51
|
Tan CS, Ch'ng YS, Loh YC, Zaini Asmawi M, Ahmad M, Yam MF. Vasorelaxation effect of Glycyrrhizae uralensis through the endothelium-dependent Pathway. JOURNAL OF ETHNOPHARMACOLOGY 2017; 199:149-160. [PMID: 28161542 DOI: 10.1016/j.jep.2017.02.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 01/06/2017] [Accepted: 02/01/2017] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Glycyrrhiza uralensis (G. uralensis) is one of the herbs used in traditional Chinese medicine (TCM) and serves as an envoy medicinal. Since G. uralensis plays a major role in the anti-hypertensive TCM formulae, we believe that G. uralensis might possess vasorelaxation activity. AIM OF THE STUDY This study is designed to investigate the vasorelaxation effect of G. uralensis from various extracts and to study its pharmacology effect. MATERIALS AND METHODS The vasorelaxation effect of G. uralensis extracts were evaluated on thoracic aortic rings isolated from Sprague Dawley rats. RESULTS Among these three extracts of G. uralensis, 50% ethanolic extract (EFG) showed the strongest vasorelaxation activity. EFG caused the relaxation of the aortic rings pre-contracted with phenylephrine either in the presence or absence of endothelium and pre-contracted with potassium chloride in endothelium-intact aortic ring. Nω-nitro-L-arginine methyl ester, methylene blue, or 1H-[1,2,4]Oxadiazolo[4,3-a]quinoxalin-1-one inhibit the vasorelaxation effect of EFG in the presence of endothelium. On the other hand, in the presence of the potassium channel blockers (tetraethylammonium and barium chloride), the vasorelaxation effect of EFG was not affected, but glibenclamide and 4-aminopyridine did inhibit the vasorelaxation effect of EFG. With indomethacin, atropine and propranolol, the vasorelaxation effect by EFG was significantly reduced. EFG was also found to be effective in reducing Ca2+ release from sarcoplasmic reticulum and the blocking of calcium channels. CONCLUSIONS The results obtained suggest that EFG is involved in the NO/sGC/cGMP pathway.
Collapse
Affiliation(s)
- Chu Shan Tan
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, USM 11800 Minden, Pulau Pinang, Malaysia
| | - Yung Sing Ch'ng
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, USM 11800 Minden, Pulau Pinang, Malaysia
| | - Yean Chun Loh
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, USM 11800 Minden, Pulau Pinang, Malaysia
| | - Mohd Zaini Asmawi
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, USM 11800 Minden, Pulau Pinang, Malaysia
| | - Mariam Ahmad
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, USM 11800 Minden, Pulau Pinang, Malaysia
| | - Mun Fei Yam
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, USM 11800 Minden, Pulau Pinang, Malaysia.
| |
Collapse
|
52
|
Casciano JC, Duchemin NJ, Lamontagne RJ, Steel LF, Bouchard MJ. Hepatitis B virus modulates store-operated calcium entry to enhance viral replication in primary hepatocytes. PLoS One 2017; 12:e0168328. [PMID: 28151934 PMCID: PMC5289456 DOI: 10.1371/journal.pone.0168328] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 11/30/2016] [Indexed: 12/13/2022] Open
Abstract
Many viruses modulate calcium (Ca2+) signaling to create a cellular environment that is more permissive to viral replication, but for most viruses that regulate Ca2+ signaling, the mechanism underlying this regulation is not well understood. The hepatitis B virus (HBV) HBx protein modulates cytosolic Ca2+ levels to stimulate HBV replication in some liver cell lines. A chronic HBV infection is associated with life-threatening liver diseases, including hepatocellular carcinoma (HCC), and HBx modulation of cytosolic Ca2+ levels could have an important role in HBV pathogenesis. Whether HBx affects cytosolic Ca2+ in a normal hepatocyte, the natural site of an HBV infection, has not been addressed. Here, we report that HBx alters cytosolic Ca2+ signaling in cultured primary hepatocytes. We used single cell Ca2+ imaging of cultured primary rat hepatocytes to demonstrate that HBx elevates the cytosolic Ca2+ level in hepatocytes following an IP3-linked Ca2+ response; HBx effects were similar when expressed alone or in the context of replicating HBV. HBx elevation of the cytosolic Ca2+ level required extracellular Ca2+ influx and store-operated Ca2+ (SOC) entry and stimulated HBV replication in hepatocytes. We used both targeted RT-qPCR and transcriptome-wide RNAseq analyses to compare levels of SOC channel components and other Ca2+ signaling regulators in HBV-expressing and control hepatocytes and show that the transcript levels of these various proteins are not affected by HBV. We also show that HBx regulation of SOC-regulated Ca2+ accumulation is likely the consequence of HBV modulation of a SOC channel regulatory mechanism. In support of this, we link HBx enhancement of SOC-regulated Ca2+ accumulation to Ca2+ uptake by mitochondria and demonstrate that HBx stimulates mitochondrial Ca2+ uptake in primary hepatocytes. The results of our study may provide insights into viral mechanisms that affect Ca2+ signaling to regulate viral replication and virus-associated diseases.
Collapse
Affiliation(s)
- Jessica C. Casciano
- Program in Molecular and Cellular Biology and Genetics, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Nicholas J. Duchemin
- Program in Molecular and Cellular Biology and Genetics, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - R. Jason Lamontagne
- Program in Microbiology and Immunology, Graduate School of Biomedical Sciences and Professional Studies, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Laura F. Steel
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Michael J. Bouchard
- Department of Biochemistry and Molecular Biology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
53
|
The ins and outs of calcium signalling in lactation and involution: Implications for breast cancer treatment. Pharmacol Res 2017; 116:100-104. [DOI: 10.1016/j.phrs.2016.12.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 10/31/2016] [Accepted: 12/07/2016] [Indexed: 01/19/2023]
|
54
|
Noury JB, Böhm J, Peche GA, Guyant-Marechal L, Bedat-Millet AL, Chiche L, Carlier RY, Malfatti E, Romero NB, Stojkovic T. Tubular aggregate myopathy with features of Stormorken disease due to a new STIM1 mutation. Neuromuscul Disord 2017; 27:78-82. [DOI: 10.1016/j.nmd.2016.10.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2016] [Revised: 09/25/2016] [Accepted: 10/12/2016] [Indexed: 12/01/2022]
|
55
|
Azimi I, Flanagan JU, Stevenson RJ, Inserra M, Vetter I, Monteith GR, Denny WA. Evaluation of known and novel inhibitors of Orai1-mediated store operated Ca 2+ entry in MDA-MB-231 breast cancer cells using a Fluorescence Imaging Plate Reader assay. Bioorg Med Chem 2016; 25:440-449. [PMID: 27856238 DOI: 10.1016/j.bmc.2016.11.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/01/2016] [Accepted: 11/03/2016] [Indexed: 12/20/2022]
Abstract
The Orai1 Ca2+ permeable ion channel is an important component of store operated Ca2+ entry (SOCE) in cells. It's over-expression in basal molecular subtype breast cancers has been linked with poor prognosis, making it a potential target for drug development. We pharmacologically characterised a number of reported inhibitors of SOCE in MDA-MB-231 breast cancer cells using a convenient Fluorescence Imaging Plate Reader (FLIPR) assay, and show that the rank order of their potencies in this assay is the same as those reported in a wide range of published assays. The assay was also used in a screening project seeking novel inhibitors. Following a broad literature survey of classes of calcium channel inhibitors we used simplified ligand structures to query the ZINC on-line database, and following two iterations of refinement selected a novel Orai1-selective dichlorophenyltriazole hit compound. Analogues of this were synthesized and evaluated in the FLIPR assay to develop structure-activity relationships (SAR) for the three domains of the hit; triazole (head), dichlorophenyl (body) and substituted phenyl (tail). For this series, the results suggested the need for a lipophilic tail domain and an out-of-plane twist between the body and tail domains.
Collapse
Affiliation(s)
- Iman Azimi
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia; Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia; Translational Research Institute, Brisbane, Queensland, Australia
| | - Jack U Flanagan
- Auckland Cancer Society Research Centre, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Ralph J Stevenson
- Auckland Cancer Society Research Centre, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Marco Inserra
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland 4072, Australia; School of Pharmacy, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Gregory R Monteith
- The School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia; Mater Research Institute, The University of Queensland, Brisbane, Queensland, Australia; Translational Research Institute, Brisbane, Queensland, Australia
| | - William A Denny
- Auckland Cancer Society Research Centre, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
56
|
Wei M, Zhou Y, Sun A, Ma G, He L, Zhou L, Zhang S, Liu J, Zhang SL, Gill DL, Wang Y. Molecular mechanisms underlying inhibition of STIM1-Orai1-mediated Ca 2+ entry induced by 2-aminoethoxydiphenyl borate. Pflugers Arch 2016; 468:2061-2074. [PMID: 27726010 DOI: 10.1007/s00424-016-1880-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/24/2016] [Accepted: 09/07/2016] [Indexed: 01/25/2023]
Abstract
Store-operated Ca2+ entry (SOCE) mediated by STIM1 and Orai1 is crucial for Ca2+ signaling and homeostasis in most cell types. 2-Aminoethoxydiphenyl borate (2-APB) is a well-described SOCE inhibitor, but its mechanisms of action remain largely elusive. Here, we show that 2-APB does not affect the dimeric state of STIM1, but enhances the intramolecular coupling between the coiled-coil 1 (CC1) and STIM-Orai-activating region (SOAR) of STIM1, with subsequent reduction in the formation of STIM1 puncta in the absence of Orai1 overexpression. 2-APB also inhibits Orai1 channels, directly inhibiting Ca2+ entry through the constitutively active, STIM1-independent Orai1 mutants, Orai1-P245T and Orai1-V102A. When unbound from STIM1, the constitutively active Orai1-V102C mutant is not inhibited by 2-APB. Thus, we used Orai1-V012C as a tool to examine whether 2-APB can also inhibit the coupling between STIM1 and Orai1. We reveal that the functional coupling between STIM1 and Orai1-V102C is inhibited by 2-APB. This inhibition on coupling is indirect, arising from 2-APB's action on STIM1, and it is most likely mediated by functional channel residues in the Orai1 N-terminus. Overall, our findings on this two-site inhibition mediated by 2-APB provide new understanding on Orai1-activation by STIM1, important to future drug design.
Collapse
Affiliation(s)
- Ming Wei
- Beijing Key Laboratory of Gene Resources and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, 100875, People's Republic of China
| | - Yandong Zhou
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Aomin Sun
- Beijing Key Laboratory of Gene Resources and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, 100875, People's Republic of China
| | - Guolin Ma
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, 77030, USA
| | - Lian He
- Institute of Biosciences and Technology, Texas A&M University Health Science Center, Houston, TX, 77030, USA
| | - Lijuan Zhou
- Beijing Key Laboratory of Gene Resources and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, 100875, People's Republic of China
| | - Shuce Zhang
- Beijing Key Laboratory of Gene Resources and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, 100875, People's Republic of China
| | - Jin Liu
- Beijing Key Laboratory of Gene Resources and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, 100875, People's Republic of China
| | - Shenyuan L Zhang
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, 76504, USA
| | - Donald L Gill
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resources and Molecular Development, College of Life Sciences, Beijing Normal University, Beijing, 100875, People's Republic of China.
| |
Collapse
|
57
|
Inhibition of Extracellular Calcium Influx Results in Enhanced IL-12 Production in LPS-Treated Murine Macrophages by Downregulation of the CaMKKβ-AMPK-SIRT1 Signaling Pathway. Mediators Inflamm 2016; 2016:6152713. [PMID: 27313401 PMCID: PMC4904125 DOI: 10.1155/2016/6152713] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 04/19/2016] [Accepted: 04/27/2016] [Indexed: 12/18/2022] Open
Abstract
Activated macrophages are the primary sources of IL-12, a key cytokine bridging innate and adaptive immunity. However, macrophages produce low amounts of IL-12 upon stimulation and the underlying regulatory mechanism remains unclear. In this study, we found a new calcium-dependent mechanism that controlled IL-12 production in LPS-treated murine macrophages. First, LPS was demonstrated to induce extracellular calcium entry in murine peritoneal macrophages and inhibition of calcium influx resulted in marked enhancement in IL-12 production. Then, withdrawal of extracellular calcium was found to suppress CaMKKβ and AMPK activation triggered by LPS while chemical inhibition or genetic knockdown of these two kinases augmented LPS induced IL-12 production. AMPK activation increased the NAD+/NADH ratio and activated Sirtuin 1 (SIRT1), a NAD+-dependent deacetylating enzyme and negative regulator of inflammation. Chemical inhibitor or siRNA of SIRT1 enhanced IL-12 release while its agonist suppressed IL-12 production. Finally, it was found that SIRT1 selectively affected the transcriptional activity of NF-κB which thereby inhibited IL-12 production. Overall, our study demonstrates a new role of transmembrane calcium mobilization in immunity modulation such that inhibition of calcium influx leads to impaired activation of CaMKKβ-AMPK-SIRT1 signaling pathway which lifts restriction on NF-κB activation and results in enhanced IL-12 production.
Collapse
|
58
|
Abstract
Ca(2+)entry into the cell via store-operated Ca(2+)release-activated Ca(2+)(CRAC) channels triggers diverse signaling cascades that affect cellular processes like cell growth, gene regulation, secretion, and cell death. These store-operated Ca(2+)channels open after depletion of intracellular Ca(2+)stores, and their main features are fully reconstituted by the two molecular key players: the stromal interaction molecule (STIM) and Orai. STIM represents an endoplasmic reticulum-located Ca(2+)sensor, while Orai forms a highly Ca(2+)-selective ion channel in the plasma membrane. Functional as well as mutagenesis studies together with structural insights about STIM and Orai proteins provide a molecular picture of the interplay of these two key players in the CRAC signaling cascade. This review focuses on the main experimental advances in the understanding of the STIM1-Orai choreography, thereby establishing a portrait of key mechanistic steps in the CRAC channel signaling cascade. The focus is on the activation of the STIM proteins, the subsequent coupling of STIM1 to Orai1, and the consequent structural rearrangements that gate the Orai channels into the open state to allow Ca(2+)permeation into the cell.
Collapse
Affiliation(s)
- Isabella Derler
- Institute of Biophysics, Johannes Kepler University of Linz, Linz, Austria; and
| | - Isaac Jardin
- Department of Physiology, University of Extremadura, Cáceres, Spain
| | - Christoph Romanin
- Institute of Biophysics, Johannes Kepler University of Linz, Linz, Austria; and
| |
Collapse
|
59
|
McKinney DA, Eum JH, Dhara A, Strand MR, Brown MR. Calcium influx enhances neuropeptide activation of ecdysteroid hormone production by mosquito ovaries. INSECT BIOCHEMISTRY AND MOLECULAR BIOLOGY 2016; 70:160-169. [PMID: 26772671 PMCID: PMC4767660 DOI: 10.1016/j.ibmb.2016.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 12/23/2015] [Accepted: 01/03/2016] [Indexed: 06/05/2023]
Abstract
A critical step in mosquito reproduction is the ingestion of a blood meal from a vertebrate host. In mosquitoes like Aedes aegypti, blood feeding stimulates the release of ovary ecdysteroidogenic hormone (OEH) and insulin-like peptide 3 (ILP3). This induces the ovaries to produce ecdysteroid hormone (ECD), which then drives egg maturation. In many immature insects, prothoracicotropic hormone (PTTH) stimulates the prothoracic glands to produce ECD that directs molting and metamorphosis. The receptors for OEH, ILP3 and PTTH are different receptor tyrosine kinases with OEH and ILP3 signaling converging downstream in the insulin pathway and PTTH activating the mitogen-activated protein kinase pathway. Calcium (Ca(2+)) flux and cAMP have also been implicated in PTTH signaling, but the role of Ca(2+) in OEH, ILP3, and cAMP signaling in ovaries is unknown. Here, we assessed whether Ca(2+) flux affects OEH, ILP3, and cAMP activity in A. aegypti ovaries and also asked whether PTTH stimulated ovaries to produce ECD. Results indicated that Ca(2+) flux enhanced but was not essential for OEH or ILP3 activity, whereas cAMP signaling was dependent on Ca(2+) flux. Recombinant PTTH from Bombyx mori fully activated ECD production by B. mori PTGs, but exhibited no activity toward A. aegypti ovaries. Recombinant PTTH from A. aegypti also failed to stimulate either B. mori PTGs or A. aegypti ovaries to produce ECD. We discuss the implications of these results in the context of mosquito reproduction and ECD biosynthesis by insects generally.
Collapse
Affiliation(s)
- David A McKinney
- Department of Entomology, University of Georgia, Athens, GA 30602, USA
| | - Jai-Hoon Eum
- Department of Entomology, University of Georgia, Athens, GA 30602, USA
| | - Animesh Dhara
- Department of Entomology, University of Georgia, Athens, GA 30602, USA
| | - Michael R Strand
- Department of Entomology, University of Georgia, Athens, GA 30602, USA
| | - Mark R Brown
- Department of Entomology, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
60
|
Loaiza A, Carretta MD, Taubert A, Hermosilla C, Hidalgo MA, Burgos RA. Differential intracellular calcium influx, nitric oxide production, ICAM-1 and IL8 expression in primary bovine endothelial cells exposed to nonesterified fatty acids. BMC Vet Res 2016; 12:38. [PMID: 26916791 PMCID: PMC4766702 DOI: 10.1186/s12917-016-0654-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 02/15/2016] [Indexed: 11/16/2022] Open
Abstract
Background Nonesterified fatty acids (NEFAs) are involved in proinflammatory processes in cattle, including in the increased expression of adhesion molecules in endothelial cells. However, the mechanisms underlying these effects are still unknown. The aim of this study was to assess the effects of NEFAs on the intracellular calcium (Ca2+i) influx, nitric oxide production, and ICAM-1 and IL-8 expression in primary bovine umbilical vein endothelial cells (BUVECs). Results Myristic (MA), palmitic (PA), stearic (SA), oleic (OA) and linoleic acid (LA) rapidly increased Ca2+i. The calcium response to all tested NEFAs showed an extracellular calcium dependence and only the LA response was significantly inhibited until the intracellular calcium was chelated. The EC50 values for MA and LA were 125 μM and 37 μM, respectively, and the MA and LA effects were dependent on calcium release from the endoplasmic reticulum stores and on the L-type calcium channels. Only the calcium response to MA was significantly reduced by GW1100, a selective G-protein-coupled free fatty acid receptor (GPR40) antagonist. We also detected a functional FFAR1/GPR40 protein in BUVECs by using western blotting and the FFAR1/GPR40 agonist TAK-875. Only LA increased the cellular nitric oxide levels in a calcium-dependent manner. LA stimulation but not MA stimulation increased ICAM-1 and IL-8-expression in BUVECs. This effect was inhibited by GW1100, an antagonist of FFAR1/GPR40, but not by U-73122, a phospholipase C inhibitor. Conclusions These findings strongly suggest that each individual NEFA stimulates endothelial cells in a different way, with clearly different effects on intracellular calcium mobilization, NO production, and IL-8 and ICAM-1 expression in primary BUVECs. These findings not only extend our understanding of NEFA-mediated diseases in ruminants, but also provide new insight into the different molecular mechanisms involved during endothelial cell activation by NEFAs. Electronic supplementary material The online version of this article (doi:10.1186/s12917-016-0654-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anitsi Loaiza
- Instituto de Farmacología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - María D Carretta
- Instituto de Farmacología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Anja Taubert
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - Carlos Hermosilla
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - María A Hidalgo
- Instituto de Farmacología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile
| | - Rafael A Burgos
- Instituto de Farmacología, Facultad de Ciencias Veterinarias, Universidad Austral de Chile, Valdivia, Chile.
| |
Collapse
|
61
|
Di A, Mehta D, Malik AB. ROS-activated calcium signaling mechanisms regulating endothelial barrier function. Cell Calcium 2016; 60:163-71. [PMID: 26905827 DOI: 10.1016/j.ceca.2016.02.002] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 02/03/2016] [Accepted: 02/04/2016] [Indexed: 02/07/2023]
Abstract
Increased vascular permeability is a common pathogenic feature in many inflammatory diseases. For example in acute lung injury (ALI) and its most severe form, the acute respiratory distress syndrome (ARDS), lung microvessel endothelia lose their junctional integrity resulting in leakiness of the endothelial barrier and accumulation of protein rich edema. Increased reactive oxygen species (ROS) generated by neutrophils (PMNs) and other inflammatory cells play an important role in increasing endothelial permeability. In essence, multiple inflammatory syndromes are caused by dysfunction and compromise of the barrier properties of the endothelium as a consequence of unregulated acute inflammatory response. This review focuses on the role of ROS signaling in controlling endothelial permeability with particular focus on ALI. We summarize below recent progress in defining signaling events leading to increased endothelial permeability and ALI.
Collapse
Affiliation(s)
- Anke Di
- Department of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612, United States
| | - Dolly Mehta
- Department of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612, United States
| | - Asrar B Malik
- Department of Pharmacology, The University of Illinois College of Medicine, Chicago, IL 60612, United States.
| |
Collapse
|
62
|
Zuccolo E, Bottino C, Diofano F, Poletto V, Codazzi AC, Mannarino S, Campanelli R, Fois G, Marseglia GL, Guerra G, Montagna D, Laforenza U, Rosti V, Massa M, Moccia F. Constitutive Store-Operated Ca2+ Entry Leads to Enhanced Nitric Oxide Production and Proliferation in Infantile Hemangioma-Derived Endothelial Colony-Forming Cells. Stem Cells Dev 2016; 25:301-19. [DOI: 10.1089/scd.2015.0240] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- Estella Zuccolo
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Cinzia Bottino
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Federica Diofano
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| | - Valentina Poletto
- Research Laboratory of Biotechnology, Center for the Study of Myelofibrosis, Foundation IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Savina Mannarino
- Department of Pediatrics, Foundation IRCCS Policlinico San Matteo, Pavia, Italy
| | - Rita Campanelli
- Research Laboratory of Biotechnology, Center for the Study of Myelofibrosis, Foundation IRCCS Policlinico San Matteo, Pavia, Italy
| | - Gabriella Fois
- Research Laboratory of Biotechnology, Center for the Study of Myelofibrosis, Foundation IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Germano Guerra
- Department of Medicine and Health Sciences “Vincenzo Tiberio,” University of Molise, Campobasso, Italy
| | - Daniela Montagna
- Laboratory of Immunology Transplantation, Foundation IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Vittorio Rosti
- Research Laboratory of Biotechnology, Center for the Study of Myelofibrosis, Foundation IRCCS Policlinico San Matteo, Pavia, Italy
| | - Margherita Massa
- Laboratory of Biotechnology, Foundation IRCCS Policlinico San Matteo, Pavia, Italy
| | - Francesco Moccia
- Laboratory of General Physiology, Department of Biology and Biotechnology “Lazzaro Spallanzani,” University of Pavia, Pavia, Italy
| |
Collapse
|
63
|
Munoz F, Hu H. The Role of Store-operated Calcium Channels in Pain. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2016; 75:139-51. [PMID: 26920011 DOI: 10.1016/bs.apha.2015.12.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Store-operated calcium channels (SOCCs) are calcium-selective cation channels. Recently, there has been explosive growth in establishing the molecular mechanisms that mediate store-operated Ca(2+) entry (SOCE) and the role of this process in normal cellular function and disease states. SOCCs and its components appear to play an important role in many Ca(2+)-dependent processes in nonexcitable cells and are implicated in several possible disorders including allergies, multiple sclerosis, cancer, and inflammatory bowel disease. Recent studies have shown that SOCCs are expressed in the central nervous system (CNS) and involved in neuronal functions and pathological conditions, including chronic pain. In this chapter, we discuss SOCE and its physiological and pathological roles in the CNS. More specifically, we discuss the expression and function of SOCCs and their downstream signaling mechanisms under chronic pain conditions.
Collapse
Affiliation(s)
- Frances Munoz
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Huijuan Hu
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
64
|
Raynal NJM, Lee JT, Wang Y, Beaudry A, Madireddi P, Garriga J, Malouf GG, Dumont S, Dettman EJ, Gharibyan V, Ahmed S, Chung W, Childers WE, Abou-Gharbia M, Henry RA, Andrews AJ, Jelinek J, Cui Y, Baylin SB, Gill DL, Issa JPJ. Targeting Calcium Signaling Induces Epigenetic Reactivation of Tumor Suppressor Genes in Cancer. Cancer Res 2015; 76:1494-505. [PMID: 26719529 DOI: 10.1158/0008-5472.can-14-2391] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 12/18/2015] [Indexed: 01/09/2023]
Abstract
Targeting epigenetic pathways is a promising approach for cancer therapy. Here, we report on the unexpected finding that targeting calcium signaling can reverse epigenetic silencing of tumor suppressor genes (TSG). In a screen for drugs that reactivate silenced gene expression in colon cancer cells, we found three classical epigenetic targeted drugs (DNA methylation and histone deacetylase inhibitors) and 11 other drugs that induced methylated and silenced CpG island promoters driving a reporter gene (GFP) as well as endogenous TSGs in multiple cancer cell lines. These newly identified drugs, most prominently cardiac glycosides, did not change DNA methylation locally or histone modifications globally. Instead, all 11 drugs altered calcium signaling and triggered calcium-calmodulin kinase (CamK) activity, leading to MeCP2 nuclear exclusion. Blocking CamK activity abolished gene reactivation and cancer cell killing by these drugs, showing that triggering calcium fluxes is an essential component of their epigenetic mechanism of action. Our data identify calcium signaling as a new pathway that can be targeted to reactivate TSGs in cancer.
Collapse
Affiliation(s)
- Noël J-M Raynal
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, Pennsylvania. Département de pharmacologie, Université de Montréal and Sainte-Justine University Hospital Research Center, Montréal, Québec, Canada
| | - Justin T Lee
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Youjun Wang
- Beijing Key Laboratory of Gene Resources and Molecular Development College of Life Sciences, Beijing Normal University, Beijing, P.R. China
| | - Annie Beaudry
- Département de pharmacologie, Université de Montréal and Sainte-Justine University Hospital Research Center, Montréal, Québec, Canada
| | - Priyanka Madireddi
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Judith Garriga
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Gabriel G Malouf
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sarah Dumont
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elisha J Dettman
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vazganush Gharibyan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Saira Ahmed
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Woonbok Chung
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Wayne E Childers
- Moulder Center for Drug Discovery Research, Philadelphia, Pennsylvania
| | | | - Ryan A Henry
- Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Andrew J Andrews
- Department of Cancer Biology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Jaroslav Jelinek
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Ying Cui
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Stephen B Baylin
- The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Donald L Gill
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, The Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Jean-Pierre J Issa
- Fels Institute for Cancer Research and Molecular Biology, Temple University School of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
65
|
Markwardt ML, Seckinger KM, Rizzo MA. Regulation of Glucokinase by Intracellular Calcium Levels in Pancreatic β Cells. J Biol Chem 2015; 291:3000-9. [PMID: 26698632 DOI: 10.1074/jbc.m115.692160] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Indexed: 01/01/2023] Open
Abstract
Glucokinase (GCK) controls the rate of glucose metabolism in pancreatic β cells, and its activity is rate-limiting for insulin secretion. Posttranslational GCK activation can be stimulated through either G protein-coupled receptors or receptor tyrosine kinase signaling pathways, suggesting a common mechanism. Here we show that inhibiting Ca(2+) release from the endoplasmic reticulum (ER) decouples GCK activation from receptor stimulation. Furthermore, pharmacological release of ER Ca(2+) stimulates activation of a GCK optical biosensor and potentiates glucose metabolism, implicating rises in cytoplasmic Ca(2+) as a critical regulatory mechanism. To explore the potential for glucose-stimulated GCK activation, the GCK biosensor was optimized using circularly permuted mCerulean3 proteins. This new sensor sensitively reports activation in response to insulin, glucagon-like peptide 1, and agents that raise cAMP levels. Transient, glucose-stimulated GCK activation was observed in βTC3 and MIN6 cells. An ER-localized channelrhodopsin was used to manipulate the cytoplasmic Ca(2+) concentration in cells expressing the optimized FRET-GCK sensor. This permitted quantification of the relationship between cytoplasmic Ca(2+) concentrations and GCK activation. Half-maximal activation of the FRET-GCK sensor was estimated to occur at ∼400 nm Ca(2+). When expressed in islets, fluctuations in GCK activation were observed in response to glucose, and we estimated that posttranslational activation of GCK enhances glucose metabolism by ∼35%. These results suggest a mechanism for integrative control over GCK activation and, therefore, glucose metabolism and insulin secretion through regulation of cytoplasmic Ca(2+) levels.
Collapse
Affiliation(s)
- Michele L Markwardt
- From the University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Kendra M Seckinger
- From the University of Maryland School of Medicine, Baltimore, Maryland 21201
| | - Mark A Rizzo
- From the University of Maryland School of Medicine, Baltimore, Maryland 21201
| |
Collapse
|
66
|
Abstract
Store-operated calcium channels (SOCs) are a major pathway for calcium signaling in virtually all metozoan cells and serve a wide variety of functions ranging from gene expression, motility, and secretion to tissue and organ development and the immune response. SOCs are activated by the depletion of Ca(2+) from the endoplasmic reticulum (ER), triggered physiologically through stimulation of a diverse set of surface receptors. Over 15 years after the first characterization of SOCs through electrophysiology, the identification of the STIM proteins as ER Ca(2+) sensors and the Orai proteins as store-operated channels has enabled rapid progress in understanding the unique mechanism of store-operate calcium entry (SOCE). Depletion of Ca(2+) from the ER causes STIM to accumulate at ER-plasma membrane (PM) junctions where it traps and activates Orai channels diffusing in the closely apposed PM. Mutagenesis studies combined with recent structural insights about STIM and Orai proteins are now beginning to reveal the molecular underpinnings of these choreographic events. This review describes the major experimental advances underlying our current understanding of how ER Ca(2+) depletion is coupled to the activation of SOCs. Particular emphasis is placed on the molecular mechanisms of STIM and Orai activation, Orai channel properties, modulation of STIM and Orai function, pharmacological inhibitors of SOCE, and the functions of STIM and Orai in physiology and disease.
Collapse
Affiliation(s)
- Murali Prakriya
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California
| | - Richard S Lewis
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois; and Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
67
|
Nurbaeva MK, Eckstein M, Snead ML, Feske S, Lacruz RS. Store-operated Ca2+ Entry Modulates the Expression of Enamel Genes. J Dent Res 2015; 94:1471-7. [PMID: 26232387 DOI: 10.1177/0022034515598144] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Dental enamel formation is an intricate process tightly regulated by ameloblast cells. The correct spatiotemporal patterning of enamel matrix protein (EMP) expression is fundamental to orchestrate the formation of enamel crystals, which depend on a robust supply of Ca2+. In the extracellular milieu, Ca2+ -EMP interactions occur at different levels. Despite its recognized role in enamel development, the molecular machinery involved in Ca2+ homeostasis in ameloblasts remains poorly understood. A common mechanism for Ca2+ influx is store-operated Ca2+ entry (SOCE). We evaluated the possibility that Ca2+ influx in enamel cells might be mediated by SOCE and the Ca2+ release-activated Ca2+ (CRAC) channel, the prototypical SOCE channel. Using ameloblast-like LS8 cells, we demonstrate that these cells express Ca2+ -handling molecules and mediate Ca2+ influx through SOCE. As a rise in the cytosolic Ca2+ concentration is a versatile signal that can modulate gene expression, we assessed whether SOCE in enamel cells had any effect on the expression of EMPs. Our results demonstrate that stimulating LS8 cells or murine primary enamel organ cells with thapsigargin to activate SOCE leads to increased expression of Amelx, Ambn, Enam, Mmp20. This effect is reversed when cells are treated with a CRAC channel inhibitor. These data indicate that Ca2+ influx in LS8 cells and enamel organ cells is mediated by CRAC channels and that Ca2+ signals enhance the expression of EMPs. Ca2+ plays an important role not only in mineralizing dental enamel but also in regulating the expression of EMPs.
Collapse
Affiliation(s)
- M K Nurbaeva
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| | - M Eckstein
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| | - M L Snead
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - S Feske
- Department of Pathology, NYU School of Medicine, New York, NY, USA
| | - R S Lacruz
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, USA
| |
Collapse
|
68
|
Idevall-Hagren O, Lü A, Xie B, De Camilli P. Triggered Ca2+ influx is required for extended synaptotagmin 1-induced ER-plasma membrane tethering. EMBO J 2015. [PMID: 26202220 DOI: 10.15252/embj.201591565] [Citation(s) in RCA: 130] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
The extended synaptotagmins (E-Syts) are ER proteins that act as Ca(2+)-regulated tethers between the ER and the plasma membrane (PM) and have a putative role in lipid transport between the two membranes. Ca(2+) regulation of their tethering function, as well as the interplay of their different domains in such function, remains poorly understood. By exposing semi-intact cells to buffers of variable Ca(2+) concentrations, we found that binding of E-Syt1 to the PI(4,5)P2-rich PM critically requires its C2C and C2E domains and that the EC50 of such binding is in the low micromolar Ca(2+) range. Accordingly, E-Syt1 accumulation at ER-PM contact sites occurred only upon experimental manipulations known to achieve these levels of Ca(2+) via its influx from the extracellular medium, such as store-operated Ca(2+) entry in fibroblasts and membrane depolarization in β-cells. We also show that in spite of their very different physiological functions, membrane tethering by E-Syt1 (ER to PM) and by synaptotagmin (secretory vesicles to PM) undergo a similar regulation by plasma membrane lipids and cytosolic Ca(2+).
Collapse
Affiliation(s)
- Olof Idevall-Hagren
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
| | - Alice Lü
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
| | - Beichen Xie
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - Pietro De Camilli
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT, USA Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
69
|
Sogkas G, Stegner D, Syed SN, Vögtle T, Rau E, Gewecke B, Schmidt RE, Nieswandt B, Gessner JE. Cooperative and alternate functions for STIM1 and STIM2 in macrophage activation and in the context of inflammation. IMMUNITY INFLAMMATION AND DISEASE 2015; 3:154-70. [PMID: 26417434 PMCID: PMC4578517 DOI: 10.1002/iid3.56] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 03/05/2015] [Accepted: 03/06/2015] [Indexed: 12/12/2022]
Abstract
Calcium (Ca(2+)) signaling in immune cells, including macrophages, controls a wide range of effector functions that are critical for host defense and contribute to inflammation and autoimmune diseases. However, receptor-mediated Ca(2+) responses consist of complex mechanisms that make it difficult to identify the pathogenesis and develop therapy. Previous studies have revealed the importance of the Ca(2+) sensor STIM1 and store-operated Ca(2+)-entry (SOCE) for Fcγ-receptor activation and IgG-induced inflammation. Here, we identify the closely related STIM2 as mediator of cell migration and cytokine production downstream of GPCR and TLR4 activation in macrophages and show that mice lacking STIM2 are partially resistant to inflammatory responses in peritonitis and LPS-induced inflammation. Interestingly, STIM2 modulates the migratory behavior of macrophages independent from STIM1 and without a strict requirement for Ca(2+) influx. While STIM2 also contributes in part to FcγR activation, the C5a-induced amplification of IgG-mediated phagocytosis is mainly dependent on STIM1. Blockade of STIM-related functions limits mortality in experimental models of AIHA and LPS-sepsis in normal mice. These results suggest benefits of Ca(2+)-inhibition for suppression of exacerbated immune reactions and illustrate the significance of alternate functions of STIM proteins in macrophage activation and in the context of innate immune inflammation.
Collapse
Affiliation(s)
- Georgios Sogkas
- Clinical Department of Immunology and Rheumatology, Hannover Medical School Germany
| | - David Stegner
- Chair of Experimental Biomedicine University Hospital and Rudolf Virchow Center DFG Research Center for Experimental Biomedicine, University of Würzburg Würzburg, Germany
| | - Shahzad N Syed
- Clinical Department of Immunology and Rheumatology, Hannover Medical School Germany
| | - Timo Vögtle
- Chair of Experimental Biomedicine University Hospital and Rudolf Virchow Center DFG Research Center for Experimental Biomedicine, University of Würzburg Würzburg, Germany
| | - Eduard Rau
- Clinical Department of Immunology and Rheumatology, Hannover Medical School Germany
| | - Britta Gewecke
- Clinical Department of Immunology and Rheumatology, Hannover Medical School Germany
| | - Reinhold E Schmidt
- Clinical Department of Immunology and Rheumatology, Hannover Medical School Germany
| | - Bernhard Nieswandt
- Chair of Experimental Biomedicine University Hospital and Rudolf Virchow Center DFG Research Center for Experimental Biomedicine, University of Würzburg Würzburg, Germany
| | | |
Collapse
|
70
|
Chen S, Zhang Z, Wu Y, Shi Q, Yan H, Mei N, Tolleson WH, Guo L. Endoplasmic Reticulum Stress and Store-Operated Calcium Entry Contribute to Usnic Acid-Induced Toxicity in Hepatic Cells. Toxicol Sci 2015; 146:116-26. [PMID: 25870318 DOI: 10.1093/toxsci/kfv075] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The use of usnic acid as a weight loss agent is a safety concern due to reports of acute liver failure in humans. Previously we demonstrated that usnic acid induces apoptosis and cytotoxicity in hepatic HepG2 cells. We also demonstrated that usnic acid induces autophagy as a survival mechanism against its cytotoxicity. In this study, we investigated and characterized further molecular mechanisms underlying the toxicity of usnic acid in HepG2 cells. We found that usnic acid causes endoplasmic reticulum (ER) stress demonstrated by the increased expression of typical ER stress markers, including CHOP, ATF-4, p-eIF2α, and spliced XBP1. Usnic acid inhibited the secretion of Gaussia luciferase measured by an ER stress reporter assay. An ER stress inhibitor 4-phenylbutyrate attenuated usnic acid-induced apoptosis. Moreover, usnic acid significantly increased the cytosolic free Ca(2+) concentration. Usnic acid increased the expression of calcium release-activated calcium channel protein 1 (CRAM1 or ORAI1) and stromal interaction molecule 1, two key components of store-operated calcium entry (SOCE), which is the major Ca(2+) influx pathway in non-excitable cells, this finding was also confirmed in primary rat hepatocytes. Furthermore, knockdown of ORAI1 prevented ER stress and ATP depletion in response to usnic acid. In contrast, overexpression of ORAI1 increased ER stress and ATP depletion caused by usnic acid. Taken together, our results suggest that usnic acid disturbs calcium homeostasis, induces ER stress, and that usnic acid-induced cellular damage occurs at least partially via activation of the Ca(2+) channel of SOCE.
Collapse
Affiliation(s)
- Si Chen
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| | - Zhuhong Zhang
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079 *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| | - Yuanfeng Wu
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| | - Qiang Shi
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| | - Hua Yan
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| | - Nan Mei
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| | - William H Tolleson
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| | - Lei Guo
- *Division of Biochemical Toxicology, Division of Genetic and Molecular Toxicology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079, Tianjin Medical University General Hospital, Tianjin 300052, China and Division of Systems Biology, National Center for Toxicological Research/U.S. FDA, Jefferson, AR 72079
| |
Collapse
|
71
|
T cell exhaustion and Interleukin 2 downregulation. Cytokine 2015; 71:339-47. [DOI: 10.1016/j.cyto.2014.11.024] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 11/13/2014] [Accepted: 11/14/2014] [Indexed: 01/30/2023]
|
72
|
Sukriti S, Tauseef M, Yazbeck P, Mehta D. Mechanisms regulating endothelial permeability. Pulm Circ 2015; 4:535-51. [PMID: 25610592 DOI: 10.1086/677356] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Accepted: 03/03/2014] [Indexed: 12/26/2022] Open
Abstract
The endothelial monolayer partitioning underlying tissue from blood components in the vessel wall maintains tissue fluid balance and host defense through dynamically opening intercellular junctions. Edemagenic agonists disrupt endothelial barrier function by signaling the opening of the intercellular junctions leading to the formation of protein-rich edema in the interstitial tissue, a hallmark of tissue inflammation that, if left untreated, causes fatal diseases, such as acute respiratory distress syndrome. In this review, we discuss how intercellular junctions are maintained under normal conditions and after stimulation of endothelium with edemagenic agonists. We have focused on reviewing the new concepts dealing with the alteration of adherens junctions after inflammatory stimulus.
Collapse
Affiliation(s)
- Sukriti Sukriti
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Mohammad Tauseef
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Pascal Yazbeck
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| | - Dolly Mehta
- Department of Pharmacology and Center for Lung and Vascular Biology, College of Medicine, University of Illinois, Chicago, Illinois, USA
| |
Collapse
|
73
|
Langfelder A, Okonji E, Deca D, Wei WC, Glitsch MD. Extracellular acidosis impairs P2Y receptor-mediated Ca(2+) signalling and migration of microglia. Cell Calcium 2015; 57:247-56. [PMID: 25623949 DOI: 10.1016/j.ceca.2015.01.004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 12/31/2014] [Accepted: 01/02/2015] [Indexed: 02/07/2023]
Abstract
Microglia are the resident macrophage and immune cell of the brain and are critically involved in combating disease and assaults on the brain. Virtually all brain pathologies are accompanied by acidosis of the interstitial fluid, meaning that microglia are exposed to an acidic environment. However, little is known about how extracellular acidosis impacts on microglial function. The activity of microglia is tightly controlled by 'on' and 'off' signals, the presence or absence of which results in generation of distinct phenotypes in microglia. Activation of G protein coupled purinergic (P2Y) receptors triggers a number of distinct behaviours in microglia, including activation, migration, and phagocytosis. Using pharmacological tools and fluorescence imaging of the murine cerebellar microglia cell line C8B4, we show that extracellular acidosis interferes with P2Y receptor-mediated Ca(2+) signalling in these cells. Distinct P2Y receptors give rise to signature intracellular Ca(2+) signals, and Ca(2+) release from stores and Ca(2+) influx are differentially affected by acidotic conditions: Ca(2+) release is virtually unaffected, whereas Ca(2+) influx, mediated at least in part by store-operated Ca(2+) channels, is profoundly inhibited. Furthermore, P2Y1 and P2Y6-mediated stimulation of migration is inhibited under conditions of extracellular acidosis, whereas basal migration independent of P2Y receptor activation is not. Taken together, our results demonstrate that an acidic microenvironment impacts on P2Y receptor-mediated Ca(2+) signalling, thereby influencing microglial responses and responsiveness to extracellular signals. This may result in altered behaviour of microglia under pathological conditions compared with microglial responses in healthy tissue.
Collapse
Affiliation(s)
- Antonia Langfelder
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Emeka Okonji
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Diana Deca
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Wei-Chun Wei
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | - Maike D Glitsch
- Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK.
| |
Collapse
|
74
|
Serrano-Flores B, Garay E, Vázquez-Cuevas FG, Arellano RO. Differential role of STIM1 and STIM2 during transient inward (T in) current generation and the maturation process in the Xenopus oocyte. BMC PHYSIOLOGY 2014; 14:9. [PMID: 25399338 PMCID: PMC4236480 DOI: 10.1186/s12899-014-0009-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 10/29/2014] [Indexed: 12/26/2022]
Abstract
BACKGROUND The Xenopus oocyte is a useful cell model to study Ca2+ homeostasis and cell cycle regulation, two highly interrelated processes. Here, we used antisense oligonucleotides to investigate the role in the oocyte of stromal interaction molecule (STIM) proteins that are fundamental elements of the store-operated calcium-entry (SOCE) phenomenon, as they are both sensors for Ca2+ concentration in the intracellular reservoirs as well as activators of the membrane channels that allow Ca2+ influx. RESULTS Endogenous STIM1 and STIM2 expression was demonstrated, and their synthesis was knocked down 48-72 h after injecting oocytes with specific antisense sequences. Selective elimination of their mRNA and protein expression was confirmed by PCR and Western blot analysis, and we then evaluated the effect of their absence on two endogenous responses: the opening of SOC channels elicited by G protein-coupled receptor (GPCR)-activated Ca2+ release, and the process of maturation stimulated by progesterone. Activation of SOC channels was monitored electrically by measuring the T in response, a Ca2+-influx-dependent Cl- current, while maturation was assessed by germinal vesicle breakdown (GVBD) scoring and electrophysiology. CONCLUSIONS It was found that STIM2, but not STIM1, was essential in both responses, and T in currents and GVBD were strongly reduced or eliminated in cells devoid of STIM2; STIM1 knockdown had no effect on the maturation process, but it reduced the T in response by 15 to 70%. Thus, the endogenous SOCE response in Xenopus oocytes depended mainly on STIM2, and its expression was necessary for entry into meiosis induced by progesterone.
Collapse
|
75
|
Audoin C, Sánchez JA, Genta-Jouve G, Alfonso A, Rios L, Vale C, Thomas OP, Botana LM. Autumnalamide, a prenylated cyclic peptide from the cyanobacterium Phormidium autumnale, acts on SH-SY5Y cells at the mitochondrial level. JOURNAL OF NATURAL PRODUCTS 2014; 77:2196-2205. [PMID: 25265024 DOI: 10.1021/np500374a] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Filamentous cyanobacteria of the genus Phormidium have been rarely studied for their chemical diversity. For the first time, the cultivable Phormidium autumnale was shown to produce a prenylated cyclic peptide named autumnalamide (1). The structure of this peptide was fully determined after a deep exploration of the spectroscopic data, including NMR and HRMS. Interestingly, a prenyl moiety was located on the guanidine end of the arginine amino acid. The absolute configurations of most amino acids were assessed using enantioselective GC/MS analysis, with (13)C NMR modeling being used for the determination of d-arginine and d-proline. The effects of 1 on sodium and calcium fluxes were studied in SH-SY5Y and hNav 1.6 HEK cells. When the Ca(2+) influx was stimulated by thapsigargin, strong inhibition was observed in the presence of 1. As a consequence, this compound may act by disrupting the normal calcium uptake of this organelle, inducing the opening of the mitochondrial permeability transition pore, which results in the indirect blockade of store-operated channels.
Collapse
Affiliation(s)
- Coralie Audoin
- Institut de Chimie de Nice-PCRE, UMR 7272 CNRS, Faculty of Science, University of Nice Sophia-Antipolis , Parc Valrose, 06108 Nice, France
| | | | | | | | | | | | | | | |
Collapse
|
76
|
Xia J, Pan R, Gao X, Meucci O, Hu H. Native store-operated calcium channels are functionally expressed in mouse spinal cord dorsal horn neurons and regulate resting calcium homeostasis. J Physiol 2014; 592:3443-61. [PMID: 24860175 DOI: 10.1113/jphysiol.2014.275065] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Store-operated calcium channels (SOCs) are calcium-selective cation channels that mediate calcium entry in many different cell types. Store-operated calcium entry (SOCE) is involved in various cellular functions. Increasing evidence suggests that impairment of SOCE is responsible for numerous disorders. A previous study demonstrated that YM-58483, a potent SOC inhibitor, strongly attenuates chronic pain by systemic or intrathecal injection and completely blocks the second phase of formalin-induced spontaneous nocifensive behaviour, suggesting a potential role of SOCs in central sensitization. However, the expression of SOCs, their molecular identity and function in spinal cord dorsal horn neurons remain elusive. Here, we demonstrate that SOCs are expressed in dorsal horn neurons. Depletion of calcium stores from the endoplasmic reticulum (ER) induced large sustained calcium entry, which was blocked by SOC inhibitors, but not by voltage-gated calcium channel blockers. Depletion of ER calcium stores activated inward calcium-selective currents, which was reduced by replacing Ca(2+) with Ba(2+) and reversed by SOC inhibitors. Using the small inhibitory RNA knockdown approach, we identified both STIM1 and STIM2 as important mediators of SOCE and SOC current, and Orai1 as a key component of the Ca(2+) release-activated Ca(2+) channels in dorsal horn neurons. Knockdown of STIM1, STIM2 or Orai1 decreased resting Ca(2+) levels. We also found that activation of neurokinin 1 receptors led to SOCE and activation of SOCs produced an excitatory action in dorsal horn neurons. Our findings reveal that a novel SOC signal is present in dorsal horn neurons and may play an important role in pain transmission.
Collapse
Affiliation(s)
- Jingsheng Xia
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, USA
| | - Rong Pan
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, USA
| | - Xinghua Gao
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, USA Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, USA
| | - Huijuan Hu
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, USA
| |
Collapse
|
77
|
Karpinsky-Semper D, Volmar CH, Brothers SP, Slepak VZ. Differential effects of the Gβ5-RGS7 complex on muscarinic M3 receptor-induced Ca2+ influx and release. Mol Pharmacol 2014; 85:758-68. [PMID: 24586057 PMCID: PMC4170115 DOI: 10.1124/mol.114.091843] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 02/28/2014] [Indexed: 11/22/2022] Open
Abstract
The G protein β subunit Gβ5 uniquely forms heterodimers with R7 family regulators of G protein signaling (RGS) proteins (RGS6, RGS7, RGS9, and RGS11) instead of Gγ. Although the Gβ5-RGS7 complex attenuates Ca(2+) signaling mediated by the muscarinic M3 receptor (M3R), the route of Ca(2+) entry (i.e., release from intracellular stores and/or influx across the plasma membrane) is unknown. Here, we show that, in addition to suppressing carbachol-stimulated Ca(2+) release, Gβ5-RGS7 enhanced Ca(2+) influx. This novel effect of Gβ5-RGS7 was blocked by nifedipine and 2-aminoethoxydiphenyl borate. Experiments with pertussis toxin, an RGS domain-deficient mutant of RGS7, and UBO-QIC {L-threonine,(3R)-N-acetyl-3-hydroxy-L-leucyl-(aR)-a-hydroxybenzenepropanoyl-2,3-idehydro-N-methylalanyl-L-alanyl-N-methyl-L-alanyl-(3R)-3-[[(2S,3R)-3-hydroxy-4- methyl-1-oxo-2-[(1-oxopropyl)amino]pentyl]oxy]-L-leucyl-N,O-dimethyl-,(7→1)-lactone (9CI)}, a novel inhibitor of Gq, showed that Gβ5-RGS7 modulated a Gq-mediated pathway. These studies indicate that Gβ5-RGS7, independent of RGS7 GTPase-accelerating protein activity, couples M3R to a nifedipine-sensitive Ca(2+) channel. We also compared the action of Gβ5-RGS7 on M3R-induced Ca(2+) influx and release elicited by different muscarinic agonists. Responses to Oxo-M [oxotremorine methiodide N,N,N,-trimethyl-4-(2-oxo-1-pyrrolidinyl)-2-butyn-1-ammonium iodide] were insensitive to Gβ5-RGS7. Pilocarpine responses consisted of a large release and modest influx components, of which the former was strongly inhibited whereas the latter was insensitive to Gβ5-RGS7. McN-A-343 [(4-hydroxy-2-butynyl)-1-trimethylammonium-3-chlorocarbanilate chloride] was the only compound whose total Ca(2+) response was enhanced by Gβ5-RGS7, attributed to, in part, by the relatively small Ca(2+) release this partial agonist stimulated. Together, these results show that distinct agonists not only have differential M3R functional selectivity, but also confer specific sensitivity to the Gβ5-RGS7 complex.
Collapse
Affiliation(s)
- Darla Karpinsky-Semper
- Department of Molecular and Cellular Pharmacology (D.K.-S., V.Z.S.) and Center for Therapeutic Innovation, Department of Psychiatry and Behavioral Sciences (C.-H.V., S.P.B.), University of Miami Miller School of Medicine, Miami, Florida
| | | | | | | |
Collapse
|
78
|
Molinari G, Colombo G, Celenza C. Respiratory allergies: a general overview of remedies, delivery systems, and the need to progress. ISRN ALLERGY 2014; 2014:326980. [PMID: 25006500 PMCID: PMC3972928 DOI: 10.1155/2014/326980] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 12/24/2013] [Indexed: 01/10/2023]
Abstract
The spread of respiratory allergies is increasing in parallel with the alarm of the scientific community. Evidently, our knowledge of the onset mechanisms of these diseases and, as a consequence, of the available remedies is inadequate. This review provides a brief, general description of current therapeutic resources and the state of research with regard to both drugs and medical devices in order to highlight their limits and the urgent need for progress. Increasing the amount of basic biochemical research will improve our knowledge of such onset mechanisms and the potential efficacy of therapeutic preparations.
Collapse
Affiliation(s)
- Giuliano Molinari
- Biochemistry Consulting Service, Giuliano Molinari, 20017 Rho, Milan, Italy
| | - Giselda Colombo
- Allergy and Immunology Unit, San Raffaele Hospital, 20132 Milan, Italy
| | - Cinzia Celenza
- Quality Assurance Service, Sandoz S.P.A., 21040 Origgio, Varese, Italy
| |
Collapse
|
79
|
Abstract
TRPC3 represents one of the first identified mammalian relative of the Drosophila trp gene product. Despite extensive biochemical and biophysical characterization as well as ambitious attempts to uncover its physiological role in native cell systems, the channel protein still represents a rather enigmatic member of the TRP superfamily. TRPC3 is significantly expressed in the brain and heart and appears of (patho)physiological importance in both non-excitable and excitable cells, being potentially involved in a wide spectrum of Ca(2+) signaling mechanisms. TRPC3 cation channels display unique gating and regulatory properties that allow for recognition and integration of multiple input stimuli including lipid mediators, cellular Ca(2+) gradients, as well as redox signals. Physiological/pathophysiological functions of this highly versatile cation channel protein are as yet incompletely understood. Its ability to associate in a dynamic manner with a variety of partner proteins enables TRPC3 to serve coordination of multiple downstream signaling pathways and control of divergent cellular functions. Here, we summarize current knowledge on ion channel features as well as possible signaling functions of TRPC3 and discuss the potential biological relevance of this signaling molecule.
Collapse
Affiliation(s)
- Michaela Lichtenegger
- Institute of Pharmaceutical Sciences - Pharmacology and Toxicology, University of Graz, A-8010, Graz, Austria
| | | |
Collapse
|
80
|
Kato K, Okamura K, Hatta M, Morita H, Kajioka S, Naito S, Yamazaki J. Involvement of IP3-receptor activation in endothelin-1-induced Ca2+ influx in rat pulmonary small artery. Eur J Pharmacol 2013; 720:255-63. [DOI: 10.1016/j.ejphar.2013.09.076] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2013] [Revised: 07/25/2013] [Accepted: 09/13/2013] [Indexed: 11/29/2022]
|
81
|
Bolaños P, Guillen A, Gámez A, Caputo C. Quantifying SOCE fluorescence measurements in mammalian muscle fibres. The effects of ryanodine and osmotic shocks. J Muscle Res Cell Motil 2013; 34:379-93. [PMID: 24129906 DOI: 10.1007/s10974-013-9360-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Accepted: 09/18/2013] [Indexed: 11/28/2022]
Abstract
We have quantified Ca(2+) entry through store operated calcium channels in mice muscle fibres, measuring the rates of change of myoplasmic [Ca(2+)], d[Ca(2+)](myo)/dt, and of Ca(2+) removal, d[Ca(2+)](Removal)/dt, turning store operated calcium entry (SOCE) ON, and OFF, by switching on or off external Ca(2+). In depleted fibres, poisoned with 10 μM cyclopiazonic acid SOCE influx was about 3 μM/s. Ryanodine (50 μM) caused a robust, nifedipine (50 μM) independent, increase in SOCE activation to 8.6 μM/s. Decreasing medium osmolarity from 300 to 220 mOsm/L, decreased SOCE to 0.9 μM/s, while increasing osmolarity from 220 to 400 mOsm/L potentiated SOCE to 43.6 μM/s. Ryanodine inhibited the effects of hypotonicity. Experiments using 2-aminoethoxydiphenyl borate, nifedipine, or Mn(2+) quenching, strongly suggest that the increased [Ca(2+)](myo) by ryanodine or hypertonic shock is mediated by potentiated SOCE activation. The Ca(2+) response decay, quantified by d[Ca(2+)](Removal)/dt, indicates a robust residual Ca(2+) removal mechanism in sarco-endoplasmic reticulum calcium ATPase poisoned fibres. SOCE high sensitivity to osmotic shocks, or to ryanodine receptor (RyR) binding, suggests its high dependency on the structural relationship between its molecular constituents, Orai1 and stromal interaction molecule and the sarcoplasmic reticulum and plasma membranes, in the triadic junctional region, where RyRs, are conspicuously present. This study demonstrates that SOCE machinery is highly sensitive to structural changes caused by binding of an agonist to its receptor or by imposed osmotical volume changes.
Collapse
Affiliation(s)
- Pura Bolaños
- Laboratorio de Fisiología Celular, Centro de Biofísica y Bioquímica, Instituto Venezolano de Investigaciones Científicas (IVIC), Caracas, Venezuela,
| | | | | | | |
Collapse
|
82
|
Russino D, McDonald E, Hejazi L, Hanson GR, Jones CE. The tachykinin peptide neurokinin B binds copper forming an unusual [CuII(NKB)2] complex and inhibits copper uptake into 1321N1 astrocytoma cells. ACS Chem Neurosci 2013; 4:1371-81. [PMID: 23875773 DOI: 10.1021/cn4000988] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Neurokinin B (NKB) is a member of the tachykinin family of neuropeptides that have neuroinflammatory, neuroimmunological, and neuroprotective functions. In a neuroprotective role, tachykinins can help protect cells against the neurotoxic processes observed in Alzheimer's disease. A change in copper homeostasis is a clear feature of Alzheimer's disease, and the dysregulation may be a contributory factor in toxicity. Copper has recently been shown to interact with neurokinin A and neuropeptide γ and can lead to generation of reactive oxygen species and peptide degradation, which suggests that copper may have a place in tachykinin function and potentially misfunction. To explore this, we have utilized a range of spectroscopic techniques to show that NKB, but not substance P, can bind Cu(II) in an unusual [Cu(II)(NKB)2] neutral complex that utilizes two N-terminal amine and two imidazole nitrogen ligands (from each molecule of NKB) and the binding substantially alters the structure of the peptide. Using 1321N1 astrocytoma cells, we show that copper can enter the cells and subsequently open plasma membrane calcium channels but when bound to neurokinin B copper ion uptake is inhibited. This data suggests a novel role for neurokinin B in protecting cells against copper-induced calcium changes and implicates the peptide in synaptic copper homeostasis.
Collapse
Affiliation(s)
- Debora Russino
- The School of Science
and Health, The University of Western Sydney, Locked bag 1797, Penrith, New South Wales 2759, Australia
| | - Elle McDonald
- The School of Science
and Health, The University of Western Sydney, Locked bag 1797, Penrith, New South Wales 2759, Australia
| | - Leila Hejazi
- Mass Spectroscopy Laboratory, The University of Western Sydney, Locked bag 1797,
Penrith, New South Wales 2759, Australia
| | - Graeme R. Hanson
- Centre for Advanced Imaging, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Christopher E. Jones
- The School of Science
and Health, The University of Western Sydney, Locked bag 1797, Penrith, New South Wales 2759, Australia
| |
Collapse
|
83
|
Jairaman A, Prakriya M. Molecular pharmacology of store-operated CRAC channels. Channels (Austin) 2013; 7:402-14. [PMID: 23807116 DOI: 10.4161/chan.25292] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Calcium influx through store-operated Ca(2+) release-activated Ca(2+) channels (CRAC channels) is a well-defined mechanism of generating cellular Ca(2+) elevations that regulates many functions including gene expression, exocytosis and cell proliferation. The identifications of the ER Ca(2+) sensing proteins, STIM1-2 and the CRAC channel proteins, Orai1-3, have led to improved understanding of the physiological roles and the activation mechanism of CRAC channels. Defects in CRAC channel function are associated with serious human diseases such as immunodeficiency and auto-immunity. In this review, we discuss several pharmacological modulators of CRAC channels, focusing specifically on the molecular mechanism of drug action and their utility in illuminating the mechanism of CRAC channel operation and their physiological roles in different cells.
Collapse
Affiliation(s)
- Amit Jairaman
- Department of Molecular Pharmacology and Biological Chemistry; Northwestern University, Feinberg School of Medicine; Chicago, IL USA
| | - Murali Prakriya
- Department of Molecular Pharmacology and Biological Chemistry; Northwestern University, Feinberg School of Medicine; Chicago, IL USA
| |
Collapse
|
84
|
Tang TH, Chang CT, Wang HJ, Erickson JD, Reichard RA, Martin AG, Shannon EK, Martin AL, Huang YW, Aronstam RS. Oxidative stress disruption of receptor-mediated calcium signaling mechanisms. J Biomed Sci 2013; 20:48. [PMID: 23844974 PMCID: PMC3716919 DOI: 10.1186/1423-0127-20-48] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 07/10/2013] [Indexed: 01/08/2023] Open
Abstract
Background Oxidative stress increases the cytosolic content of calcium in the cytoplasm through a combination of effects on calcium pumps, exchangers, channels and binding proteins. In this study, oxidative stress was produced by exposure to tert-butyl hydroperoxide (tBHP); cell viability was assessed using a dye reduction assay; receptor binding was characterized using [3H]N-methylscopolamine ([3H]MS); and cytosolic and luminal endoplasmic reticulum (ER) calcium concentrations ([Ca2+]i and [Ca2+]L, respectively) were measured by fluorescent imaging. Results Activation of M3 muscarinic receptors induced a biphasic increase in [Ca2+]i: an initial, inositol trisphosphate (IP3)-mediated release of Ca2+ from endoplasmic reticulum (ER) stores followed by a sustained phase of Ca2+ entry (i.e., store-operated calcium entry; SOCE). Under non-cytotoxic conditions, tBHP increased resting [Ca2+]i; a 90 minute exposure to tBHP (0.5-10 mM ) increased [Ca2+]i from 26 to up to 127 nM and decreased [Ca2+]L by 55%. The initial response to 10 μM carbamylcholine was depressed by tBHP in the absence, but not the presence, of extracellular calcium. SOCE, however, was depressed in both the presence and absence of extracellular calcium. Acute exposure to tBHP did not block calcium influx through open SOCE channels. Activation of SOCE following thapsigargin-induced depletion of ER calcium was depressed by tBHP exposure. In calcium-free media, tBHP depressed both SOCE and the extent of thapsigargin-induced release of Ca2+ from the ER. M3 receptor binding parameters (ligand affinity, guanine nucleotide sensitivity, allosteric modulation) were not affected by exposure to tBHP. Conclusions Oxidative stress induced by tBHP affected several aspects of M3 receptor signaling pathway in CHO cells, including resting [Ca2+]i, [Ca2+]L, IP3 receptor mediated release of calcium from the ER, and calcium entry through the SOCE. tBHP had little effect on M3 receptor binding or G protein coupling. Thus, oxidative stress affects multiple aspects of calcium homeostasis and calcium dependent signaling.
Collapse
|
85
|
Gao R, Gao X, Xia J, Tian Y, Barrett JE, Dai Y, Hu H. Potent analgesic effects of a store-operated calcium channel inhibitor. Pain 2013; 154:2034-2044. [PMID: 23778292 DOI: 10.1016/j.pain.2013.06.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Revised: 05/17/2013] [Accepted: 06/11/2013] [Indexed: 11/30/2022]
Abstract
Chronic pain often accompanies immune responses and immune cells are known to be involved in chronic pain. Store-operated calcium (SOC) channels are calcium-selective cation channels and play an important role in the immune system. YM-58483, a potent SOC channel inhibitor, has been shown to inhibit cytokine production from immune cells and attenuate antigen-induced hypersensitivity reactions. Here, we report that YM-58483 has analgesic actions in chronic pain and produces antinociceptive effects in acute pain and prevents the development of chronic pain in mice. Oral administration of 10mg/kg or 30 mg/kg YM-58483 dramatically attenuated complete Freund adjuvant (CFA)-induced thermal hyperalgesia and prevented the development of thermal and mechanical hypersensitivity in a dose-dependent manner. Analgesic effects were observed when YM-58483 was administered systemically, intrathecally and intraplantarly. YM-58483 decreased spared nerve injury (SNI)-induced thermal and mechanical hypersensitivity and prevented the development of SNI-induced pain hypersensitivity. Pretreatment with YM-58483 strongly reduced both the first and second phases of formalin-induced spontaneous nocifensive behavior in a dose-dependent manner. YM-58483 produced antinociception in acute pain induced by heat or chemical or mechanical stimuli at a dose of 30 mg/kg. YM-58483 diminished CFA-induced paw edema, and reduced production of TNF-α, IL-1β and PGE2 in the CFA-injected paw. In vitro, SOC entry in nociceptors was more robust than in nonnociceptors, and the inhibition of SOC entry by YM-58483 in nociceptors was much greater than in nonnociceptors. Our findings indicate that YM-58483 is a potent analgesic and suggest that SOC channel inhibitors may represent a novel class of therapeutics for pain.
Collapse
Affiliation(s)
- Ruby Gao
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, USA Department of Pharmacology of Chinese Materia Medica, China Pharmaceutical University, Nanjing, China
| | | | | | | | | | | | | |
Collapse
|
86
|
Viitanen TM, Sukumaran P, Löf C, Törnquist K. Functional coupling of TRPC2 cation channels and the calcium-activated anion channels in rat thyroid cells: implications for iodide homeostasis. J Cell Physiol 2013; 228:814-23. [PMID: 23018590 DOI: 10.1002/jcp.24230] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 09/18/2012] [Indexed: 01/28/2023]
Abstract
The initial step in a synthesis of thyroid hormones is the uptake of iodide from the circulation. Iodide (I(-)) is transported into thyroid cells via a Na(+)/I(-) symporter (NIS), which is electrogenic and thus sensitive to alterations in membrane potential (V(m)). I(-) is then released to the lumen of thyroid follicles where the hormones are synthesised and stored. The mechanisms of I(-) release to follicle lumen are poorly characterised. Our whole-cell voltage clamp recordings revealed the presence of a Ca(2+) activated Cl(-) current (CaCC) in Fisher rat thyroid cell line 5 (FRTL-5). Transcripts of anoctamin 1 (ANO1) and anoctamin 10 (ANO10), putative molecular constituents of CaCC, were detected. The anion channels underlying CaCC are highly permeable to I(-). Both niflumic acid (NFA) and 2-aminoethyl diphenylborinate (2-APB), antagonists of CaCC and transient receptor potential channels, respectively, inhibited CaCC. Canonical transient receptor potential channel 2 (TRPC2) is the only TRPC member present in FRTL-5 cells. The activation rate of CaCC was markedly slower in shTRPC2 knock-down cells, indicating that Ca(2+) entry via TRPC2 contributes to CaCC activation. The uptake of iodide was enhanced and the resting V(m) was more depolarised in TRPC2 knock-down cells. We suggest that the interplay between TRPC2 and ANO1 may have dual effects on iodide transport, modulating I(-) release via ANO channels and I(-) uptake via the V(m) sensitive NIS.
Collapse
Affiliation(s)
- Tero M Viitanen
- Department of Biosciences, Åbo Akademi University, Turku, Finland
| | | | | | | |
Collapse
|
87
|
Ozkan MH, Ozturk EI, Uma S. Electrical field stimulation (EFS)-induced relaxations turn into contractions upon removal of extracellular calcium in rat mesenteric artery. Pharmacol Res 2013; 70:60-5. [DOI: 10.1016/j.phrs.2013.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 12/03/2012] [Accepted: 01/04/2013] [Indexed: 10/27/2022]
|
88
|
Cheon B, Lee HC, Wakai T, Fissore RA. Ca2+ influx and the store-operated Ca2+ entry pathway undergo regulation during mouse oocyte maturation. Mol Biol Cell 2013; 24:1396-410. [PMID: 23468522 PMCID: PMC3639051 DOI: 10.1091/mbc.e13-01-0065] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Changes in Ca2+ homeostasis render oocytes competent to undergo [Ca2+]i oscillations and activation. During mouse oocyte maturation Ca2+ influx and SOCE are down-regulated, whereas [Ca2+]ER content increases. Bypassing the down-regulation of Ca2+ influx disturbs oocyte maturation. In preparation for fertilization, mammalian oocytes undergo optimization of the mechanisms that regulate calcium homeostasis. Among these changes is the increase in the content of the Ca2+ stores ([Ca2+]ER), a process that requires Ca2+ influx. Nevertheless, the mechanism(s) that mediates this influx remains obscure, although is known that [Ca2+]ER can regulate Ca2+ influx via store-operated Ca2+ entry (SOCE). We find that during maturation, as [Ca2+]ER increases, Ca2+ influx decreases. We demonstrate that mouse oocytes/eggs express the two molecular components of SOCE—stromal interaction molecule 1 (Stim1) and Orai1—and expression of human (h) Stim1 increases Ca2+ influx in a manner that recapitulates endogenous SOCE. We observe that the cellular distribution of hStim1 and hOrai1 during maturation undergoes sweeping changes that curtail their colocalization during the later stages of maturation. Coexpression of hStim1 and hOrai1 enhances influx throughout maturation but increases basal Ca2+ levels only in GV oocytes. Further, expression of a constitutive active form of hStim1 plus Orai1, which increases basal Ca2+ throughout maturation, disturbs resumption of meiosis. Taken together, our results demonstrate that Ca2+ influx and SOCE are regulated during maturation and that alteration of Ca2+ homeostasis undermines maturation in mouse oocytes.
Collapse
Affiliation(s)
- Banyoon Cheon
- Molecular and Cellular Biology Program, University of Massachusetts, Amherst, MA 01002, USA
| | | | | | | |
Collapse
|
89
|
Wang HJ, Martin AG, Chao PK, Reichard RA, Martin AL, Huang YW, Chan MH, Aronstam RS. Honokiol blocks store operated calcium entry in CHO cells expressing the M3 muscarinic receptor: honokiol and muscarinic signaling. J Biomed Sci 2013; 20:11. [PMID: 23432810 PMCID: PMC3599152 DOI: 10.1186/1423-0127-20-11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 02/22/2013] [Indexed: 01/15/2023] Open
Abstract
Background Honokiol, a cell-permeable phenolic compound derived from the bark of magnolia trees and present in Asian herbal teas, has a unique array of pharmacological actions, including the inhibition of multiple autonomic responses. We determined the effects of honokiol on calcium signaling underlying transmission mediated by human M3 muscarinic receptors expressed in Chinese hamster ovary (CHO) cells. Receptor binding was determined in radiolabelled ligand binding assays; changes in intracellular calcium concentrations were determined using a fura-2 ratiometric imaging protocol; cytotoxicity was determined using a dye reduction assay. Results Honokiol had a potent (EC50 ≈ 5 μmol/l) inhibitory effect on store operated calcium entry (SOCE) that was induced by activation of the M3 receptors. This effect was specific, rapid and partially reversible, and was seen at concentrations not associated with cytotoxicity, inhibition of IP3 receptor-mediated calcium release, depletion of ER calcium stores, or disruption of M3 receptor binding. Conclusions It is likely that an inhibition of SOCE contributes to honokiol disruption of parasympathetic motor functions, as well as many of its beneficial pharmacological properties.
Collapse
Affiliation(s)
- Hsiu-Jen Wang
- Department of Biological Sciences, Missouri University of Science & Technology, 400 W, 11th St, Rolla MO 65409, USA
| | | | | | | | | | | | | | | |
Collapse
|
90
|
Böhm J, Chevessier F, Maues De Paula A, Koch C, Attarian S, Feger C, Hantaï D, Laforêt P, Ghorab K, Vallat JM, Fardeau M, Figarella-Branger D, Pouget J, Romero NB, Koch M, Ebel C, Levy N, Krahn M, Eymard B, Bartoli M, Laporte J. Constitutive activation of the calcium sensor STIM1 causes tubular-aggregate myopathy. Am J Hum Genet 2013; 92:271-8. [PMID: 23332920 DOI: 10.1016/j.ajhg.2012.12.007] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 10/31/2012] [Accepted: 12/14/2012] [Indexed: 01/20/2023] Open
Abstract
Tubular aggregates are regular arrays of membrane tubules accumulating in muscle with age. They are found as secondary features in several muscle disorders, including alcohol- and drug-induced myopathies, exercise-induced cramps, and inherited myasthenia, but also exist as a pure genetic form characterized by slowly progressive muscle weakness. We identified dominant STIM1 mutations as a genetic cause of tubular-aggregate myopathy (TAM). Stromal interaction molecule 1 (STIM1) is the main Ca(2+) sensor in the endoplasmic reticulum, and all mutations were found in the highly conserved intraluminal Ca(2+)-binding EF hands. Ca(2+) stores are refilled through a process called store-operated Ca(2+) entry (SOCE). Upon Ca(2+)-store depletion, wild-type STIM1 oligomerizes and thereby triggers extracellular Ca(2+) entry. In contrast, the missense mutations found in our four TAM-affected families induced constitutive STIM1 clustering, indicating that Ca(2+) sensing was impaired. By monitoring the calcium response of TAM myoblasts to SOCE, we found a significantly higher basal Ca(2+) level in TAM cells and a dysregulation of intracellular Ca(2+) homeostasis. Because recessive STIM1 loss-of-function mutations were associated with immunodeficiency, we conclude that the tissue-specific impact of STIM1 loss or constitutive activation is different and that a tight regulation of STIM1-dependent SOCE is fundamental for normal skeletal-muscle structure and function.
Collapse
Affiliation(s)
- Johann Böhm
- Département de Médecine Translationelle et Neurogénétique, Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Horinouchi T, Terada K, Higashi T, Miwa S. Endothelin Receptor Signaling: New Insight Into Its Regulatory Mechanisms. J Pharmacol Sci 2013; 123:85-101. [DOI: 10.1254/jphs.13r02cr] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
92
|
Derler I, Schindl R, Fritsch R, Heftberger P, Riedl MC, Begg M, House D, Romanin C. The action of selective CRAC channel blockers is affected by the Orai pore geometry. Cell Calcium 2012; 53:139-51. [PMID: 23218667 PMCID: PMC3580291 DOI: 10.1016/j.ceca.2012.11.005] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 11/02/2012] [Accepted: 11/03/2012] [Indexed: 11/10/2022]
Abstract
As the molecular composition of calcium-release activated calcium (CRAC) channels has been unknown for two decades, elucidation of selective inhibitors has been considerably hampered. By the identification of the two key components of CRAC channels, STIM1 and Orai1 have emerged as promising targets for CRAC blockers. The aim of this study was to thoroughly characterize the effects of two selective CRAC channel blockers on currents derived from STIM1/Orai heterologoulsy expressed in HEK293 cells. The novel compounds GSK-7975A and GSK-5503A were tested for effects on STIM1 mediated Orai1 or Orai3 currents by whole-cell patch-clamp recordings and for the effects on STIM1 oligomerisation or STIM1/Orai coupling by FRET microscopy. To investigate their site of action, inhibitory effects of these molecules were explored using Orai pore mutants. The GSK blockers inhibited Orai1 and Orai3 currents with an IC50 of approximately 4 μM and exhibited a substantially slower rate of onset than the typical pore blocker La3+, together with almost no current recovery upon wash-out over 4 min. For the less Ca2+-selective Orai1 E106D pore mutant, ICRAC inhibition was significantly reduced. FRET experiments indicated that neither STIM1–STIM1 oligomerization nor STIM1–Orai1 coupling was affected by these compounds. These CRAC channel blockers are acting downstream of STIM1 oligomerization and STIM1/Orai1 interaction, potentially via an allosteric effect on the selectivity filter of Orai. The elucidation of these CRAC current blockers represents a significant step toward the identification of CRAC channel-selective drug compounds.
Collapse
Affiliation(s)
- Isabella Derler
- Institute of Biophysics, University of Linz, 4040 Linz, Austria
| | | | | | | | | | | | | | | |
Collapse
|
93
|
Lodola F, Laforenza U, Bonetti E, Lim D, Dragoni S, Bottino C, Ong HL, Guerra G, Ganini C, Massa M, Manzoni M, Ambudkar IS, Genazzani AA, Rosti V, Pedrazzoli P, Tanzi F, Moccia F, Porta C. Store-operated Ca2+ entry is remodelled and controls in vitro angiogenesis in endothelial progenitor cells isolated from tumoral patients. PLoS One 2012; 7:e42541. [PMID: 23049731 PMCID: PMC3458053 DOI: 10.1371/journal.pone.0042541] [Citation(s) in RCA: 115] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Accepted: 07/09/2012] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Endothelial progenitor cells (EPCs) may be recruited from bone marrow to sustain tumor vascularisation and promote the metastatic switch. Understanding the molecular mechanisms driving EPC proliferation and tubulogenesis could outline novel targets for alternative anti-angiogenic treatments. Store-operated Ca(2+) entry (SOCE), which is activated by a depletion of the intracellular Ca(2+) pool, regulates the growth of human EPCs, where is mediated by the interaction between the endoplasmic reticulum Ca(2+)-sensor, Stim1, and the plasmalemmal Ca(2+) channel, Orai1. As oncogenesis may be associated to the capability of tumor cells to grow independently on Ca(2+) influx, it is important to assess whether SOCE regulates EPC-dependent angiogenesis also in tumor patients. METHODOLOGY/PRINCIPAL FINDINGS The present study employed Ca(2+) imaging, recombinant sub-membranal and mitochondrial aequorin, real-time polymerase chain reaction, gene silencing techniques and western blot analysis to investigate the expression and the role of SOCE in EPCs isolated from peripheral blood of patients affected by renal cellular carcinoma (RCC; RCC-EPCs) as compared to control EPCs (N-EPCs). SOCE, activated by either pharmacological (i.e. cyclopiazonic acid) or physiological (i.e. ATP) stimulation, was significantly higher in RCC-EPCs and was selectively sensitive to BTP-2, and to the trivalent cations, La(3+) and Gd(3+). Furthermore, 2-APB enhanced thapsigargin-evoked SOCE at low concentrations, whereas higher doses caused SOCE inhibition. Conversely, the anti-angiogenic drug, carboxyamidotriazole (CAI), blocked both SOCE and the intracellular Ca(2+) release. SOCE was associated to the over-expression of Orai1, Stim1, and transient receptor potential channel 1 (TRPC1) at both mRNA and protein level The intracellular Ca(2+) buffer, BAPTA, BTP-2, and CAI inhibited RCC-EPC proliferation and tubulogenesis. The genetic suppression of Stim1, Orai1, and TRPC1 blocked CPA-evoked SOCE in RCC-EPCs. CONCLUSIONS SOCE is remodelled in EPCs from RCC patients and stands out as a novel molecular target to interfere with RCC vascularisation due to its ability to control proliferation and tubulogenesis.
Collapse
Affiliation(s)
- Francesco Lodola
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Umberto Laforenza
- Section of Human Physiology, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Elisa Bonetti
- Clinical Epidemiology Laboratory Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Dmitry Lim
- Department of Pharmaceutical Sciences, University of Eastern Piedmont “Amedeo Avogadro”, Novara, Italy
| | - Silvia Dragoni
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Cinzia Bottino
- Section of Human Physiology, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Hwei Ling Ong
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Germano Guerra
- Department of Health Sciences, University of Molise, Campobasso, Italy
| | - Carlo Ganini
- Medical Oncology IRCCS Policlinico San Matteo, Pavia, Italy
| | - Margherita Massa
- Laboratory of Biotechnology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Indu S. Ambudkar
- Secretory Physiology Section, Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Armando A. Genazzani
- Department of Pharmaceutical Sciences, University of Eastern Piedmont “Amedeo Avogadro”, Novara, Italy
| | - Vittorio Rosti
- Clinical Epidemiology Laboratory Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | | | - Franco Tanzi
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Francesco Moccia
- Department of Biology and Biotechnology “Lazzaro Spallanzani”, University of Pavia, Pavia, Italy
| | - Camillo Porta
- Medical Oncology IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
94
|
Ercan E, Chung SH, Bhardwaj R, Seedorf M. Di-Arginine Signals and the K-Rich Domain Retain the Ca2+Sensor STIM1 in the Endoplasmic Reticulum. Traffic 2012; 13:992-1003. [DOI: 10.1111/j.1600-0854.2012.01359.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 03/28/2012] [Accepted: 04/12/2012] [Indexed: 11/30/2022]
Affiliation(s)
| | - Shan-Hua Chung
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH); DKFZ-ZMBH Alliance; Im Neuenheimer Feld 282; 69120; Heidelberg; Germany
| | - Rajesh Bhardwaj
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH); DKFZ-ZMBH Alliance; Im Neuenheimer Feld 282; 69120; Heidelberg; Germany
| | - Matthias Seedorf
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH); DKFZ-ZMBH Alliance; Im Neuenheimer Feld 282; 69120; Heidelberg; Germany
| |
Collapse
|
95
|
Inhibition of store-operated Ca2+ channels prevent ethanol-induced intracellular Ca2+ increase and cell injury in a human hepatoma cell line. Toxicol Lett 2012; 208:254-61. [DOI: 10.1016/j.toxlet.2011.11.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Revised: 11/06/2011] [Accepted: 11/08/2011] [Indexed: 12/13/2022]
|
96
|
Structure, regulation and biophysics of I(CRAC), STIM/Orai1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:383-410. [PMID: 22453951 DOI: 10.1007/978-94-007-2888-2_16] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Ca(2+) release activated Ca(2+) (CRAC) channels mediate robust Ca(2+) influx when the endoplasmic reticulum Ca(2+) stores are depleted. This essential process for T-cell activation as well as degranulation of mast cells involves the Ca(2+) sensor STIM1, located in the endoplasmic reticulum and the Ca(2+) selective Orai1 channel in the plasma membrane. Our review describes the CRAC signaling pathway, the activation of which is initiated by a drop in the endoplasmic Ca(2+) level sensed by STIM1. This in term induces multimerisation and puncta-formation of STIM1 proteins is followed by their coupling to and activation of Orai channels. Consequently Ca(2+) entry is triggered through the Orai pore into the cytosol with subsequent closure of the channel by Ca(2+)-dependent inactivation. We will portray a mechanistic view of the events coupling STIM1 to Orai activation based on their structure and biophysics.
Collapse
|
97
|
Moreno C, Vaca L. SOC and now also SIC: store-operated and store-inhibited channels. IUBMB Life 2011; 63:856-63. [PMID: 21901816 DOI: 10.1002/iub.547] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2011] [Accepted: 07/02/2011] [Indexed: 12/19/2022]
Abstract
There is a specialized form of calcium influx that involves a close communication between endoplasmic reticulum and the channels at the plasma membrane. In one side store depletion activates channels known as store-operated channels (SOC), which are responsible of the well-studied store-operated calcium entry (SOCE). SOC comprises two different types of channels. Orai, which is exclusively activated by store depletion being the channel responsible of the calcium release-activated calcium current, and transient receptor potential canonical channel, which in contrast, is activated by store depletion only under specific conditions and carries nonselective cationic currents. On the other hand, it has been recently shown that store depletion also inhibits calcium channels. The first member identified, of what we named as store-inhibited channels (SIC), is the L-type voltage-gated calcium channel. Stores control both SOC and SIC by means of the multifunctional protein STIM1. The identification of SOC and SIC opens a new scenario for the role of store depletion in the modulation of different calcium entry pathways, which may satisfy different cellular processes.
Collapse
Affiliation(s)
- Claudia Moreno
- Departamento de Biología Celular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Del. Coyoacán, 04510 México DF, México
| | | |
Collapse
|
98
|
Zbidi H, Jardin I, Woodard GE, Lopez JJ, Berna-Erro A, Salido GM, Rosado JA. STIM1 and STIM2 are located in the acidic Ca2+ stores and associates with Orai1 upon depletion of the acidic stores in human platelets. J Biol Chem 2011; 286:12257-70. [PMID: 21321120 PMCID: PMC3069429 DOI: 10.1074/jbc.m110.190694] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 02/11/2011] [Indexed: 11/06/2022] Open
Abstract
Mammalian cells accumulate Ca2+ into agonist-sensitive acidic organelles, vesicles that possess a vacuolar proton-ATPase. Acidic Ca2+ stores include secretory granules and lysosome-related organelles. Current evidence clearly indicates that acidic Ca2+ stores participate in cell signaling and function, including the activation of store-operated Ca2+ entry in human platelets upon depletion of the acidic stores, although the mechanism underlying the activation of store-operated Ca2+ entry controlled by the acidic stores remains unclear. STIM1 has been presented as the endoplasmic reticulum Ca2+ sensor, but its role sensing intraluminal Ca2+ concentration in the acidic stores has not been investigated. Here we report that STIM1 and STIM2 are expressed in the lysosome-related organelles and dense granules in human platelets isolated by immunomagnetic sorting. Depletion of the acidic Ca2+ stores using the specific vacuolar proton-ATPase inhibitor, bafilomycin A1, enhanced the association between STIM1 and STIM2 as well as between these proteins and the plasma membrane channel Orai1. Depletion of the acidic Ca2+ stores also induces time-dependent co-immunoprecipitation of STIM1 with the TRPC proteins hTRPC1 and hTRPC6, as well as between Orai1 and both TRPC proteins. In addition, bafilomycin A1 enhanced the association between STIM2 and SERCA3. These findings demonstrate the location of STIM1 and STIM2 in the acidic Ca2+ stores and their association with Ca2+ channels and ATPases upon acidic stores discharge.
Collapse
Affiliation(s)
- Hanene Zbidi
- From the Department of Physiology (Cell Physiology Research Group) University of Extremadura, 10003 Cáceres, Spain
| | - Isaac Jardin
- From the Department of Physiology (Cell Physiology Research Group) University of Extremadura, 10003 Cáceres, Spain
| | | | - Jose J. Lopez
- Hémostase et Dynamique Cellulaire Vasculaire U770, INSERM, 94276 Le Kremlin-Bicêtre, France
| | - Alejandro Berna-Erro
- From the Department of Physiology (Cell Physiology Research Group) University of Extremadura, 10003 Cáceres, Spain
| | - Ginés M. Salido
- From the Department of Physiology (Cell Physiology Research Group) University of Extremadura, 10003 Cáceres, Spain
| | - Juan A. Rosado
- From the Department of Physiology (Cell Physiology Research Group) University of Extremadura, 10003 Cáceres, Spain
| |
Collapse
|
99
|
Yamashita M, Somasundaram A, Prakriya M. Competitive modulation of Ca2+ release-activated Ca2+ channel gating by STIM1 and 2-aminoethyldiphenyl borate. J Biol Chem 2011; 286:9429-42. [PMID: 21193399 PMCID: PMC3059062 DOI: 10.1074/jbc.m110.189035] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 12/03/2010] [Indexed: 11/06/2022] Open
Abstract
Activation of Ca(2+) release-activated Ca(2+) channels by depletion of intracellular Ca(2+) stores involves physical interactions between the endoplasmic reticulum Ca(2+) sensor, STIM1, and the channels composed of Orai subunits. Recent studies indicate that the Orai3 subtype, in addition to being store-operated, is also activated in a store-independent manner by 2-aminoethyldiphenyl borate (2-APB), a small molecule with complex pharmacology. However, it is unknown whether the store-dependent and -independent activation modes of Orai3 channels operate independently or whether there is cross-talk between these activation states. Here we report that in addition to causing direct activation, 2-APB also regulates store-operated gating of Orai3 channels, causing potentiation at low doses and inhibition at high doses. Inhibition of store-operated gating by 2-APB was accompanied by the suppression of several modes of Orai3 channel regulation that depend on STIM1, suggesting that high doses of 2-APB interrupt STIM1-Orai3 coupling. Conversely, STIM1-bound Orai3 (and Orai1) channels resisted direct gating by high doses of 2-APB. The rate of direct 2-APB activation of Orai3 channels increased linearly with the degree of STIM1-Orai3 uncoupling, suggesting that 2-APB has to first disengage STIM1 before it can directly gate Orai3 channels. Collectively, our results indicate that the store-dependent and -independent modes of Ca(2+) release-activated Ca(2+) channel activation are mutually exclusive: channels bound to STIM1 resist 2-APB gating, whereas 2-APB antagonizes STIM1 gating.
Collapse
Affiliation(s)
- Megumi Yamashita
- From the Department of Molecular Pharmacology and Biological Chemistry, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Agila Somasundaram
- From the Department of Molecular Pharmacology and Biological Chemistry, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| | - Murali Prakriya
- From the Department of Molecular Pharmacology and Biological Chemistry, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611
| |
Collapse
|