1
|
Liu R, Krüger K, Pilat C, Fan W, Xiao Y, Seimetz M, Ringseis R, Baumgart-Vogt E, Eder K, Weissmann N, Mooren FC. Excessive Accumulation of Intracellular Ca 2+ After Acute Exercise Potentiated Impairment of T-cell Function. Front Physiol 2021; 12:728625. [PMID: 34899372 PMCID: PMC8662941 DOI: 10.3389/fphys.2021.728625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 10/18/2021] [Indexed: 11/26/2022] Open
Abstract
Ca2+ is an important intracellular second messenger known to regulate several cellular functions. This research aimed to investigate the mechanisms of exercise-induced immunosuppression by measuring intracellular calcium levels, Ca2+-regulating gene expression, and agonist-evoked proliferation of murine splenic T lymphocytes. Mice were randomly assigned to the control, sedentary group (C), and three experimental groups, which performed a single bout of intensive and exhaustive treadmill exercise. Murine splenic lymphocytes were separated by density-gradient centrifugation immediately (E0), 3h (E3), and 24h after exercise (E24). Fura-2/AM was used to monitor cytoplasmic free Ca2+ concentration in living cells. The combined method of carboxyfluorescein diacetate succinimidyl ester (CFSE) labeling and flow cytometry was used for the detection of T cell proliferation. The transcriptional level of Ca2+-regulating genes was quantified by using qPCR. Both basal intracellular Ca2+ levels and agonist (ConA, OKT3, or thapsigargin)-induced Ca2+ transients were significantly elevated at E3 group (p<0.05 vs. control). However, mitogen-induced cell proliferation was significantly decreased at E3 group (p<0.05 vs. control). In parallel, the transcriptional level of plasma membrane Ca2+-ATPases (PMCA), sarco/endoplasmic reticulum Ca2+-ATPases (SERCA), TRPC1, and P2X7 was significantly downregulated, and the transcriptional level of IP3R2 and RyR2 was significantly upregulated in E3 (p<0.01 vs. control). In summary, this study demonstrated that acute exercise affected intracellular calcium homeostasis, most likely by enhancing transmembrane Ca2+ influx into cells and by reducing expression of Ca2+-ATPases such as PMCA and SERCA. However, altered Ca2+ signals were not transduced into an enhanced T cell proliferation suggesting other pathways to be responsible for the transient exercise-associated immunosuppression.
Collapse
Affiliation(s)
- Renyi Liu
- Department of Physical Education, China University of Geosciences (Wuhan), Wuhan, China.,Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Giessen, Germany
| | - Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Giessen, Germany
| | - Christian Pilat
- Department of Exercise Physiology and Sports Therapy, Institute of Sports Science, Justus-Liebig-University Giessen, Giessen, Germany
| | - Wei Fan
- Institute for Anatomy and Cell Biology II, Justus-Liebig-University Giessen, Giessen, Germany
| | - Yu Xiao
- Institute for Anatomy and Cell Biology II, Justus-Liebig-University Giessen, Giessen, Germany
| | - Michael Seimetz
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Robert Ringseis
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Eveline Baumgart-Vogt
- Institute for Anatomy and Cell Biology II, Justus-Liebig-University Giessen, Giessen, Germany
| | - Klaus Eder
- Institute of Animal Nutrition and Nutrition Physiology, Justus-Liebig-University Giessen, Giessen, Germany
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Lung Center (DZL), Justus-Liebig-University Giessen, Giessen, Germany
| | - Frank Christoph Mooren
- Department of Rehabilitation Sciences, Faculty of Health, University of Witten/Herdecke, Witten, Germany
| |
Collapse
|
2
|
Cheng HH, Liang WZ, Kuo CC, Hao LJ, Chou CT, Jan CR. The exploration of effect of terfenadine on Ca 2+ signaling in renal tubular cells. J Recept Signal Transduct Res 2019; 39:73-79. [PMID: 31184240 DOI: 10.1080/10799893.2019.1620777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Terfenadine, an antihistamine used for the treatment of allergic conditions, affected Ca2+-related physiological responses in various models. However, the effect of terfenadine on cytosolic free Ca2+ levels ([Ca2+]i) and its related physiology in renal tubular cells is unknown. This study examined whether terfenadine altered Ca2+ signaling and caused cytotoxicity in Madin-Darby canine kidney (MDCK) renal tubular cells. The Ca2+-sensitive fluorescent dye fura-2 was used to measure [Ca2+]i. Cell viability was measured by the fluorescent reagent 4-[3-[4-lodophenyl]-2-4(4-nitrophenyl)-2H-5-tetrazolio-1,3-benzene disulfonate] water soluble tetrazolium-1 (WST-1) assay. Terfenadine at concentrations of 100-1000 μM induced [Ca2+]i rises concentration dependently. The response was reduced by approximately 35% by removing extracellular Ca2+. In Ca2+-free medium, treatment with the endoplasmic reticulum Ca2+ pump inhibitor 2,5-di-tert-butylhydroquinone (BHQ) partly inhibited terfenadine-evoked [Ca2+]i rises. Conversely, treatment with terfenadine abolished BHQ-evoked [Ca2+]i rises. Inhibition of phospholipase C (PLC) with U73122 inhibited 95% of terfenadine-induced Ca2+ release. Terfenadine-induced Ca2+ entry was supported by Mn2+-caused quenching of fura-2 fluorescence. Terfenadine-induced Ca2+ entry was partly inhibited by an activator of protein kinase C (PKC), phorbol 12-myristate 13 acetate (PMA) and by three modulators of store-operated Ca2+ channels (nifedipine, econazole, and SKF96365). Terfenadine at 200-300 μM decreased cell viability, which was not reversed by pretreatment with the Ca2+ chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-acetoxymethyl ester (BAPTA/AM). Together, in MDCK cells, terfenadine induced [Ca2+]i rises by evoking PLC-dependent Ca2+ release from the endoplasmic reticulum and Ca2+ entry via PKC-sensitive store-operated Ca2+ entry. Furthermore, terfenadine caused cell death that was not triggered by preceding [Ca2+]i rises.
Collapse
Affiliation(s)
- He-Hsiung Cheng
- a Department of Medicine , Chang Bing Show Chwan Memorial Hospital , Changhua , Taiwan
| | - Wei-Zhe Liang
- b Department of Medical Education and Research , Kaohsiung Veterans General Hospital , Kaohsiung , Taiwan.,c Department of Pharmacy , Tajen University , Pingtung , Taiwan
| | - Chun-Chi Kuo
- d Department of Nursing , Tzu Hui Institute of Technology , Pingtung , Taiwan
| | - Lyh-Jyh Hao
- e Department of Metabolism , Kaohsiung Veterans General Hospital Tainan Branch , Tainan , Taiwan
| | - Chiang-Ting Chou
- f Department of Nursing, Division of Basic Medical Sciences , Chang Gung University of Science and Technology , Chia-Yi , Taiwan
| | - Chung-Ren Jan
- b Department of Medical Education and Research , Kaohsiung Veterans General Hospital , Kaohsiung , Taiwan
| |
Collapse
|
3
|
Lopez JJ, Salido GM, Rosado JA. Cardiovascular and Hemostatic Disorders: SOCE and Ca 2+ Handling in Platelet Dysfunction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 993:453-472. [PMID: 28900928 DOI: 10.1007/978-3-319-57732-6_23] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Among the Ca2+ entry mechanisms in platelets, store-operated Ca2+ entry (SOCE) plays a prominent role as it is necessary to achieve full activation of platelet functions and replenish intracellular Ca2+ stores. In platelets, as in other non-excitable cells, SOCE has been reported to involve the activation of plasma membrane channels by the ER Ca2+ sensor STIM1. Despite electrophysiological studies are not possible in human platelets, indirect analyses have revealed that the Ca2+-permeable channels involve Orai1 and, most likely, TRPC1 subunits. A relevant role for the latter has not been found in mouse platelets. There is a body of evidence revealing a number of abnormalities in SOCE or in its molecular regulators that result in qualitative platelet disorders and, as a consequence, altered platelet responsiveness upon stimulation with multiple physiological agonists. Platelet SOCE abnormalities include STIM1 and Orai1 mutations. This chapter summarizes the current knowledge in this field, as well as the disorders associated to platelet SOCE dysfunction.
Collapse
Affiliation(s)
- Jose J Lopez
- Cell Physiology Research Group, Department of Physiology, University of Extremadura, Cáceres, Spain
| | - Gines M Salido
- Cell Physiology Research Group, Department of Physiology, University of Extremadura, Cáceres, Spain
| | - Juan A Rosado
- Cell Physiology Research Group, Department of Physiology, University of Extremadura, Cáceres, Spain.
| |
Collapse
|
4
|
Bouron A, Chauvet S, Dryer S, Rosado JA. Second Messenger-Operated Calcium Entry Through TRPC6. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:201-49. [PMID: 27161231 DOI: 10.1007/978-3-319-26974-0_10] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Canonical transient receptor potential 6 (TRPC6) proteins assemble into heteromultimeric structures forming non-selective cation channels. In addition, many TRPC6-interacting proteins have been identified like some enzymes, channels, pumps, cytoskeleton-associated proteins, immunophilins, or cholesterol-binding proteins, indicating that TRPC6 are engaged into macromolecular complexes. Depending on the cell type and the experimental conditions used, TRPC6 activity has been reported to be controlled by diverse modalities. For instance, the second messenger diacylglycerol, store-depletion, the plant extract hyperforin or H2O2 have all been shown to trigger the opening of TRPC6 channels. A well-characterized consequence of TRPC6 activation is the elevation of the cytosolic concentration of Ca(2+). This latter response can reflect the entry of Ca(2+) through open TRPC6 channels but it can also be due to the Na(+)/Ca(2+) exchanger (operating in its reverse mode) or voltage-gated Ca(2+) channels (recruited in response to a TRPC6-mediated depolarization). Although TRPC6 controls a diverse array of biological functions in many tissues and cell types, its pathophysiological functions are far from being fully understood. This chapter covers some key features of TRPC6, with a special emphasis on their biological significance in kidney and blood cells.
Collapse
Affiliation(s)
- Alexandre Bouron
- Université Grenoble Alpes, 38000, Grenoble, France.
- CNRS, iRTSV-LCBM, 38000, Grenoble, France.
| | - Sylvain Chauvet
- Université Grenoble Alpes, 38000, Grenoble, France
- CNRS, iRTSV-LCBM, 38000, Grenoble, France
| | - Stuart Dryer
- University of Houston, Houston, TX, USA
- Baylor College of Medicine, Houston, TX, USA
| | - Juan A Rosado
- Departamento de Fisiología, University of Extremadura, Cáceres, Spain
| |
Collapse
|
5
|
Albarran L, Lopez JJ, Salido GM, Rosado JA. Historical Overview of Store-Operated Ca(2+) Entry. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:3-24. [PMID: 27161222 DOI: 10.1007/978-3-319-26974-0_1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Calcium influx is an essential mechanism for the activation of cellular functions both in excitable and non-excitable cells. In non-excitable cells, activation of phospholipase C by occupation of G protein-coupled receptors leads to the generation of inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG), which, in turn, initiate two Ca(2+) entry pathways: Ca(2+) release from intracellular Ca(2+) stores, signaled by IP3, leads to the activation of store-operated Ca(2+) entry (SOCE); on the other hand, DAG activates a distinct second messenger-operated pathway. SOCE is regulated by the filling state of the intracellular calcium stores. The search for the molecular components of SOCE has identified the stromal interaction molecule 1 (STIM1) as the Ca(2+) sensor in the endoplasmic reticulum and Orai1 as a store-operated channel (SOC) subunit. Furthermore, a number of reports have revealed that several members of the TRPC family of channels also take part of the SOC macromolecular complex. This introductory chapter summarizes the early pieces of evidence that led to the concept of SOCE and the components of the store-operated signaling pathway.
Collapse
Affiliation(s)
- Letizia Albarran
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, Av. Universidad s/n, 10003, Cáceres, Spain
| | - Jose J Lopez
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, Av. Universidad s/n, 10003, Cáceres, Spain
| | - Ginés M Salido
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, Av. Universidad s/n, 10003, Cáceres, Spain
| | - Juan A Rosado
- Departamento de Fisiología, University of Extremadura, Cáceres, Spain.
| |
Collapse
|
6
|
Berna-Erro A, Jardín I, Smani T, Rosado JA. Regulation of Platelet Function by Orai, STIM and TRP. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 898:157-81. [PMID: 27161229 DOI: 10.1007/978-3-319-26974-0_8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Agonist-induced changes in cytosolic Ca(2+) concentration ([Ca(2+)]c) are central events in platelet physiology. A major mechanism supporting agonist-induced Ca(2+) signals is store-operated Ca(2+) entry (SOCE), where the Ca(2+) sensor STIM1 and the channels of the Orai family, as well as TRPC members are the key elements. STIM1-dependent SOCE plays a major role in collagen-stimulated Ca(2+) signaling, phosphatidylserine exposure and thrombin generation. Furthermore, studies involving Orai1 gain-of-function mutants and platelets from Orai1-deficient mice have revealed the importance of this channel in thrombosis and hemostasis to those found in STIM1-deficient mice indicating that SOCE might play a prominent role in thrombus formation. Moreover, increase in TRPC6 expression might lead to thrombosis in humans. The role of STIM1, Orai1 and TRPCs, and thus SOCE, in thrombus formation, suggests that therapies directed against SOCE and targeting these molecules during cardiovascular and cerebrovascular events could significantly improve traditional anti-thrombotic treatments.
Collapse
Affiliation(s)
- Alejandro Berna-Erro
- Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona, 08003, Spain
| | - Isaac Jardín
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, Cáceres, 10003, Spain
| | - Tarik Smani
- Department of Medical Physiology and Biophysic, Institute of Biomedicine of Seville (IBiS), University Hospital of Virgen del Rocío/CSIC/University of Seville, Sevilla, 41013, Spain
| | - Juan A Rosado
- Departamento de Fisiología, University of Extremadura, Cáceres, Spain.
| |
Collapse
|
7
|
Redondo PC, Rosado JA. Store-operated calcium entry: unveiling the calcium handling signalplex. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 316:183-226. [PMID: 25805125 DOI: 10.1016/bs.ircmb.2015.01.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Store-operated Ca(2+) entry (SOCE) is an important mechanism for Ca(2+) influx in non-excitable cells, also present in excitable cells. The activation of store-operated channels (SOCs) is finely regulated by the filling state of the intracellular agonist-sensitive Ca(2+) compartments, and both, the mechanism of sensing the Ca(2+) stores and the nature and functional properties of the SOCs, have been a matter of intense investigation and debate. The identification of STIM1 as the endoplasmic reticulum Ca(2+) sensor and both Orai1, as the pore-forming subunit of the channels mediating the Ca(2+)-selective store-operated current, and the members of the TRPC subfamily of proteins, as the channels mediating the cation-permeable SOCs, has shed new light on the underlying events. This review summarizes the initial hypothesis and the current advances on the mechanism of activation of SOCE.
Collapse
Affiliation(s)
- Pedro C Redondo
- Department of Physiology, University of Extremadura, Cáceres, Spain
| | - Juan A Rosado
- Department of Physiology, University of Extremadura, Cáceres, Spain
| |
Collapse
|
8
|
Harper MT, Londono JEC, Quick K, Londono JC, Flockerzi V, Philipp SE, Birnbaumer L, Freichel M, Poole AW. Transient Receptor Potential Channels Function as a Coincidence Signal Detector Mediating Phosphatidylserine Exposure. Sci Signal 2013; 6:ra50. [DOI: 10.1126/scisignal.2003701] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
9
|
Jardin I, Dionisio N, Frischauf I, Berna-Erro A, Woodard GE, López JJ, Salido GM, Rosado JA. The polybasic lysine-rich domain of plasma membrane-resident STIM1 is essential for the modulation of store-operated divalent cation entry by extracellular calcium. Cell Signal 2013; 25:1328-37. [PMID: 23395841 DOI: 10.1016/j.cellsig.2013.01.025] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 01/09/2013] [Accepted: 01/23/2013] [Indexed: 01/16/2023]
Abstract
STIM1 acts as an endoplasmic reticulum Ca(2+) sensor that communicates the filling state of the intracellular stores to the store-operated channels. In addition, STIM1 is expressed in the plasma membrane, with the Ca(2+) binding EF-hand motif facing the extracellular medium; however, its role sensing extracellular Ca(2+) concentrations in store-operated Ca(2+) entry (SOCE), as well as the underlying mechanism remains unclear. Here we report that divalent cation entry stimulated by thapsigargin (TG) is attenuated by extracellular Ca(2+) in a concentration-dependent manner. Expression of the Ca(2+)-binding defective STIM1(D76A) mutant did not alter the surface expression of STIM1 but abolishes the regulation of divalent cation entry by extracellular Ca(2+). Orai1 and TRPC1 have been shown to play a major role in SOCE. Expression of the STIM1(D76A) mutant did not alter Orai1 phosphoserine content. TRPC1 silencing significantly attenuated TG-induced Mn(2+) entry. Expression of the STIM1(K684,685E) mutant impaired the association of plasma membrane STIM1 with TRPC1, as well as the regulation of TG-induced divalent cation entry by extracellular Ca(2+), which suggests that TRPC1 might be involved in the regulation of divalent cation entry by extracellular Ca(2+) mediated by plasma membrane-resident STIM1. Expression of the STIM1(D76A) or STIM1(K684,685E) mutants reduced store-operated divalent cation entry and resulted in loss of dependence on the extracellular Ca(2+) concentration, providing evidence for a functional role of plasma membrane-resident STIM1 in the regulation of store-operated divalent cation entry, which at least involves the EF-hand motif and the C-terminal polybasic lysine-rich domain.
Collapse
Affiliation(s)
- Isaac Jardin
- Institute of Biophysics, University of Linz, A-4040 Linz, Austria
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Lysophosphatidic acid promotes cell migration through STIM1- and Orai1-mediated Ca2+(i) mobilization and NFAT2 activation. J Invest Dermatol 2012; 133:793-802. [PMID: 23096711 PMCID: PMC3572452 DOI: 10.1038/jid.2012.370] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Lysophosphatidic acid (LPA) enhances cell migration and promotes wound healing in vivo, but the intracellular signaling pathways regulating these processes remain incompletely understood. Here we investigated the involvement of agonist-induced Ca2+ entry and STIM1 and Orai1 proteins in regulating nuclear factor of activated T cell (NFAT) signaling and LPA-induced keratinocyte cell motility. As monitored by Fluo-4 imaging, stimulation with 10 μℳ LPA in 60 μℳ Ca2+o evoked Ca2+i transients owing to store release, whereas addition of LPA in physiological 1.2 mℳ Ca2+o triggered store release coupled to extracellular Ca2+ entry. Store-operated Ca2+ entry (SOCE) was blocked by the SOCE inhibitor diethylstilbestrol (DES), STIM1 silencing using RNA interference (RNAi), and expression of dominant/negative Orai1R91W. LPA induced significant NFAT activation as monitored by nuclear translocation of green fluorescent protein-tagged NFAT2 and a luciferase reporter assay, which was impaired by DES, expression of Orai1R91W, and inhibition of calcineurin using cyclosporin A (CsA). By using chemotactic migration assays, LPA-induced cell motility was significantly impaired by STIM1, CsA, and NFAT2 knockdown using RNAi. These data indicate that in conditions relevant to epidermal wound healing, LPA induces SOCE and NFAT activation through Orai1 channels and promotes cell migration through a calcineurin/NFAT2-dependent pathway.
Collapse
|
11
|
Dionisio N, Galán C, Jardín I, Salido GM, Rosado JA. Lipid rafts are essential for the regulation of SOCE by plasma membrane resident STIM1 in human platelets. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2011; 1813:431-7. [PMID: 21255618 DOI: 10.1016/j.bbamcr.2011.01.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Revised: 12/26/2010] [Accepted: 01/10/2011] [Indexed: 01/23/2023]
Abstract
STIM1 is a transmembrane protein essential for the activation of store-operated Ca²+ entry (SOCE), a major Ca²+ influx mechanism. STIM1 is either located in the endoplasmic reticulum, communicating the Ca²+ concentration in the stores to plasma membrane channels or in the plasma membrane, where it might sense the extracellular Ca²+ concentration. Plasma membrane-located STIM1 has been reported to mediate the SOCE sensitivity to extracellular Ca²+ through its interaction with Orai1. Here we show that plasma membrane lipid raft domains are essential for the regulation of SOCE by extracellular Ca²+. Treatment of platelets with the SERCA inhibitor thapsigargin (TG) induced Mn²+ entry, which was inhibited by increasing concentrations of extracellular Ca²+. Platelet treatment with methyl-β-cyclodextrin, which removes cholesterol and disrupts the lipid raft domains, impaired the inactivation of Ca²+ entry induced by extracellular Ca²+. Methyl-β-cyclodextrin also abolished translocation of STIM1 to the plasma membrane stimulated by treatment with TG and prevented TG-evoked co-immunoprecipitation between plasma membrane-located STIM1 and the Ca²+ permeable channel Orai1. These findings suggest that lipid raft domains are essential for the inactivation of SOCE by extracellular Ca²+ mediated by the interaction between plasma membrane-located STIM1 and Orai1.
Collapse
Affiliation(s)
- Natalia Dionisio
- Department of Physiology, Cell Physiology Research Group, University of Extremadura, Cáceres, Spain
| | | | | | | | | |
Collapse
|
12
|
Harper MT, Molkentin JD, Poole AW. Protein kinase C alpha enhances sodium-calcium exchange during store-operated calcium entry in mouse platelets. Cell Calcium 2010; 48:333-40. [PMID: 21094527 DOI: 10.1016/j.ceca.2010.10.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2010] [Revised: 10/15/2010] [Accepted: 10/18/2010] [Indexed: 11/20/2022]
Abstract
A rise in intracellular calcium concentration ([Ca(2+)](i)) is necessary for platelet activation. A major component of the [Ca(2+)](i) elevation occurs through store-operated Ca(2+) entry (SOCE). The aim of this study was to understand the contribution of the classical PKC isoform, PKCα to platelet SOCE, using platelets from PKCα-deficient mice. SOCE was reduced by approximately 50% in PKCα(-/-) platelets, or following treatment with bisindolylmaleimide I, a PKC inhibitor. However, TG-induced Mn(2+) entry was unaffected, which suggests that divalent cation entry through store-operated channels is not directly regulated. Blocking the autocrine action of secreted ADP or 5-HT on its receptors did not reproduce the effect of PKCα deficiency. In contrast, SN-6, a Na(+)/Ca(2+) exchanger inhibitor, did reduce SOCE to the same extent as loss of PKCα, as did replacing extracellular Na(+) with NMDG(+). These treatments had no further effect in PKCα(-/-) platelets. These data suggest that PKCα enhances the extent of SOCE in mouse platelets by regulating Ca(2+) entry through the Na(+)/Ca(2+) exchanger.
Collapse
Affiliation(s)
- Matthew T Harper
- School of Physiology and Pharmacology, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, United Kingdom
| | | | | |
Collapse
|
13
|
Onopiuk M, Wierzbicka K, Brutkowski W, Szczepanowska J, Zabłocki K. Caspase-dependent inhibition of store-operated Ca(2+) entry into apoptosis-committed Jurkat cells. Biochem Biophys Res Commun 2010; 399:198-202. [PMID: 20643097 DOI: 10.1016/j.bbrc.2010.07.054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 07/15/2010] [Indexed: 11/15/2022]
Abstract
Activation of T-cells triggers store-operated Ca(2+) entry, which begins a signaling cascade leading to induction of appropriate gene expression and eventually lymphocyte proliferation and differentiation. The simultaneous enhancement of Fas ligand gene expression in activated cells allows the immune response to be limited by committing the activated cells to apoptosis. In apoptotic cells the store-operated calcium entry is significantly inhibited. It has been documented that moderate activation of Fas receptor may cause reversible inhibition of store-operated channels by ceramide released from hydrolyzed sphingomyelin. Here we show that activation of Fas receptor in T-cells results in caspase-dependent decrease of cellular STIM1 and Orai1 protein content. This effect may be responsible for the substantial inhibition of Ca(2+) entry into Jurkat cells undergoing apoptosis. In turn, this inhibition might prevent overloading of cells with calcium and protect them against necrosis.
Collapse
Affiliation(s)
- Marta Onopiuk
- Department of Biochemistry, The Nencki Institute of Experimental Biology, Warsaw, Poland
| | | | | | | | | |
Collapse
|
14
|
Harper MT, Poole AW. Protein kinase Ctheta negatively regulates store-independent Ca2+ entry and phosphatidylserine exposure downstream of glycoprotein VI in platelets. J Biol Chem 2010; 285:19865-73. [PMID: 20388711 PMCID: PMC2888397 DOI: 10.1074/jbc.m109.085654] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 03/23/2010] [Indexed: 01/17/2023] Open
Abstract
Platelet activation must be tightly controlled to provide an effective, but not excessive, response to vascular injury. Cytosolic calcium is a critical regulator of platelet function, including granule secretion, integrin activation, and phosphatidylserine (PS) exposure. Here we report that the novel protein kinase C isoform, PKCtheta, plays an important role in negatively regulating Ca(2+) signaling downstream of the major collagen receptor, glycoprotein VI (GPVI). This limits PS exposure and so may prevent excessive platelet procoagulant activity. Stimulation of GPVI resulted in significantly higher and more sustained Ca(2+) signals in PKCtheta(-/-) platelets. PKCtheta acts at multiple distinct sites. PKCtheta limits secretion, reducing autocrine ADP signaling that enhances Ca(2+) release from intracellular Ca(2+) stores. PKCtheta thereby indirectly regulates activation of store-operated Ca(2+) entry. However, PKCtheta also directly and negatively regulates store-independent Ca(2+) entry. This pathway, activated by the diacylglycerol analogue, 1-oleoyl-2-acetyl-sn-glycerol, was enhanced in PKCtheta(-/-) platelets, independently of ADP secretion. Moreover, LOE-908, which blocks 1-oleoyl-2-acetyl-sn-glycerol-induced Ca(2+) entry but not store-operated Ca(2+) entry, blocked the enhanced GPVI-dependent Ca(2+) signaling and PS exposure seen in PKCtheta(-/-) platelets. We propose that PKCtheta normally acts to restrict store-independent Ca(2+) entry during GPVI signaling, which results in reduced PS exposure, limiting platelet procoagulant activity during thrombus formation.
Collapse
Affiliation(s)
- Matthew T. Harper
- From the Department of Physiology and Pharmacology, School of Medical Sciences, University of Bristol, BS8 1TD Bristol, United Kingdom
| | - Alastair W. Poole
- From the Department of Physiology and Pharmacology, School of Medical Sciences, University of Bristol, BS8 1TD Bristol, United Kingdom
| |
Collapse
|
15
|
Ca2+ entry through a non-selective cation channel in Aplysia bag cell neurons. Neuroscience 2009; 162:1023-38. [DOI: 10.1016/j.neuroscience.2009.05.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2009] [Revised: 05/03/2009] [Accepted: 05/05/2009] [Indexed: 11/20/2022]
|
16
|
Jardín I, López JJ, Redondo PC, Salido GM, Rosado JA. Store-operated Ca2+ entry is sensitive to the extracellular Ca2+ concentration through plasma membrane STIM1. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1614-22. [PMID: 19631699 DOI: 10.1016/j.bbamcr.2009.07.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 07/03/2009] [Accepted: 07/13/2009] [Indexed: 01/06/2023]
Abstract
Store-operated Ca(2+) entry (SOCE) is a major mechanism for Ca(2+) influx in platelets and other cells activated by a reduction in Ca(2+) concentration in the intracellular stores. SOCE has been reported to be regulated by extracellular Ca(2+), although the underlying mechanism remains unclear. Here we have examined the involvement of plasma membrane-located STIM1 (PM-STIM1) in the regulation of SOCE by extracellular Ca(2+). Treatment of platelets with the SERCA inhibitor thapsigargin (TG) induced Mn(2+) entry, which was inhibited by extracellular Ca(2+) in a concentration-dependent manner. Incubation of platelets with a specific antibody, which recognizes the extracellular amino acid sequence 25-139 of PM-STIM1 that contains the Ca(2+)-binding domain, prevented the inactivation of Ca(2+) entry induced by extracellular Ca(2+). TG induced translocation of STIM1 to the plasma membrane (PM), an event that was found to be Ca(2+)-dependent. In addition, TG stimulated association of PM-STIM1 with Orai1, an event that was not prevented by stabilization of the membrane cytoskeleton using jasplakinolide. These findings suggest that PM-STIM1 is important for the inactivation of SOCE by extracellular Ca(2+), an event that is likely to be mediated by interaction with Orai1.
Collapse
Affiliation(s)
- Isaac Jardín
- Department of Physiology (Cellular Physiology Research Group), University of Extremadura, 10071 Cáceres, Spain
| | | | | | | | | |
Collapse
|
17
|
Kerrigan MJP, Hall AC. Control of chondrocyte regulatory volume decrease (RVD) by [Ca2+]i and cell shape. Osteoarthritis Cartilage 2008; 16:312-22. [PMID: 17855127 DOI: 10.1016/j.joca.2007.07.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2007] [Accepted: 07/16/2007] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Optimal matrix metabolism by articular chondrocytes is controlled by the 'set-point' volume which is determined mainly by membrane transporters. The signal transduction pathway(s) for the key membrane transporter which responds to cell swelling ('osmolyte channel') and mediates regulatory volume decrease (RVD) is poorly understood, so here the role of Ca2+ and the effects of 2D culture have been clarified. METHODS Changes to the volume and intracellular calcium levels ([Ca2+]i) of freshly isolated and 2D cultured bovine articular chondrocytes subjected to hypotonic challenge using a 43% reduction in medium osmolarity were studied by single-cell fluorescence microscopy. The effects of ethylene glycol tetraacetic acid (EGTA), REV5901 and Gd(3+) were studied and the role of Ca2+ influx determined by Mn2+ quench. RESULTS In freshly isolated cells, approximately 50% of chondrocytes exhibited 'robust RVD' (6[120]). RVD was inhibited by REV 5901 (4+/-2% responding) (3[23]) and 2 mM EGTA (18+/-5% responding) (4[166]) whereas Gd3+ had no effect (3[89]). The hypotonic challenge resulted in a Gd3+-insensitive rise in [Ca2+]i that did not correlate with RVD in all cells. Following 2D culture, chondrocytes also demonstrated Gd3+-insensitive RVD, but in contrast, the [Ca2+]i rise was blocked by this agent. CONCLUSIONS The data suggested that in freshly isolated and 2D cultured chondrocytes, the rise in [Ca2+]i occurring during hypotonic challenge could be related to RVD, but only in some cells. However, with 2D culture, the Ca2+ response switched to being Gd3+-sensitive, suggesting that as a result of changes to chondrocyte shape, stretch-activated cation channels although present, do not appear to play a role in volume regulation.
Collapse
Affiliation(s)
- M J P Kerrigan
- School of Biosciences, Department of Human and Health Sciences, University of Westminster, London, UK.
| | | |
Collapse
|
18
|
McElroy SP, Gurney AM, Drummond RM. Pharmacological profile of store-operated Ca(2+) entry in intrapulmonary artery smooth muscle cells. Eur J Pharmacol 2008; 584:10-20. [PMID: 18308301 DOI: 10.1016/j.ejphar.2008.01.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2007] [Revised: 12/19/2007] [Accepted: 01/15/2008] [Indexed: 10/22/2022]
Abstract
Store-operated Ca(2+) entry (SOCE) plays an important role in the contraction and proliferation of pulmonary artery smooth muscle cells (PASMCs). The aim of this study was to characterise the pharmacological properties of the SOCE pathway in freshly isolated PASMCs from rat lung and to determine whether this Ca(2+) entry pathway is sensitive to nitric oxide donor drugs. Following depletion of Ca(2+) from the sarcoplasmic reticulum, by treating cells with thapsigargin, re-addition of Ca(2+) produced an increase in cytosolic fluo-4 fluorescence that was sustained for the period that extracellular Ca(2+) was present. Thapsigargin also increased the rate of quench of fura-2 fluorescence, confirming that SOCE was activated. The SOCE pathway was not affected by nifedipine or verapamil; however, it was inhibited by the divalent cations Ni(2+) (10 microM) and Cd(2+) (10 microM) by 47+/-5% and 49+/-5% respectively. SOCE was also inhibited 42+/-5% by 2-aminoethoxydiphenyl borate (2-APB; 75 microM) and 58+/-4% by Gd(3+) (10 microM), although La(3+) (100 microM) had little effect. None of the NO donors examined, including sodium nitroprusside, glyceryl trinitrate, and 2-(N,N-diethylamino)-diazenolate-2-oxide had any effect on SOCE. Thus, the pulmonary vasorelaxation produced by NO does not involve direct inhibition of SOCE in PASMCs. Western blot and immunocytochemistry using antibodies directed against specific TRPC subunits detected the presence of TRPC1, 3, and 6 in pulmonary artery and the pharmacological profile of SOCE in PASMCs favours a role for TRPC1 in mediating the underlying channels that are activated by store depletion.
Collapse
Affiliation(s)
- Stuart P McElroy
- Division of Physiology and Pharmacology, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0NR, United Kingdom
| | | | | |
Collapse
|
19
|
Jardín I, Redondo PC, Salido GM, Rosado JA. Phosphatidylinositol 4,5-bisphosphate enhances store-operated calcium entry through hTRPC6 channel in human platelets. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2007; 1783:84-97. [PMID: 17719101 DOI: 10.1016/j.bbamcr.2007.07.007] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2007] [Revised: 06/29/2007] [Accepted: 07/19/2007] [Indexed: 10/23/2022]
Abstract
Phosphatidylinositol 4,5-bisphosphate (PIP2) is a versatile regulator of TRP channels. We report that inclusion of a PIP2 analogue, PIP2 1,2-dioctanoyl, does not induce non-capacitative Ca2+ entry per se but enhanced Ca2+ entry stimulated either by thrombin or by selective depletion of the Ca2+ stores in platelets, the dense tubular system, using 10 nM TG, and the acidic stores, using 20 microM 2,5-di-(tert-butyl)-1,4-hydroquinone (TBHQ). Reduction of PIP2 levels by blocking PIP2 resynthesis with Li+ or introducing a monoclonal anti-PIP2 antibody, or sequestering PIP2 using poly-lysine, attenuated Ca2+ entry induced by thrombin, TG and TBHQ, and reduced thrombin-evoked, but not TG- or TBHQ-induced, Ca2+ release from the stores. Incubation with the anti-hTRPC1 antibody did not alter the stimulation of Ca2+ entry by PIP2, whilst introduction of anti-hTRPC6 antibody directed towards the C-terminus of hTRPC6 reduced Ca2+ and Mn2+ entry induced by thrombin, TG or TBHQ, and abolished the stimulation of Ca2+ entry by PIP2. The anti-hTRPC6 antibody, but not the anti-hTRPC1 antibody or PIP2, reduced non-capacitative Ca2+ entry by the DAG analogue 1-oleoyl-2-acetyl-sn-glycerol. In summary, hTRPC6 plays a role both in store-operated and in non-capacitative Ca2+ entry. PIP2 enhances store-operated Ca2+ entry in human platelets, most probably by stimulation of hTRPC6 channels.
Collapse
Affiliation(s)
- Isaac Jardín
- Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10071 Cáceres, Spain
| | | | | | | |
Collapse
|
20
|
Liao JY, Li LL, Wei Q, Yue JC. Heregulinβ activates store-operated Ca2+ channels through c-erbB2 receptor level-dependent pathway in human breast cancer cells. Arch Biochem Biophys 2007; 458:244-52. [PMID: 17214955 DOI: 10.1016/j.abb.2006.12.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Revised: 11/23/2006] [Accepted: 12/03/2006] [Indexed: 11/21/2022]
Abstract
The heregulinbeta (HRGbeta) is a ligand to activate c-erbB2/c-erbB3 interaction and can subsequently increases cytosolic [Ca(2+)](i). In the two human breast cancer cell lines, MCF-7 shows a low c-erbB2 expression level, whereas SK-BR-3 overexpress c-erbB2 receptor. In this article, we have found that in MCF-7, HRGbeta induced Ca(2+) release from the endoplasmic reticulums (ER) and subsequently activated Ca(2+) entry via store-operated Ca(2+) channel (SOC). However, in SK-BR-3, HRGbeta failed to induce Ca(2+) release and Ca(2+)entry. RNA interference to decrease c-erbB2 level in SK-BR-3 resulted in reactivation of HRGbeta-evoked Ca(2+) release and Ca(2+) entry via SOC, which was similar to that of MCF-7. In addition, in the absence of HRGbeta, a constitutive activation of SOC was observed in SK-BR-3 rather than in MCF-7 and c-erbB2-siRNA treated SK-BR-3. Compared to the cells with low c-erbB2 level, c-erbB2 might tend to interact with c-erbB3 in the resting state in the cells with high c-erbB2 level, which resulted in different [Ca(2+)](i) responses to HRGbeta. In SK-BR-3, the Ca(2+) mobilization in the presence or in the absence of HRGbeta was completely blocked by PLC inhibitor U73122. In summary, our results indicate that HRGbeta-induced SOC was regulated by c-erbB2 level and dependent on activation of PLC in human breast cancer cells.
Collapse
Affiliation(s)
- Jie-Ying Liao
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, Chaoyang District, Beijing 100101, China
| | | | | | | |
Collapse
|
21
|
Atucha NM, Iyú D, Alcaraz A, Rosa V, Martínez-Prieto C, Ortiz MC, Rosado JA, García-Estañ J. Altered calcium signalling in platelets from bile-duct-ligated rats. Clin Sci (Lond) 2007; 112:167-74. [PMID: 16948638 DOI: 10.1042/cs20060226] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In the present study, we have analysed the mechanisms of Ca2+ entry and release in platelets obtained from BDL (bile-duct-ligated) rats, 11–13 days and 4 weeks after surgery. Platelets were washed and loaded with fura-2, and [Ca2+]i (cytosolic Ca2+ concentration) was determined in cell suspensions by means of fluorescence spectroscopy. Basal [Ca2+]i was similar in platelets from BDL rats compared with those from their respective controls, both in the absence and presence of extracellular Ca2+. Platelet stimulation with thrombin in the absence and presence of extracellular Ca2+ induced a rapid rise in [Ca2+]i that was of greater magnitude in platelets from BDL rats than in controls. Ca2+ storage was significantly elevated in platelets from BDL rats, as well as the activity of SERCA (sarcoplasmic/endoplasmic-reticulum Ca2+-ATPase). Capacitative Ca2+ entry, as evaluated by inhibition of SERCA with thapsigargin, was also altered in platelets from BDL rats, having lower rates of Ca2+ entry. In conclusion, chronic BDL alters intracellular Ca2+ homoeostasis in platelets, such that an enhanced Ca2+ release is evoked by thrombin, which may be due to an increased amount of Ca2+ stored in the intracellular organelles and secondary to an enhanced activity of SERCA. These alterations are already evident before cirrhosis has completely developed and occurs during the cholestasis phase.
Collapse
Affiliation(s)
- Noemí M Atucha
- Department of Physiology, Faculty of Medicine, University of Murcia, 30100 Murcia, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Redondo PC, Harper MT, Rosado JA, Sage SO. A role for cofilin in the activation of store-operated calcium entry by de novo conformational coupling in human platelets. Blood 2006; 107:973-9. [PMID: 16234361 DOI: 10.1182/blood-2005-05-2015] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractStore-operated Ca2+ entry (SOCE) is a major mechanism for Ca2+ influx in platelets and other cells. De novo conformational coupling between elements in the plasma membrane and Ca2+ stores, where the actin cytoskeleton plays an important regulatory role, has been proposed as the most likely mechanism to activate SOCE in platelets. Here we have examined for the first time changes in platelet F-actin levels on a subsecond time scale. Using stopped-flow fluorimetry and a quenched-flow approach, we provide evidence for the involvement of cofilin in actin filament reorganization and SOCE in platelets. Thrombin (0.1 U/mL) evoked an initial decrease in F-actin that commenced within 0.1 second and reached a minimum 0.9 second after stimulation, prior to the activation of SOCE. F-actin then increased, exceeding basal levels approximately 2.5 seconds after stimulation. Thrombin also induced cofilin dephosphorylation and activation, which paralleled the changes observed in F-actin, and rapid Btk activation. Inhibition of cofilin dephosphorylation by LFM-A13 resulted in the loss of net actin depolymerization and an increased delay in SOCE initiation. These results suggest that cofilin is important for the rapid actin remodeling necessary for the activation of SOCE in platelets through de novo conformational coupling.
Collapse
Affiliation(s)
- Pedro C Redondo
- Department of Physiology, University of Extremadura, Cáceres, Spain
| | | | | | | |
Collapse
|
23
|
Labiós M, Martínez M, Gabriel F, Guiral V, Ruiz-Aja S, Aznar J. Cytoplasmic free calcium mobilization in platelets, expression of P-selectin, phosphatidylserine, and microparticle formation, measured by whole blood flow cytometry, in hypertensive patients. Effect of doxazosin GITS. Thromb Res 2006; 117:403-9. [PMID: 16461073 DOI: 10.1016/j.thromres.2005.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2004] [Revised: 03/23/2005] [Accepted: 03/29/2005] [Indexed: 10/25/2022]
Abstract
The effects of doxazosin on expression of CD62 (P-selectin) and phosphatidylserine on platelet membrane and platelet calcium flux were studied in 50 uncomplicated essential hypertensive patients (World Health Organization stages 1-2) and 80 normotensive control subjects, matched for age, sex, and cardiovascular risk factors. Hypertensive patients showed greater in vivo platelet activation at baseline than control patients (percentage of CD62-positive platelets, 4.1+/-2.2% versus 2.4+/-1.5%, p<0.001; percentage of phosphatidylserine-positive platelets, 0.8+/-0.5% versus 0.5+/-0.3%, p<0.001). Increased platelet activation was associated with significant changes in the mobilization of free intraplatelet calcium, evaluated by a whole blood flow cytometric kinetic method. With this method, an arbitrary Ca(2+) mobilization index was defined as the ratio of cytoplasmic free calcium before activation with thrombin to the slope of the calcium removal rate following the action of the agonist. This index was significantly higher in untreated hypertensive patients than in normotensive controls (0.12+/-0.06 versus 0.05+/-0.08, p<0.001). Treatment of hypertensive patients with doxazosin gastrointestinal therapeutic system (4 mg/day as a single dose) for 2 months normalized both platelet activation and Ca(2+) mobilization. Changes in the expression of CD62 and phosphatidylserine in the platelet membrane after treatment with doxazosin gastrointestinal therapeutic system may be related to normalization of the kinetics of cytoplasmic free Ca(2+). Normalization of platelet activation may represent an additional beneficial effect to the known antihypertensive action of doxazosin gastrointestinal therapeutic system.
Collapse
Affiliation(s)
- Manuel Labiós
- Hypertension Unit, Clinic University Hospital, Valencia, Spain
| | | | | | | | | | | |
Collapse
|
24
|
Ben-Amor N, Redondo PC, Bartegi A, Pariente JA, Salido GM, Rosado JA. A role for 5,6-epoxyeicosatrienoic acid in calcium entry by de novo conformational coupling in human platelets. J Physiol 2005; 570:309-23. [PMID: 16308346 PMCID: PMC1464301 DOI: 10.1113/jphysiol.2005.100800] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
A major pathway for Ca(2+) entry in non-excitable cells is activated following depletion of intracellular Ca(2+) stores. A de novo conformational coupling between elements in the plasma membrane (PM) and Ca(2+) stores has been proposed as the most likely mechanism to activate this capacitative Ca(2+) entry (CCE) in several cell types, including platelets. Here we report that a cytochrome P450 metabolite, 5,6-EET, might be a component of the de novo conformational coupling in human platelets. In these cells, 5,6-EET induces divalent cation entry without having any detectable effect on Ca(2+) store depletion. 5,6-EET-induced Ca(2+) entry was sensitive to the CCE blockers 2-APB, lanthanum, SKF-96365 and nickel and impaired by incubation with anti-hTRPC1 antibody. Ca(2+) entry stimulated by low concentrations of thapsigargin, which selectively depletes the dense tubular system and induces EET production, was impaired by the cytochrome P450 inhibitor 17-ODYA, which has no effect on CCE mediated by depletion of the acidic stores using 2,5-di-(tert-butyl)-1,4-hydroquinone. We have found that 5,6-EET-induced Ca(2+) entry requires basal levels of H(2)O(2), which might maintain a redox state favourable for this event. Finally, our results indicate that 5,6-EET induces the activation of tyrosine kinase proteins and the reorganization of the actin cytoskeleton, which might provide a support for the transport of portions of the Ca(2+) store towards the PM to facilitate de novo coupling between IP(3)R type II and hTRPC1 detected by coimmunoprecipitation. We propose that the involvement of 5,6-EET in TG-induced coupling between IP(3)R type II and hTRPC1 and subsequently CCE is compatible with the de novo conformational coupling in human platelets.
Collapse
Affiliation(s)
- Nidhal Ben-Amor
- Unité de Recherche de Biochimie, Institute Superieur de Biotechnologie, Monastir, Tunisia
| | | | | | | | | | | |
Collapse
|
25
|
Tran L, Farinas J, Ruslim-Litrus L, Conley PB, Muir C, Munnelly K, Sedlock DM, Cherbavaz DB. Agonist-induced calcium response in single human platelets assayed in a microfluidic device. Anal Biochem 2005; 341:361-8. [PMID: 15907883 DOI: 10.1016/j.ab.2005.02.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2005] [Indexed: 10/25/2022]
Abstract
To facilitate drug discovery directed toward platelet-specific targets, we developed a platelet isolation and fluorophore-loading method that yields functionally responsive platelets in which we were able to detect agonist-induced calcium flux using a microfluidics-based screening platform. The platelet preparation protocol was designed to minimize preparation-induced platelet activation and to optimize signal strength. Measurement of platelet activation, as monitored by ratiometric determination of agonist-induced calcium flux in fluor-loaded human platelets, was optimized in a macrosample cuvette format in preparation for detection in a microfluidic chip-based assay. For the microfluidic device used in these studies, a cell density of 1 to 2 x 10(6) platelets per milliliter and a nominal flow rate of 5 to 10 nl per second provided optimal event resolution of 5 to 20 platelets traversing the detection volume per unit time. Platelets responded in a dose-dependent manner to adenosine diphosphate and protease-activating peptide (PAR) 1 thrombin receptor-activating peptide (TRAP). The work presented here constitutes proof-of-principle experiments demonstrating the enabling application of a microfluidic device to conduct high-throughput signaling studies and drug discovery screening against human platelet targets.
Collapse
Affiliation(s)
- Louie Tran
- Caliper Technologies, Mountain View, CA 94043, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Asplund Persson AK, Palmér L, Gunnarsson P, Grenegård M. Characterisation of GEA 3175 on human platelets; comparison with S-nitroso-N-acetyl-D,L-penicillamine. Eur J Pharmacol 2005; 496:1-9. [PMID: 15288569 DOI: 10.1016/j.ejphar.2004.06.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2004] [Accepted: 06/02/2004] [Indexed: 10/26/2022]
Abstract
By comparing the effect of two nitric oxide (NO)-containing compounds, we found that S-nitroso-N-acetyl-D,L-penicillamine (SNAP), but not GEA 3175 (1,2,3,4-Oxatriazolium,3-(3-chloro-2-metylphenyl)-5-[[(4-methylphenyl)sulfonyl]amino]-, hydroxide inner salt), released NO. Despite this, both drugs elevated cyclic guanosine 3',5'-monophosphate (cGMP) levels in human platelets. However, SNAP was more effective after short exposure times (5 and 20 s). The compounds also inhibited thrombin-induced rises in cytosolic Ca2+. Time studies revealed that the action of SNAP rapidly declined by increasing the length of incubation (from 5 s to 30 min). This desensibilisation phenomenon mainly involved the release of Ca2+ from intracellular stores. In comparison, GEA 3175-induced inhibition of cytosolic Ca2+ signalling was much more long-lasting. The soluble guanylyl cyclase (sGC) inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ) reversed the effect of GEA 3175 on cytosolic Ca2+. Consequently, this inhibition depends solely on the increase in cGMP. In summary, differences between GEA 3175 and SNAP were observed in NO releasing, cGMP elevating and Ca2+ suppressive properties.
Collapse
Affiliation(s)
- Anna K Asplund Persson
- Department of Medicine and Care, Division of Pharmacology, Faculty of Health Sciences, Linköping University, SE-581 85 Linköping, Sweden.
| | | | | | | |
Collapse
|
27
|
Chernysh O, Condrescu M, Reeves JP. Calcium-dependent regulation of calcium efflux by the cardiac sodium/calcium exchanger. Am J Physiol Cell Physiol 2004; 287:C797-806. [PMID: 15151903 DOI: 10.1152/ajpcell.00176.2004] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Allosteric regulation by cytosolic Ca2+of Na+/Ca2+exchange activity in the Ca2+efflux mode has received little attention because it has been technically difficult to distinguish between the roles of Ca2+as allosteric activator and transport substrate. In this study, we used transfected Chinese hamster ovary cells to compare the Ca2+efflux activities in nontransfected cells and in cells expressing either the wild-type exchanger or a mutant, Δ(241–680), that operates constitutively; i.e., its activity does not require allosteric Ca2+activation. Expression of the wild-type exchanger did not significantly lower the cytosolic Ca2+concentration ([Ca2+]i) compared with nontransfected cells. During Ca2+entry through store-operated Ca2+channels, Ca2+efflux by the wild-type exchanger became evident only after [Ca2+]iapproached 100–200 nM. A subsequent decline in [Ca2+]iwas observed, suggesting that the activation process was time dependent. In contrast, Ca2+efflux activity was evident under all experimental conditions in cells expressing the constitutive exchanger mutant. After transient exposure to elevated [Ca2+]i, the wild-type exchanger behaved similarly to the constitutive mutant for tens of seconds after [Ca2+]ihad returned to resting levels. We conclude that Ca2+efflux activity by the wild-type exchanger is allosterically activated by Ca2+, perhaps in a time-dependent manner, and that the activated state is briefly retained after the return of [Ca2+]ito resting levels.
Collapse
Affiliation(s)
- Olga Chernysh
- Dept. of Pharmacology and Physiology, Graduate School of Biomedical Sciences, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, 185 South Orange Avenue, Room H649, Newark, NJ 07103, USA
| | | | | |
Collapse
|
28
|
Nilsson UK, Andersson RGG, Ekeroth J, Hallin EC, Konradsson P, Lindberg J, Svensson SPS. Lack of stereospecificity in lysophosphatidic acid enantiomer-induced calcium mobilization in human erythroleukemia cells. Lipids 2004; 38:1057-64. [PMID: 14669971 DOI: 10.1007/s11745-006-1161-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Lysophosphatidic acid (LPA) is a lipid mediator that, among several other cellular responses, can stimulate cells to mobilize calcium (Ca2+). LPA is known to activate at least three different subtypes of G protein-coupled receptors. These receptors can then stimulate different kinds of G proteins. In the present study, LPA and LPA analogs were synthesized from (R)- and (S)-glycidol and used to characterize the ability to stimulate Ca2+ mobilization. The cytosolic Ca2+ concentration ([Ca2+]i) was measured in fura-2-acetoxymethylester-loaded human erythroleukemia (HEL) cells. Furthermore, a reverse transcriptase polymerase chain reaction was used to characterize LPA receptor subtypes expressed in HEL cells. The results show that HEL cells mainly express LPA1 and LPA2, although LPA3 might possibly be expressed as well. Moreover, LPA and its analogs concentration-dependently increased [Ca2+]i in HEL cells. The response involved both influx of extracellular Ca2+ and release of Ca2+ from intracellular stores. This is the first time the unnatural (S)-enantiomer of LPA, (S)-3-O-oleoyl-1-O-phosphoryl-glycerol, has been synthesized and studied according to its ability to activate cells. The results indicate that this group of receptors does not discriminate between (R)- and (S)-enantiomers of LPA and its analogs. When comparing ether analogs having different hydrocarbon chain lengths, the tetradecyl analog (14 carbons) was found to be the most effective in increasing [Ca2+]i. Pertussis toxin treatment of the HEL cells resulted in an even more efficient Ca2+ mobilization stimulated by LPA and its analogs. Furthermore, at repeated incubation with the same ligand no further increase in [Ca2+]i was obtained. When combining LPA with the ether analogs no suppression of the new Ca2+ signal occurred. All these findings may be of significance in the process of searching for specific agonists and antagonists of the LPA receptor subtypes.
Collapse
Affiliation(s)
- Ulrika K Nilsson
- Division of Pharmacology, Department of Medicine and Care, Faculty of Health Sciences, Linköpings Universitet, SE-581 85 Linköping, Sweden.
| | | | | | | | | | | | | |
Collapse
|
29
|
Yao J, Li Q, Chen J, Muallem S. Subpopulation of store-operated Ca2+ channels regulate Ca2+-induced Ca2+ release in non-excitable cells. J Biol Chem 2004; 279:21511-9. [PMID: 15016819 DOI: 10.1074/jbc.m314028200] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Ca2+-induced Ca2+ release (CICR) is a well characterized activity in skeletal and cardiac muscles mediated by the ryanodine receptors. The present study demonstrates CICR in the non-excitable parotid acinar cells, which resembles the mechanism described in cardiac myocytes. Partial depletion of internal Ca2+ stores leads to a minimal activation of Ca2+ influx. Ca2+ influx through this pathway results in an explosive mobilization of Ca2+ from the majority of the stores by CICR. Thus, stimulation of parotid acinar cells in Ca2+ -free medium with 0.5 microm carbachol releases approximately 5% of the Ca2+ mobilizable by 1 mm carbachol. Addition of external Ca2+ induced the same Ca2+ release observed in maximally stimulated cells. Similar results were obtained by a short treatment with 2.5-10 microm cyclopiazonic acid, an inhibitor of the sarco/endoplasmic reticulum Ca2+ ATPase pump. The Ca2+ release induced by the addition of external Ca2+ was largely independent of IP(3)Rs because it was reduced by only approximately 30% by the inhibition of the inositol 1,4,5-trisphosphate receptors with caffeine or heparin. Measurements of Ca2+ -activated outward current and [Ca2+](i) suggested that most CICR triggered by Ca2+ influx occurred away from the plasma membrane. Measurement of the response to several concentrations of cyclopiazonic acid revealed that Ca2+ influx that regulates CICR is associated with a selective portion of the internal Ca2+ pool. The minimal activation of Ca2+ influx by partial store depletion was confirmed by the measurement of Mn2+ influx. Inhibition of Ca2+ influx with SKF96365 or 2-aminoethoxydiphenyl borate prevented activation of CICR observed on addition of external Ca2+. These findings provide evidence for activation of CICR by Ca2+ influx in non-excitable cells, demonstrate a previously unrecognized role for Ca2+ influx in triggering CICR, and indicate that CICR in non-excitable cells resembles CICR in cardiac myocytes with the exception that in cardiac cells Ca2+ influx is mediated by voltage-regulated Ca2+ channels whereas in non-excitable cells Ca2+ influx is mediated by store-operated channels.
Collapse
Affiliation(s)
- Jian Yao
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9040, USA
| | | | | | | |
Collapse
|
30
|
Brownlow SL, Sage SO. Rapid agonist-evoked coupling of type II Ins(1,4,5)P3 receptor with human transient receptor potential (hTRPC1) channels in human platelets. Biochem J 2003; 375:697-704. [PMID: 12908873 PMCID: PMC1223726 DOI: 10.1042/bj20030929] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2003] [Revised: 08/07/2003] [Accepted: 08/11/2003] [Indexed: 02/02/2023]
Abstract
Depletion of intracellular Ca2+ stores results in the activation of SMCE (store-mediated Ca2+ entry) in many cells. The mechanism of activation of SMCE is poorly understood. In human platelets, a secretion-like coupling model may be involved. This proposes that store depletion results in trafficking of portions of the endoplasmic reticulum to the plasma membrane, enabling coupling between proteins in the two membranes. In support of this, we have shown that, in human platelets, agonist-evoked Ca2+ store depletion results in de novo and reversible coupling of the Ins P3RII [type II inositol (1,4,5)trisphosphate receptor] with the putative Ca2+ entry channel hTRPC1 [human canonical transient receptor potential 1 (protein); Rosado, Brownlow and Sage (2002) J. Biol. Chem. 277, 42157-42163]. A crucial test of the hypothesis that this coupling activates SMCE is that it should occur rapidly enough to account for agonist-evoked Ca2+ entry. In the present study, we have used quenched- and stopped-flow approaches to determine the latencies of thrombin-evoked coupling of Ins P3RII with hTRPC1 and of thrombin-evoked bivalent cation entry using Mn2+ quenching of fura 2 fluorescence. Thrombin-evoked Mn2+ entry was detected with a latency of 0.81+/-0.07 s (S.E.M., n =7) or 1.36+/-0.09 s (S.E.M., n =7) at a concentration of 1.0 or 0.1 unit/ml respectively. Coupling between Ins P3RII and hTRPC1, assessed at 100 ms intervals, was first detected with a latency of 0.9 or 1.4 s after stimulation with thrombin at a concentration of 1.0 or 0.1 unit/ml respectively. These results support the hypothesis that de novo coupling of Ins P3RII with hTRPC1 could activate SMCE in human platelets.
Collapse
Affiliation(s)
- Sharon L Brownlow
- Department of Physiology, University of Cambridge, Downing Street, Cambridge CB2 3EG, UK
| | | |
Collapse
|
31
|
Bryant AE. Biology and pathogenesis of thrombosis and procoagulant activity in invasive infections caused by group A streptococci and Clostridium perfringens. Clin Microbiol Rev 2003; 16:451-62. [PMID: 12857777 PMCID: PMC164226 DOI: 10.1128/cmr.16.3.451-462.2003] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Group A streptococcal necrotizing fasciitis/myonecrosis and Clostridium perfringens gas gangrene are two of the most fulminant gram-positive infections in humans. Tissue destruction associated with these infections progresses rapidly to involve an entire extremity. Multiple-organ failure is common, and morbidity and mortality remain high. Systemic activation of coagulation and dysregulation of the anticoagulation pathways contribute to the pathogenesis of many diverse disease entities of infectious etiology, and it has been our hypothesis that microvascular thrombosis contributes to reduced tissue perfusion, hypoxia, and subsequent regional tissue necrosis and organ failure in these invasive gram-positive infections. This article reviews the coagulation, anticoagulation, and fibrinolytic systems from cellular players to cytokines to novel antithrombotic therapies and discusses the mechanisms contributing to occlusive microvascular thrombosis and tissue destruction in invasive group A streptococcal and C. perfringens infections. A thorough understanding of these mechanisms may suggest novel therapeutic targets for patients with these devastating infections.
Collapse
Affiliation(s)
- Amy E Bryant
- Infectious Diseases Section, Veterans Affairs Medical Center, Boise, and Department of Microbiology, University of Idaho, Moscow, Idaho.
| |
Collapse
|
32
|
Calderaro V, Boccellino M, Cirillo G, Quagliuolo L, Cirillo D, Giovane A. Cyclosporine A amplifies Ca2+ signaling pathway in LLC-PK1 cells through the inhibition of plasma membrane Ca2+ pump. J Am Soc Nephrol 2003; 14:1435-42. [PMID: 12761243 DOI: 10.1097/01.asn.0000065632.32856.4c] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Cyclosporine A (CsA), a neutral, highly hydrophobic cyclic peptide with 11 amino acids, is currently the most widely used immunosuppressive drug for preventing graft rejection and autoimmune diseases. Despite its efficacy, the use of CsA is limited by severe side effects, mainly nephrotoxicity and arterial hypertension. Single cell microfluorimetry was used to evaluate the role of CsA on Ca(2+) signaling pathway in intact cells of the porcine proximal tubule-like cell line LLC-PK1; the assay of the in vitro activity of the plasma membrane Ca(2+) pump (PMCA) was carried out through the preparation and isolation of membranes. The addition of CsA to incubation medium at doses ranging from 0.1 to 2 microM did not change the basal level of intracellular calcium ([Ca(2+)](i)), whereas it affected the [Ca(2+)](i) response to thapsigargin (TG), a powerful inhibitor of microsomal Ca(2+) pump. In control studies, 5 microM TG produced a biphasic response: [Ca(2+)](i) peaked with a 60-s lag, and it then declined to a plateau of elevated [Ca(2+)](i), which remains above basal. However, it became evident that CsA strengthened the Ca(2+) response to TG because the addition of 5 microM TG to cells exposed to 400 nM CsA did not affect the peak response to TG, but it markedly affected the subsequent sustained phase ([Ca(2+)](i) = 156 +/- 4.84 versus 130 +/- 3.28 nmol, mean +/- SEM, n = 6, P < 0.001). In membrane preparations, 200 nM CsA brought about, in the presence of 10 microM calmodulin (CaM), a significant decrease of plasma membrane Ca(2+) pump (PMCA) activity (46.96 +/- 0.26 versus 53.48 +/- 1.96 nmol x mg of protein(-1) x min(-1), n = 6, P < 0.02), a value similar to that obtained in the presence of equimolar amounts of cyclosporine H (CsH), a non-immunosuppressive analogue of CsA. These findings suggest that in this cell line CsA affects the Ca(2+) export pathway through the reduction of the PMCA activity with consequent amplification and strengthening of [Ca(2+)](i) response after exposure to agents that trigger intracellular Ca(2+) release. The increased cell sensitivity during Ca(2+) signaling events ensuing from the impairment of this "defense system" may be regarded as one of the basic mechanisms involved in the development of the side effects induced by CsA.
Collapse
Affiliation(s)
- Vincenzo Calderaro
- Institute of Internal Medicine and Nephrology, Second University of Naples, Naples, Italy.
| | | | | | | | | | | |
Collapse
|
33
|
Monteiro M, Gonçalves M, Sansonetty F, O'Connor J. Flow Cytometric Analysis of Calcium Mobilization in Whole‐Blood Platelets. ACTA ACUST UNITED AC 2003; Chapter 9:Unit 9.20. [DOI: 10.1002/0471142956.cy0920s24] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
| | | | - Filipe Sansonetty
- Escola de Ciências de Saúde, Universidade do Minho IPATIMUP Braga Portugal
| | | |
Collapse
|
34
|
Zhang BX, Ma X, Yeh CK, Lifschitz MD, Zhu MX, Katz MS. Epidermal growth factor-induced depletion of the intracellular Ca2+ store fails to activate capacitative Ca2+ entry in a human salivary cell line. J Biol Chem 2002; 277:48165-71. [PMID: 12368284 DOI: 10.1074/jbc.m208077200] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Epidermal growth factor (EGF) is a multifunctional factor known to influence proliferation and function of a variety of cells. The actions of EGF are mediated by EGF receptor tyrosine kinase pathways, including stimulation of phospholipase Cgamma and mobilization of intracellular Ca(2+) ([Ca(2+)](i)). Generally, agonist-mediated Ca(2+) mobilization involves both Ca(2+) release from internal stores and Ca(2+) influx activated by store depletion (i.e. capacitative or store-operated Ca(2+) influx). However, the role of capacitative Ca(2+) entry in EGF-mediated Ca(2+) mobilization is still largely unknown. In this study, we compared [Ca(2+)](i) signals elicited by EGF with those induced by agents (the muscarinic receptor agonist carbachol and thapsigargin (Tg)) known to activate capacitative Ca(2+) entry. Unlike carbachol and Tg, EGF (5 nm) elicited a transient [Ca(2+)](i) signal without a plateau phase in the presence of extracellular Ca(2+) and also failed to accelerate Mn(2+) entry. Repletion of extracellular Ca(2+) to cells stimulated with EGF in the absence of Ca(2+) elicited an increase in [Ca(2+)](i), indicating that EGF indeed stimulates Ca(2+) influx. However, the influx was activated at lower EGF concentrations than those required to stimulate Ca(2+) release. Interestingly, the phospholipase C inhibitor completely inhibited Ca(2+) release induced by both EGF and carbachol and also reduced Ca(2+) influx responsive to carbachol but had no effect on Ca(2+) influx induced by EGF. EGF-induced Ca(2+) influx was potentiated by low concentrations (<5 ng/ml) of oligomycin, a mitochondrial inhibitor that blocks capacitative Ca(2+) influx in other systems. Transient expression of the hTRPC3 protein enhanced Ca(2+) influx responsive to carbachol but did not increase EGF-activated Ca(2+) influx. Both EGF and carbachol depleted internal Ca(2+) stores. Our results demonstrate that EGF-induced Ca(2+) release from internal stores does not activate capacitative Ca(2+) influx. Rather, EGF stimulates Ca(2+) influx via a mechanism distinct from capacitative Ca(2+) influx induced by carbachol and Tg.
Collapse
Affiliation(s)
- Bin-Xian Zhang
- Medical Research Service, South Texas Veterans Health Care System, Audie L. Murphy Division, San Antonio 78229, USA
| | | | | | | | | | | |
Collapse
|
35
|
Rosado JA, Brownlow SL, Sage SO. Endogenously expressed Trp1 is involved in store-mediated Ca2+ entry by conformational coupling in human platelets. J Biol Chem 2002; 277:42157-63. [PMID: 12196544 DOI: 10.1074/jbc.m207320200] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Physical interaction between transient receptor potential (Trp) channels and inositol 1,4,5-trisphosphate receptors (IP(3)Rs) has been presented as a candidate mechanism for the activation of store-mediated Ca(2+) entry. The role of a human homologue of Drosophila transient receptor potential channel, hTrp1, in the conduction of store-mediated Ca(2+) entry was examined in human platelets. Incubation of platelets with a specific antibody, which recognizes the extracellular amino acid sequence 557-571 of hTrp1, inhibited both store depletion-induced Ca(2+) and Mn(2+) entry in a concentration-dependent manner. Stimulation of platelets with the physiological agonist thrombin activated coupling between the IP(3) receptor type II and endogenously expressed hTrp1. This event was reversed by refilling of the internal Ca(2+) stores but maintained after removal of the agonist if the stores were not allowed to refill. Inhibition of IP(3) recycling using Li(+) or inhibition of IP(3)Rs with xestospongin C or treatment with jasplakinolide, to stabilize the cortical actin filament network, abolished thrombin-induced coupling between hTrp1 and IP(3)R type II. Incubation with the anti-hTrp1 antibody inhibited thrombin-evoked Ca(2+) entry without affecting Ca(2+) release from intracellular stores. These results provide evidence for the involvement of hTrp1 in the activation of store-mediated Ca(2+) entry by coupling to IP(3)R type II in normal human cells.
Collapse
Affiliation(s)
- Juan A Rosado
- Department of Physiology, University of Cambridge, United Kingdom
| | | | | |
Collapse
|
36
|
Abstract
A heterogeneous distribution of ion channels on the cell surface is a prerequisite for several cellular functions. Thus, there has been considerable interest in methods allowing the mapping of ion channel distributions. Here we report on a novel ratiometric imaging technique appropriate to measure spatially resolved ion flux signals by using ion sensitive dyes. However, given that certain relevant cell properties like the surface to volume ratio may exhibit significant spatial heterogeneities, the local influx signal cannot be interpreted as a measure of the local open channel concentration or flux density. To overcome this problem, we suggest an internal normalization procedure, which, in analogy to, but clearly distinct from, well-established ratioing techniques, eliminates effects which would otherwise obscure the desired result. Ratioing is performed on flux signals from a given cell, triggered by two different, subsequent stimuli. If the two stimuli address different ion channels, the flux density distribution caused by two channel types can be determined relative to each other. In cases where one of the stimuli triggers a spatially homogeneous flux signal, ratioing yields an ion flux density map for a given channel type. Thus distribution patterns of ion channels active during a given stimulus may be derived.
Collapse
Affiliation(s)
- S Munck
- Bioimaging Zentrum der Ludwig-Maximilians-Universität München, Martinsried, Germany
| | | | | |
Collapse
|
37
|
Sun D, Steele JE. Regulation of intracellular calcium in dispersed fat body trophocytes of the cockroach, Periplaneta americana, by hypertrehalosemic hormone. JOURNAL OF INSECT PHYSIOLOGY 2001; 47:1399-1408. [PMID: 12770146 DOI: 10.1016/s0022-1910(01)00130-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Incubation of trophocytes from dissaggregated fat body of Periplaneta americana with either of the hypertrehalosemic hormones, HTH-I or HTH-II, leads to an increase in the cytosolic concentration of Ca(2+) from approximately 80 to approximately 310nM with a rise time of approximately 110s. The Ca(2+) concentration then declines to the resting level during the ensuing 5min. In the absence of extracellular Ca(2+) the increase in [Ca(2+)](i) due to HTH is limited to approximately 100nM. The calmodulin inhibitors calmidazolium and W-7 also limit to a similar degree the ability of HTH to increase [Ca(2+)](i). Phorbol 12-myristate 13-acetate, an activator of protein kinase C, was shown to block Ca(2+) entry through the plasma membrane. Additional evidence to support the view that HTH enhances Ca(2+) influx has been obtained by measuring the quenching of fura-2 fluorescence when Ca(2+) is replaced with Mn(2+).
Collapse
Affiliation(s)
- D Sun
- Department of Zoology, The University of Western Ontario, Ont., N6A 5B7, London, Canada
| | | |
Collapse
|
38
|
Potocnik SJ, Hill MA. Pharmacological evidence for capacitative Ca(2+) entry in cannulated and pressurized skeletal muscle arterioles. Br J Pharmacol 2001; 134:247-56. [PMID: 11564642 PMCID: PMC1572963 DOI: 10.1038/sj.bjp.0704270] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Arteriolar myogenic tone shows a marked dependency on extracellular Ca(2+). The contribution played by mechanisms such as intracellular Ca(2+) release and capacitative entry, however, are less certain. The present studies aimed to demonstrate functional evidence for involvement of such mechanisms in myogenic tone and reactivity. Single cremaster arterioles were denuded of endothelium, pressurized under no-flow conditions and loaded with fura 2-AM for measurement of changes in intracellular Ca(2+) [Ca(2+)](i). The cell permeable, putative, IP(3) receptor antagonist 2APB (2 aminoethoxydiphenyl borate) was used to determine the possible role of IP(3) receptor-mediated mechanisms in arteriolar myogenic tone and reactivity. Arterioles dilated in response to increasing concentrations of 2APB (1 - 300 microM) without a concomitant change in global [Ca(2+)](i). Also 2APB (50 microM) completely inhibited the myogenic constriction in response to a step change in luminal pressure (50 - 120 mmHg) with no apparent effect on pressure-mediated increases in [Ca(2+)](i). 2APB markedly attenuated the constrictor response and [Ca(2+)](i) increase stimulated by phenylephrine but not KCl. Capacitative Ca(2+) influx in arterioles was demonstrated either by re-addition of extracellular [Ca(2+)] following pre-treatment with 1 or 10 microM nifedipine in Ca(2+) free buffer or exposure of vessels to thapsigargin (1 microM) to induce store depletion. In both cases 2APB inhibited the increase in [Ca(2+)](i). Capacitative Ca(2+) entry showed an inverse relationship with intraluminal pressure over the range 10 - 120 mmHg. Consistent with an effect on a Ca(2+) entry pathway, 2APB had no effect on intracellular (caffeine releasable) Ca(2+) stores while decreasing the rate of Mn(2+) quench of fura 2 fluorescence. The results provide functional evidence for capacitative Ca(2+) entry in intact arteriolar smooth muscle. The effectiveness of 2APB in inhibiting both non-voltage gated Ca(2+) entry and responsiveness to an acute pressure step is consistent with the involvement of an axis involving IP(3)-mediated and or capacitative Ca(2+) entry mechanisms in myogenic reactivity. Given the lack of effect of 2APB on pressure-induced changes in global [Ca(2+)](i) it is suggested that such mechanisms participate on a localized level to couple the myogenic stimulus to contraction.
Collapse
Affiliation(s)
- Simon J Potocnik
- Microvascular Biology Group, School of Medical Sciences, Division of Biosciences, RMIT University, Plenty Road, Bundoora, Victoria 3083, Australia
| | - Michael A Hill
- Microvascular Biology Group, School of Medical Sciences, Division of Biosciences, RMIT University, Plenty Road, Bundoora, Victoria 3083, Australia
- Author for correspondence:
| |
Collapse
|
39
|
Hill MA, Zou H, Potocnik SJ, Meininger GA, Davis MJ. Invited review: arteriolar smooth muscle mechanotransduction: Ca(2+) signaling pathways underlying myogenic reactivity. J Appl Physiol (1985) 2001; 91:973-83. [PMID: 11457816 DOI: 10.1152/jappl.2001.91.2.973] [Citation(s) in RCA: 204] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The smooth muscle of arterioles responds to an increase in intraluminal pressure with vasoconstriction and with vasodilation when pressure is decreased. Such myogenic vasoconstriction provides a level of basal tone that enables arterioles to appropriately adjust diameter in response to neurohumoral stimuli. Key in this process of mechanotransduction is the role of changes in intracellular Ca(2+). However, it is becoming clear that considerable complexity exists in the spatiotemporal characteristics of the Ca(2+) signal and that changes in intracellular Ca(2+) may play roles other than direct effects on the contractile process via activation of myosin light-chain phosphorylation. The involvement of Ca(2+) may extend to modulation of ion channels and release of Ca(2+) from the sarcoplasmic reticulum, alterations in Ca(2+) sensitivity, and coupling between cells within the vessel wall. The purpose of this brief review is to summarize the current literature relating to Ca(2+) and the arteriolar myogenic response. Consideration is given to coupling of Ca(2+) changes to the mechanical stimuli, sources of Ca(2+), involvement of ion channels, and spatiotemporal aspects of intracellular Ca(2+) signaling.
Collapse
Affiliation(s)
- M A Hill
- Microvascular Biology Group, School of Medical Sciences, RMIT University, Bundoora, Victoria 3083, Australia.
| | | | | | | | | |
Collapse
|
40
|
Rosado JA, Rosenzweig I, Harding S, Sage SO. Tumor necrosis factor-alpha inhibits store-mediated Ca2+ entry in the human hepatocellular carcinoma cell line HepG2. Am J Physiol Cell Physiol 2001; 280:C1636-44. [PMID: 11350760 DOI: 10.1152/ajpcell.2001.280.6.c1636] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tumor necrosis factor-alpha (TNF-alpha) is an important component of the early signaling pathways leading to liver regeneration and proliferation, but it is also responsible for several hepatotoxic effects. We have investigated the effect of TNF-alpha on thapsigargin (TG)-induced store-mediated Ca2+ entry (SMCE) in the human hepatocellular carcinoma cell line HepG2. In these cells, short-term (10 min) exposure to TNF-alpha slightly increased SMCE. In contrast, long-term (12 h) exposure to TNF-alpha significantly reduced SMCE. This effect was reversed by coincubation with atrial natriuretic peptide (ANP), which itself had no effect on SMCE. Cytochalasin D and latrunculin A, inhibitors of actin polymerization, abolished SMCE. Long-term exposure of HepG2 cells to TNF-alpha abolished TG-induced actin polymerization and membrane association of Ras proteins. When TNF-alpha was added in combination with ANP, these effects were reduced. These findings suggest that in HepG2 cells, TNF-alpha inhibits SMCE by affecting reorganization of the actin cytoskeleton, probably by interfering with the activation of Ras proteins, and that ANP protects against these inhibitory effects of TNF-alpha.
Collapse
Affiliation(s)
- J A Rosado
- Department of Physiology, University of Cambridge, Cambridge CB2 3EG, United Kingdom
| | | | | | | |
Collapse
|
41
|
Wieclawska B, Rozalski M, Trojanowski Z, Watala C. Modulators of intraplatelet calcium concentration affect the binding of thrombospondin to blood platelets in healthy donors and patients with type 2 diabetes mellitus. Eur J Haematol 2001; 66:396-403. [PMID: 11488939 DOI: 10.1034/j.1600-0609.2001.066006396.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
UNLABELLED Thrombospondin (TSP), which is secreted from alpha-granules of activated platelets, binds to its surface receptor (CD36) in the presence of Ca2+. OBJECTIVES We monitored how the modulation of intraplatelet Ca2+ affects TSP binding to CD36 on platelets from healthy donors and patients with type 2 diabetes mellitus. We also aimed to verify whether the impaired Ca2+ mobilisation in diabetes influences TSP binding upon the pharmacological modulation of calcium transport. METHODS Whole blood cytometry was used to monitor TSP release/binding and CD36 presentation in platelets from 28 type 2 patients and 33 healthy donors. RESULTS No significant changes in TSP and CD36 levels were revealed between the groups in circulating platelets and TRAP-, collagen- or thrombin-activated platelets. In healthy donors, 1 microM thapsigargin (TG) elevated the TRAP-activated TSP binding (by up to 50%, p<0.001), 5 mM EGTA reversed the effect (by up to 85%, p<0.001), and overcame the effect of TG when used together. Less profoundly expressed effects occurred in the NIDDM group. In both groups TG increased the presentation of CD36 in TRAP-stimulated platelets (p<0.05), whereas EGTA lowered the TRAP-stimulated increase in CD36 (p<0.001). The inhibition of CD36 by EGTA was stronger in healthy volunteers (41% vs. 32%, respectively, p<0.05), whereas the activation by TG was higher in the NIDDM group (11% vs. 27%, p<0.05). When acting together the suppressive effects of EGTA on TG-dependent Ca2+ mobilisation were much attenuated in diabetic subjects (p<0.05). CONCLUSION Both the release of TSP and CD36 presentation are under the influence of agents modulating intracellular Ca2+. Diabetic platelets seem more vulnerable to the releasers of cytosolic [Ca2+] and more resistant to the blockers of cytosolic [Ca2+] mobilisation.
Collapse
Affiliation(s)
- B Wieclawska
- Laboratory of Haemostasis and Haemostatic Disorders, Medical University of Lodz, Lodz, Poland
| | | | | | | |
Collapse
|
42
|
Hoque KM, Pal A, Nair GB, Chattopadhyay S, Chakrabarti MK. Evidence of calcium influx across the plasma membrane depends upon the initial rise of cytosolic calcium with activation of IP(3) in rat enterocytes by heat-stable enterotoxin of Vibrio cholerae non-O1. FEMS Microbiol Lett 2001; 196:45-50. [PMID: 11257546 DOI: 10.1111/j.1574-6968.2001.tb10538.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
In response to heat-stable enterotoxin of Vibrio cholerae non-O1, the initial rise of cytosolic Ca(2+) occurred with activation of IP(3). Chelation of extracellular Ca(2+) with EGTA and suspension of cells in Ca(2+) free buffer both demonstrated the involvement of internal stores in the rise of [Ca(2+)]i. Cells pretreated with dantrolene resulted in decrease of [Ca(2+)]i response which suggested that the rise of intracellular level of Ca(2+) was mostly due to the mobilization from IP(3) sensitive stores. When the cytosolic Ca(2+) was chelated by loading the cells with BAPTA, NAG-ST could not induce Ca(2+) entry to the cell as assessed by Mn(2+) quenching of fura-2 fluorescence which suggested that calcium influx across the plasma membrane depends upon initial rise of this bivalent cation that maintained the sustained phase of [Ca(2+)]i response. Addition of toxin to the fura-2-loaded cells, preincubated with lanthanum chloride, resulted in reduction of [Ca(2+)]i level with a short duration of irregular sustained phase further suggesting that the influx of Ca(2+) across the plasma membrane might be through the calcium channel.
Collapse
Affiliation(s)
- K M Hoque
- Pathophysiology Division, national Institute of Cholera and Enteric Diseases, Calcutta, India
| | | | | | | | | |
Collapse
|
43
|
|
44
|
Sugatani J, Iwai T, Watanabe M, Machida K, Tanaka T, Maeda T, Miwa M. Inhibition of rabbit platelet aggregation by nucleoside 5'-alkylphosphates: correlation with inhibition of agonist-induced calcium influx. Biochem Pharmacol 2000; 60:197-205. [PMID: 10825464 DOI: 10.1016/s0006-2952(00)00323-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We investigated the effects of uridine 5'-alkylphosphates on agonist-induced aggregation, increased intracellular calcium concentration [Ca(2+)](i), and Ca(2+) (Mn(2+)) influx in washed rabbit platelets. Uridine 5'-hexadecylphosphate (UMPC16) and uridine 5'-eicosylphosphate (UMPC20) at a concentration of 1 x 10(-5) M inhibited platelet aggregation induced by platelet-activating factor (PAF), thrombin, arachidonic acid, and ADP. UMPC16 did not cause significant interference in the binding of [(3)H-acetyl]PAF to platelets. The inhibition of PAF-induced platelet aggregation by UMPC16 was dependent upon the addition time; UMPC16 was ineffective at 60 sec when the extracellular calcium uptake reached the maximum level in PAF-stimulated platelets. Furthermore, UMPC16 inhibited guanosine 5'-O-(3-thiotriphosphate)-induced platelet aggregation but did not affect ionophore A23187- and calcium-independent agonist phorbol 12-myristate 13-acetate-induced platelet aggregation. UMPC16 markedly inhibited the Ca(2+) (Mn(2+)) influx induced by PAF and ADP, and partly inhibited the [Ca(2+)](i) increase induced by the receptor-mediated stimulation. On the other hand, UMPC16 did not affect the [Ca(2+)](i) increase and Ca(2+) (Mn(2+)) influx induced by ionomycin. These experiments suggest that inhibition of calcium influx associated with receptor-mediated platelet activation may be involved in the action of UMPC16.
Collapse
Affiliation(s)
- J Sugatani
- Department of Pharmaco-Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 422-8526, Shizuoka, Japan.
| | | | | | | | | | | | | |
Collapse
|
45
|
Cusack NJ, Hourani SM. Platelet P2 receptors: from curiosity to clinical targets. JOURNAL OF THE AUTONOMIC NERVOUS SYSTEM 2000; 81:37-43. [PMID: 10869698 DOI: 10.1016/s0165-1838(00)00151-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Adenosine 5'-diphosphate (ADP) is a paracrine mediator that activates human blood platelets, causing them to become adhesive and thereby contributing to their role in hemostasis. The actions of ADP were initially thought to be mediated by a unique ADP receptor termed P2(T) found only on platelets and antagonized by ATP, but it appears that at least two P2Y receptor subtypes are involved, a P2Y(1) receptor linked in some way to control of intracellular-free calcium levels and another P2Y receptor linked via an inhibitory G protein to adenylate cyclase. In addition, the presence of excitatory P2X(1) receptors that mediate the influx of monovalent and divalent cations in response to both ADP and ATP has been demonstrated. The precise contribution that each of these P2 receptors make to the overall phenomena associated with platelet aggregation, adhesion and hemostasis is yet to be defined. Antithrombotic agents that interfere with the actions of ADP are marketed, and P2 receptor antagonists are entering clinical trials for acute treatments of thrombosis. This review seeks to summarize the present state of knowledge of platelet P2 receptor pharmacology and therapeutics.
Collapse
Affiliation(s)
- N J Cusack
- Discovery Therapeutics, Inc., 2028 Dabney Road, Suite E-17, Richmond, VA 23230, USA.
| | | |
Collapse
|
46
|
Bonnefoy A, Liu Q, Legrand C, Frojmovic MM. Efficiency of platelet adhesion to fibrinogen depends on both cell activation and flow. Biophys J 2000; 78:2834-43. [PMID: 10827966 PMCID: PMC1300871 DOI: 10.1016/s0006-3495(00)76826-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The kinetics of adhesion of platelets to fibrinogen (Fg) immobilized on polystyrene latex beads (Fg-beads) was determined in suspensions undergoing Couette flow at well-defined homogeneous shear rates. The efficiency of platelet adhesion to Fg-beads was compared for ADP-activated versus "resting" platelets. The effects of the shear rate (100-2000 s(-1)), Fg density on the beads (24-2882 Fg/microm(2)), the concentration of ADP used to activate the platelets, and the presence of soluble fibrinogen were assessed. "Resting" platelets did not specifically adhere to Fg-beads at levels detectable with our methodology. The apparent efficiency of platelet adhesion to Fg-beads readily correlated with the proportion of platelets "quantally" activated by doses of ADP, i.e., only ADP-activated platelets appeared to adhere to Fg-beads, with a maximal adhesion efficiency of 6-10% at shear rates of 100-300 s(-1), decreasing with increasing shear rates up to 2000 s(-1). The adhesion efficiency was found to decrease by only threefold when decreasing the density of Fg at the surface of the beads by 100-fold, with only moderate decreases in the presence of physiologic concentrations of soluble Fg. These adhesive interactions were also compared using activated GPIIbIIIa-coated beads. Our studies provide novel model particles for studying platelet adhesion relevant to hemostasis and thrombosis, and show how the state of activation of the platelet and the local flow conditions regulate Fg-dependent adhesion.
Collapse
Affiliation(s)
- A Bonnefoy
- Unité 353 INSERM, Institut d'Hématologie, Université Paris VII, Hôpital St. Louis, Paris, France
| | | | | | | |
Collapse
|
47
|
Rosado JA, Sage SO. Farnesylcysteine analogues inhibit store-regulated Ca2+ entry in human platelets: evidence for involvement of small GTP-binding proteins and actin cytoskeleton. Biochem J 2000; 347 Pt 1:183-92. [PMID: 10727417 PMCID: PMC1220946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
We have investigated the mechanism of Ca(2+) entry into fura-2-loaded human platelets by preventing the prenylation of proteins such as small GTP-binding proteins. The farnesylcysteine analogues farnesylthioacetic acid (FTA) and N-acetyl-S-geranylgeranyl-L-cysteine (AGGC), which are inhibitors of the methylation of prenylated and geranylgeranylated proteins respectively, significantly decreased thrombin-evoked increases in intracellular free Ca(2+) concentration ([Ca(2+)](i)) in the presence, but not in the absence, of external Ca(2+), suggesting a relatively selective inhibition of Ca(2+) entry over internal release. Both these compounds and N-acetyl-S-farnesyl-L-cysteine, which had similar effects to those of FTA, also decreased Ca(2+) entry evoked by the depletion of intracellular Ca(2+) stores with thapsigargin. The inactive control N-acetyl-S-geranyl-L-cysteine was without effect. Patulin, an inhibitor of prenylation that is inert with respect to methyltransferases, also decreased store-regulated Ca(2+) entry. Cytochalasin D, an inhibitor of actin polymerization, significantly decreased store-regulated Ca(2+) entry in a time-dependent manner. Both cytochalasin D and the farnesylcysteine analogues FTA and AGGC inhibited actin polymerization; however, when evoking the same extent of decrease in actin filament formation, FTA and AGGC showed greater inhibitory effects on Ca(2+) entry, indicating a cytoskeleton-independent component in the regulation of Ca(2+) entry by small GTP-binding-protein. These findings suggest that prenylated proteins such as small GTP-binding proteins are involved in store-regulated Ca(2+) entry through actin cytoskeleton-dependent and cytoskeleton-independent mechanisms in human platelets.
Collapse
Affiliation(s)
- J A Rosado
- Department of Physiology, University of Cambridge, Downing Street, Cambridge CB2 3EG, U.K
| | | |
Collapse
|
48
|
Kondo I. Protein kinase C potentiates capacitative Ca2+ entry that links to steroidogenesis in bovine adrenocortical cells. JAPANESE JOURNAL OF PHARMACOLOGY 2000; 82:210-7. [PMID: 10887951 DOI: 10.1254/jjp.82.210] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
I investigated the role of protein kinase C (PKC) in regulation of the capacitative Ca2+ entry and steroidogenesis in bovine adrenocortical (BA) cells. Thapsigargin (TG)-treatment depleted intracellular Ca2+ stores followed by induction of Ca2+ influx from the extracellular pool and also increasing of Mn2+ influx as an indicator of divalent cation influx in BA cells. Calphostin C, a PKC inhibitor, inhibited the TG-induced [Ca2+]i elevation dose-dependently (0.1-1 microM) and attenuated Mn2+ entry. Phorbol 12-myristate 13-acetate (PMA), an activator of PKC, potentiated the elevation of [Ca2+]i and enhanced Mn2+ entry by TG treatment. These results suggest that PKC may modulate capacitative Ca2+ entry in BA cells. In the presence of extracellular Ca2+, TG enhanced cortisol production in BA cells. Calphostin C attenuated the TG-induced steroidogenesis dose-dependently (0.25-1 microM). PMA enhanced the steroidogenesis dose-dependently (1-100 nM). These results suggested that PKC may have a modulatory effect on the capacitative Ca2+ entry that links to steroidogenesis in BA cells.
Collapse
Affiliation(s)
- I Kondo
- Department of Anesthesiology, Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
49
|
Kitamura T, Murayama T, Nomura Y. Enhancement of Ca2+-induced noradrenaline release by vanadate in PC12 cells: possible involvement of tyrosine phosphorylation. Brain Res 2000; 854:165-71. [PMID: 10784118 DOI: 10.1016/s0006-8993(99)02299-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Tyrosine phosphorylation has been shown to participate in the signal cascade after receptor stimulation with neurotransmitters and neurotrophins. However, the role of tyrosine phosphorylation in the process(es) of neurotransmitter release has not been well established. The effects of orthovanadate (Na3VO4), an inhibitor of protein-tyrosine phosphatases, on cytosolic free Ca2+ concentrations ([Ca2+]i), phosphotyrosine accumulation and noradrenaline (NA) release in neurosecretory PC12 cells were investigated. Addition of Na3VO4 enhanced ionomycin-stimulated [3H]NA release in a concentration-dependent manner, although Na3VO4 alone had no effect. Na3VO4 also enhanced [3H]NA release induced by P2 receptor stimulation with adenosine 5'-O-(3-thiotriphosphate) (ATPgammaS) or by depolarization with 50 mM KCl, which stimulated a [Ca2+]i increase. A cell permeable inhibitor of protein-tyrosine phosphatases, L-p-bromotetramisole oxalate, at 0.3 mM enhanced ionomycin-stimulated [3H]NA release, although pervanadate had no effect. Addition of 5 mM Na3VO4 stimulated phosphotyrosine accumulation in several protein bands such as p130cas, but did not increase [Ca2+]i in PC12 cells. These findings suggest that the tyrosine phosphorylation pathway regulates Ca2+-stimulated NA release without changes of [Ca2+]i in PC12 cells.
Collapse
Affiliation(s)
- T Kitamura
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | | | | |
Collapse
|
50
|
Gende OA. Lithium opens store-operated channels in human platelets. Biochem Biophys Res Commun 2000; 267:546-50. [PMID: 10631099 DOI: 10.1006/bbrc.1999.2002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ca(2+) release from internal stores as a result of activation of phospholipase C or inhibition of the endoplasmic reticulum pump is accompanied by Ca(2+) influx from the extracellular space. Measurement of intracellular calcium concentration and fluorescence quenching in Fura2-loaded cells showed that platelets preincubated in lithium have significantly higher basal, but lower agonist-stimulated influx of Mn(2+) (acting as a surrogate of Ca(2+) influx), than platelets reloaded with calcium in a normal sodium medium. There is no difference in the basal entry of divalent ion in platelets preincubated in sodium, lithium, or N-methyl glucamine in the absence of calcium. In platelets preincubated in lithium there is a higher basal Mn(2+) entry without further increase upon store depletion by thapsigargin. In contrast, a significant increase in the divalent ion influx was found in sodium or N-methyl glucamine attributable to the opening of channels sensitive to store depletion. In the absence of extracellular calcium, the empty store opens channels and Li(+) did not have additional effect on channels that are already open. The refilling of the stores with Ca(2+) suppresses Mn(2+) entry after sodium or NMG preincubation, but not after lithium preincubation. We propose that lithium induces a calcium influx throughout store-operated channels. This hypothesis may explain the lack of additivity, in cell preincubated in lithium, of basal entry and thapsigargin-triggered entry of calcium.
Collapse
Affiliation(s)
- O A Gende
- Centro de Investigaciones Cardiovasculares, Universidad Nacional de La Plata, 60 Y 120, La Plata, 1900, Argentina.
| |
Collapse
|