression, is directly linked to the expression and function of CaSR because blocking CaSR induction by shRNA circumvented such reversal. We further report that methylation and demethylation of the CaSR gene promoter underlie CaSR expression. Due to the malignant nature of the CaSR null cells, inclusion of the CaSR null phenotype in disease management may improve on the mortality of this disease. Because CaSR is a robust promoter of differentiation and mediates its action through diverse mechanisms and pathways, inactivation of CaSR may serve as a new paradigm in colon carcinogenesis.
Collapse
Affiliation(s)
- Navneet Singh
- Department of Microbiology, Immunology and Cell Biology, Simmons Cancer Institute, Southern Illinois University, School of Medicine, Springfield, IL
| | | |
Collapse
|
43
|
Singh N, Promkan M, Liu G, Varani J, Chakrabarty S. Role of calcium sensing receptor (CaSR) in tumorigenesis. Best Pract Res Clin Endocrinol Metab 2013; 27:455-63. [PMID: 23856272 DOI: 10.1016/j.beem.2013.04.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The extracellular Ca(2+)-sensing receptor (CaSR) is a robust promoter of differentiation in colonic epithelial cells and functions as a tumor suppressor in colon cancer. CaSR mediates its biologic effects through diverse mechanisms. Loss of CaSR expression activates a myriad of stem cell-like molecular features that drive and sustain the malignant and drug-resistant phenotypes of colon cancer. This CaSR-null phenotype, however, is not irreversible and induction of CaSR expression in CaSR-null cells promotes cell death mechanisms and restores drug sensitivity. The CaSR also functions as a tumor suppressor in breast cancer and promotes cellular sensitivity to cytotoxic drugs. BRCA1 and CaSR functions intersect in breast cancer cells, and CaSR activation can rescue breast cancer cells from the deleterious effect of BRCA1 mutations.
Collapse
Affiliation(s)
- Navneet Singh
- Southern Illinois University School of Medicine, Department of Medical Microbiology, Immunology and Cell Biology and Simmons Cancer Institute, Springfield, IL, USA.
| | | | | | | | | |
Collapse
|
44
|
Macleod RJ. CaSR function in the intestine: Hormone secretion, electrolyte absorption and secretion, paracrine non-canonical Wnt signaling and colonic crypt cell proliferation. Best Pract Res Clin Endocrinol Metab 2013; 27:385-402. [PMID: 23856267 DOI: 10.1016/j.beem.2013.05.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Expression and function of the CaSR have been shown in some mammalian taste buds and basal cells of the esophagus. Signaling cascades responsible for CaSR-mediated stimulation of H(+)-K(+)-ATPase on human parietal cells have been defined. Transgenic mice and reductionistic cell culture models have shown that the CaSR promotes gastrin secretion from G cells, cholecystokinin (CCK) secretion from duodenal I cells and BMP-2 secretion from sub-epithelial myofibroblasts. In addition, the CaSR mediates a novel paracrine relationship between myofibroblasts and overlying epithelial cells in the colon. Thus, CaSR activators stimulate secretion of Wnt5a from myofibroblasts and expression of the Wnt5a receptor Ror2 in epithelial cells. CaSR-mediated Wnt5a/Ror2 engagement stimulates epithelial differentiation and reduces expression of the receptor for tumor necrosis factor (TNFR1). CaSR activators also modulate intestinal motility, inhibit Cl(-) secretion and stimulate Na(+) absorption in both the small intestine and colon. Colonic epithelia from conditional and global CaSR knockout mice exhibit increased proliferation with increased Wnt/β-catenin signaling, demonstrating that the CaSR negatively modulates colonic epithelial growth.
Collapse
Affiliation(s)
- R John Macleod
- Department of Biomedical and Molecular Sciences, GIDRU, Queen's University, Kingston, Ontario K7L 3N6, Canada.
| |
Collapse
|
45
|
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: 3.3] [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
|
46
|
Sarkar P, Kumar S. Calcium sensing receptor modulation for cancer therapy. Asian Pac J Cancer Prev 2013; 13:3561-8. [PMID: 23098435 DOI: 10.7314/apjcp.2012.13.8.3561] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
The calcium sensing receptor (CaSR) is a member of the largest family of cell surface receptors, the G protein-coupled receptors involved in calcium homeostasis. The role of the CaSR in neoplasia appears to be homeostatic; loss of normal CaSR-induced response to extracellular calcium is observed in cancers of the colon and ovary, while increased release of PTHrP is observed in cancers of the breast, prostate and Leydig cells. Currently CaSR can be considered as a molecule that can either promote or prevent tumor growth depending on the type of cancer. Therefore, recognition of the multifaceted role of CaSR in gliomas and other malignant tumors in general is fundamental to elucidating the mechanisms of tumor progression and the development of novel therapeutic agents. Emphasis should be placed on development of drug-targeting methods to modulate CaSR activity in cancer cells.
Collapse
Affiliation(s)
- Puja Sarkar
- IGNOU-I2IT Centre of Excellence for Advanced Education and Research, Pune, Maharashtra, India
| | | |
Collapse
|
47
|
Li Z, Xu J, Xu P, Liu S, Yang Z. Wnt/β-catenin signalling pathway mediates high glucose induced cell injury through activation of TRPC6 in podocytes. Cell Prolif 2013; 46:76-85. [PMID: 23294354 DOI: 10.1111/cpr.12010] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2012] [Accepted: 08/26/2012] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Diabetic nephropathy is a major complication of diabetes and a frequent cause of end-stage renal disease and recent studies suggest that podocyte damage may play a role in the pathogenesis of this. At early onset of diabetic nephropathy there is podocyte drop-out, which is thought to provoke glomerular albuminuria and subsequent glomerular injury; however, the underlying molecular mechanisms of this remain poorly understood. Here we report that we tested the hypothesis that early diabetic podocyte injury is caused, at least in part, by up-regulation of transient receptor potential cation channel 6 (TRPC6), which is regulated by the canonical Wnt signalling pathway, in mouse podocytes. MATERIALS AND METHODS Mechanism of injury initiation in mouse podocytes, by high concentration of D-glucose (HG, 30 mM), was investigated by MTT, flow cytometry, real-time quantitative PCR, and western blot analysis. RESULTS HG induced apoptosis and reduced viability of differentiated podocytes. It caused time-dependent up-regulation of TRPC6 and activation of the canonical Wnt signalling pathway, in mouse podocytes. In these cells, blockade of the Wnt signalling pathway by dickkopf related protein 1 (Dkk1) resulted in effective reduction of TRPC6 up-regulation and amelioration of podocyte apoptosis. Furthermore, reduction of cell viability induced by HG was attenuated by treatment with Dkk1. CONCLUSION These findings indicate that the Wnt/β-catenin signalling pathway may potentially be active in pathogenesis of TRPC6-mediated diabetic podocyte injury.
Collapse
Affiliation(s)
- Z Li
- Department of Physiology, College of Medicine, Nankai University, 300071, Tianjin, China
| | | | | | | | | |
Collapse
|
48
|
Firth AL, Won JY, Park WS. Regulation of ca(2+) signaling in pulmonary hypertension. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2013; 17:1-8. [PMID: 23439762 PMCID: PMC3579099 DOI: 10.4196/kjpp.2013.17.1.1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 12/12/2012] [Accepted: 12/18/2012] [Indexed: 01/08/2023]
Abstract
Understanding the cellular and molecular mechanisms involved in the development and progression of pulmonary hypertension (PH) remains imperative if we are to successfully improve the quality of life and life span of patients with the disease. A whole plethora of mechanisms are associated with the development and progression of PH. Such complexity makes it difficult to isolate one particular pathway to target clinically. Changes in intracellular free calcium concentration, the most common intracellular second messenger, can have significant impact in defining the pathogenic mechanisms leading to its development and persistence. Signaling pathways leading to the elevation of [Ca(2+)](cyt) contribute to pulmonary vasoconstriction, excessive proliferation of smooth muscle cells and ultimately pulmonary vascular remodeling. This current review serves to summarize the some of the most recent advances in the regulation of calcium during pulmonary hypertension.
Collapse
Affiliation(s)
- Amy L Firth
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California, USA
| | | | | |
Collapse
|
49
|
Peterlik M, Kállay E, Cross HS. Calcium nutrition and extracellular calcium sensing: relevance for the pathogenesis of osteoporosis, cancer and cardiovascular diseases. Nutrients 2013; 5:302-27. [PMID: 23340319 PMCID: PMC3571650 DOI: 10.3390/nu5010302] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2012] [Revised: 01/10/2013] [Accepted: 01/11/2013] [Indexed: 02/07/2023] Open
Abstract
Through a systematic search in Pubmed for literature, on links between calcium malnutrition and risk of chronic diseases, we found the highest degree of evidence for osteoporosis, colorectal and breast cancer, as well as for hypertension, as the only major cardiovascular risk factor. Low calcium intake apparently has some impact also on cardiovascular events and disease outcome. Calcium malnutrition can causally be related to low activity of the extracellular calcium-sensing receptor (CaSR). This member of the family of 7-TM G-protein coupled receptors allows extracellular Ca2+ to function as a "first messenger" for various intracellular signaling cascades. Evidence demonstrates that Ca2+/CaSR signaling in functional linkage with vitamin D receptor (VDR)-activated pathways (i) promotes osteoblast differentiation and formation of mineralized bone; (ii) targets downstream effectors of the canonical and non-canonical Wnt pathway to inhibit proliferation and induce differentiation of colorectal cancer cells; (iii) evokes Ca2+ influx into breast cancer cells, thereby activating pro-apoptotic intracellular signaling. Furthermore, Ca2+/CaSR signaling opens Ca2+-sensitive K+ conductance channels in vascular endothelial cells, and also participates in IP(3)-dependent regulation of cytoplasmic Ca2+, the key intermediate of cardiomyocyte functions. Consequently, impairment of Ca2+/CaSR signaling may contribute to inadequate bone formation, tumor progression, hypertension, vascular calcification and, probably, cardiovascular disease.
Collapse
Affiliation(s)
- Meinrad Peterlik
- Department of Pathophysiology, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria.
| | | | | |
Collapse
|
50
|
Höbaus J, Thiem U, Hummel DM, Kallay E. Role of calcium, vitamin D, and the extrarenal vitamin D hydroxylases in carcinogenesis. Anticancer Agents Med Chem 2013; 13:20-35. [PMID: 23094918 PMCID: PMC3826118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2012] [Revised: 05/05/2012] [Accepted: 05/16/2012] [Indexed: 06/01/2023]
Abstract
Vitamin D deficiency and low calcium intake are considered risk factors for several cancers. Vitamin D, synthesized in the skin or ingested through the diet, is transformed through two hydroxylation steps to the active metabolite, 1α,25-dihydroxyvitamin D3 (1,25-D3). 25-hydroxylases in the liver are responsible for the first hydroxylation step. The ultimate activation is performed by the renal 25-hydroxyvitamin D 1α-hydroxylase (CYP27B1), while the 1,25-dihydroxyvitamin D 24-hydroxylase (CYP24A1) in the kidneys degrades the active metabolite. These two renal vitamin D hydroxylases control the endocrine serum 1,25-D3 levels, and are responsible for maintaining mineral homeostasis. In addition, the active vitamin D hormone 1,25-D3 regulates cellular proliferation, differentiation, and apoptosis in multiple tissues in a paracrine/autocrine manner. Interestingly, it is the low serum level of the precursor 25- hydroxyvitamin D3 (25-D3) that predisposes to numerous cancers and other chronic diseases, and not the serum concentration of the active vitamin D hormone. The extra-renal autocrine/paracrine vitamin D system is able to synthesize and degrade locally the active 1,25- D3 necessary to maintain normal cell growth and to counteract mitogenic stimuli. Thus, vitamin D hydroxylases play a prominent role in this process. The present review describes the role of the vitamin D hydroxylases in cancer pathogenesis and the cross-talk between the extra-renal autocrine/paracrine vitamin D system and calcium in cancer prevention.
Collapse
Affiliation(s)
- Julia Höbaus
- Department of Pathophysiology and Allergy Research
| | - Ursula Thiem
- Department of Pathophysiology and Allergy Research
- Division of Nephrology and Dialysis, Medical University of Vienna
| | | | - Enikö Kallay
- Department of Pathophysiology and Allergy Research
| |
Collapse
|