1
|
Pick T, Gamayun I, Tinschert R, Cavalié A. Kinetics of the thapsigargin-induced Ca2+ mobilisation: A quantitative analysis in the HEK-293 cell line. Front Physiol 2023; 14:1127545. [PMID: 37051019 PMCID: PMC10083721 DOI: 10.3389/fphys.2023.1127545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
Thapsigargin (TG) inhibits the sarco/endoplasmic reticulum Ca2+ ATPase (SERCA) pump and, when applied acutely, it initiates a Ca2+ mobilisation that begins with the loss of Ca2+ from the endoplasmic reticulum (ER) and culminates with store-operated Ca2+ entry (SOCE) from the extracellular space. Using the popular model cell line HEK-293, we quantified TG-induced changes in cytosolic and ER Ca2+ levels using FURA-2 and the FRET-based ER Ca2+ sensor D1ER, respectively. Our analysis predicts an ER Ca2+ leak of 5–6 µM⋅s−1 for the typical basal ER Ca2+ level of 335–407 µM in HEK-293 cells. The resulting cytosolic Ca2+ transients reached peak amplitudes of 0.6–1.0 µM in the absence of external Ca2+ and were amplified by SOCE that amounted to 28–30 nM⋅s−1 in 1 mM external Ca2+. Additionally, cytosolic Ca2+ transients were shaped by a Ca2+ clearance of 10–13 nM⋅s−1. Using puromycin (PURO), which enhances the ER Ca2+ leak, we show that TG-induced cytosolic Ca2+ transients are directly related to ER Ca2+ levels and to the ER Ca2+ leak. A one-compartment model incorporating ER Ca2+ leak and cytosolic Ca2+ clearance accounted satisfactorily for the basic features of TG-induced Ca2+ transients and underpinned the rule that an increase in amplitude associated with shortening of TG-induced cytosolic Ca2+ transients most likely reflects an increase in ER Ca2+ leak.
Collapse
Affiliation(s)
- Tillman Pick
- *Correspondence: Tillman Pick, ; Adolfo Cavalié,
| | | | | | | |
Collapse
|
2
|
Sánchez-Vázquez VH, Martínez-Martínez E, Gallegos-Gómez ML, Arias JM, Pallafacchina G, Rizzuto R, Guerrero-Hernández A. Heterogeneity of the endoplasmic reticulum Ca 2+ store determines colocalization with mitochondria. Cell Calcium 2023; 109:102688. [PMID: 36538845 DOI: 10.1016/j.ceca.2022.102688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 11/14/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
Contact sites between the endoplasmic reticulum (ER) and mitochondria play a pivotal role in cell signaling, and the interaction between these organelles is dynamic and finely regulated. We have studied the role of ER Ca2+ concentration ([Ca2+]ER) in modulating this association in HeLa and HEK293 cells and human fibroblasts. According to Manders' coefficient, ER-mitochondria colocalization varied depending on the ER marker; it was the highest with ER-Tracker and the lowest with ER Ca2+ indicators (Mag-Fluo-4, erGAP3, and G-CEPIA1er) in both HeLa cells and human fibroblasts. Only GEM-CEPIA1er displayed a high colocalization with elongated mitochondria in HeLa cells, this ER Ca2+ indicator reveals low Ca2+ regions because this ion quenches its fluorescence. On the contrary, the typical rounded and fragmented mitochondria of HEK293 cells colocalized with Mag-Fluo-4 and, to a lesser extent, with GEM-CEPIA1er. The ablation of the three IP3R isoforms in HEK293 cells increased mitochondria-GEM-CEPIA1er colocalization. This pattern of colocalization was inversely correlated with the rate of ER Ca2+ leak evoked by thapsigargin (Tg). Moreover, Tg and Histamine in the absence of external Ca2+ increased mitochondria-ER colocalization. On the contrary, in the presence of external Ca2+, both Bafilomycin A1 and Tg reduced the mitochondria-ER interaction. Notably, knocking down MCU decreased mitochondria-ER colocalization. Overall, our data suggest that the [Ca2+] is not homogenous within the ER lumen and that mitochondria-ER interaction is modulated by the ER Ca2+ leak and the [Ca2+]i.
Collapse
Affiliation(s)
| | | | | | - Juan M Arias
- Programa de Neurociencias-UIICSE, Facultad de Estudios Superiores Iztacala, UNAM; Tlalnepantla de Baz, Estado de México, 54090, Mexico
| | - Giorgia Pallafacchina
- CNR, Neuroscience Institute, Padua, 35131. Italy; Department of Biomedical Sciences, University of Padua, Padua, 35131. Italy
| | - Rosario Rizzuto
- Department of Biomedical Sciences, University of Padua, Padua, 35131. Italy
| | | |
Collapse
|
3
|
Dagnino-Acosta A, Guerrero-Hernandez A. PKC Inhibits Sec61 Translocon-Mediated Sarcoplasmic Reticulum Ca2+ Leak in Smooth Muscle Cells. Front Physiol 2022; 13:925023. [PMID: 35837019 PMCID: PMC9275787 DOI: 10.3389/fphys.2022.925023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/03/2022] [Indexed: 01/13/2023] Open
Abstract
PKC inhibitors stimulate Ca2+ release from internal stores in diverse cell types. Our data indicate that this action cannot be explained by an increased agonist-induced IP3 production or an overloaded SR Ca2+ pool in smooth muscle cells from guinea pig urinary bladder. The incubation of these cells with three different PKC inhibitors, such as Go6976, Go6983, and BIM 1, resulted in a higher SR Ca2+ leak revealed by inhibition of the SERCA pump with thapsigargin. This SR Ca2+ leakage was sensitive to protein translocation inhibitors such as emetine and anisomycin. Since this increased SR Ca2+ leak did not result in a depleted SR Ca2+ store, we have inferred there was a compensatory increase in SERCA pump activity, resulting in a higher steady-state. This new steady-state increased the frequency of Spontaneous Transient Outward Currents (STOCs), which reflect the activation of high conductance, Ca2+-sensitive potassium channels in response to RyR-mediated Ca2+ sparks. This increased STOC frequency triggered by PKC inhibition was restored to normal by inhibiting translocon-mediated Ca2+ leak with emetine. These results suggest a critical role of PKC-mediated translocon phosphorylation in regulating SR Ca2+ steady-state, which, in turn, alters SR Ca2+ releasing activity.
Collapse
Affiliation(s)
- Adan Dagnino-Acosta
- Centro Universitario de Investigaciones Biomédicas, CONACYT-Universidad de Colima, Colima, Mexico
| | - Agustín Guerrero-Hernandez
- Departamento de Bioquímica, Centro de Investigación y de Estudios Avanzados, Mexico City, Mexico
- *Correspondence: Agustín Guerrero-Hernandez,
| |
Collapse
|
4
|
PKC-Mediated Orai1 Channel Phosphorylation Modulates Ca2+ Signaling in HeLa Cells. Cells 2022; 11:cells11132037. [PMID: 35805121 PMCID: PMC9266177 DOI: 10.3390/cells11132037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 05/15/2022] [Accepted: 05/27/2022] [Indexed: 12/04/2022] Open
Abstract
The overexpression of the Orai1 channel inhibits SOCE when using the Ca2+ readdition protocol. However, we found that HeLa cells overexpressing the Orai1 channel displayed enhanced Ca2+ entry and a limited ER depletion in response to the combination of ATP and thapsigargin (TG) in the presence of external Ca2+. As these effects require the combination of an agonist and TG, we decided to study whether the phosphorylation of Orai1 S27/S30 residues had any role using two different mutants: Orai1-S27/30A (O1-AA, phosphorylation-resistant) and Orai1-S27/30D (O1-DD, phosphomimetic). Both O1-wt and O1-AA supported enhanced Ca2+ entry, but this was not the case with O1-E106A (dead-pore mutant), O1-DD, and O1-AA-E106A, while O1-wt, O1-E106A, and O1-DD inhibited the ATP and TG-induced reduction of ER [Ca2+], suggesting that the phosphorylation of O1 S27/30 interferes with the IP3R activity. O1-wt and O1-DD displayed an increased interaction with IP3R in response to ATP and TG; however, the O1-AA channel decreased this interaction. The expression of mCherry-O1-AA increased the frequency of ATP-induced sinusoidal [Ca2+]i oscillations, while mCherry-O1-wt and mCherry-O1-DD decreased this frequency. These data suggest that the combination of ATP and TG stimulates Ca2+ entry, and the phosphorylation of Orai1 S27/30 residues by PKC reduces IP3R-mediated Ca2+ release.
Collapse
|
5
|
SUPER-RESOLUTION MICROSCOPY FOR THE STUDY OF STORE-OPERATED CALCIUM ENTRY. Cell Calcium 2022; 104:102595. [DOI: 10.1016/j.ceca.2022.102595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 11/19/2022]
|
6
|
Pick T, Beck A, Gamayun I, Schwarz Y, Schirra C, Jung M, Krause E, Niemeyer BA, Zimmermann R, Lang S, Anken EV, Cavalié A. Remodelling of Ca 2+ homeostasis is linked to enlarged endoplasmic reticulum in secretory cells. Cell Calcium 2021; 99:102473. [PMID: 34560367 DOI: 10.1016/j.ceca.2021.102473] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/27/2021] [Accepted: 09/08/2021] [Indexed: 11/30/2022]
Abstract
The endoplasmic reticulum (ER) is extensively remodelled during the development of professional secretory cells to cope with high protein production. Since ER is the principal Ca2+ store in the cell, we characterised the Ca2+ homeostasis in NALM-6 and RPMI 8226 cells, which are commonly used as human pre-B and antibody secreting plasma cell models, respectively. Expression levels of Sec61 translocons and the corresponding Sec61-mediated Ca2+ leak from ER, Ca2+ storage capacity and store-operated Ca2+ entry were significantly enlarged in the secretory RPMI 8226 cell line. Using an immunoglobulin M heavy chain producing HeLa cell model, we found that the enlarged Ca2+ storage capacity and Ca2+ leak from ER are linked to ER expansion. Our data delineates a developmental remodelling of Ca2+ homeostasis in professional secretory cells in which a high Sec61-mediated Ca2+ leak and, thus, a high Ca2+ turnover in the ER is backed up by enhanced store-operated Ca2+ entry.
Collapse
Affiliation(s)
- Tillman Pick
- Experimental and Clinical Pharmacology and Toxicology, Pre-clinical Center for Molecular Signalling (PZMS), Saarland University, 66421 Homburg, Germany.
| | - Andreas Beck
- Experimental and Clinical Pharmacology and Toxicology, Pre-clinical Center for Molecular Signalling (PZMS), Saarland University, 66421 Homburg, Germany
| | - Igor Gamayun
- Experimental and Clinical Pharmacology and Toxicology, Pre-clinical Center for Molecular Signalling (PZMS), Saarland University, 66421 Homburg, Germany
| | - Yvonne Schwarz
- Molecular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, 66421 Homburg, Germany
| | - Claudia Schirra
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, 66421 Homburg, Germany
| | - Martin Jung
- Medical Biochemistry and Molecular Biology, Pre-clinical Centre for Molecular Signalling (PZMS), Saarland University, 66421 Homburg, Germany
| | - Elmar Krause
- Cellular Neurophysiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, 66421 Homburg, Germany
| | - Barbara A Niemeyer
- Molecular Biophysics, Center for Integrative Physiology and Molecular Medicine (CIPMM), Saarland University, 66421 Homburg, Germany
| | - Richard Zimmermann
- Medical Biochemistry and Molecular Biology, Pre-clinical Centre for Molecular Signalling (PZMS), Saarland University, 66421 Homburg, Germany
| | - Sven Lang
- Medical Biochemistry and Molecular Biology, Pre-clinical Centre for Molecular Signalling (PZMS), Saarland University, 66421 Homburg, Germany
| | - Eelco van Anken
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute and Università Vita-Salute San Raffaele, Milan, Italy
| | - Adolfo Cavalié
- Experimental and Clinical Pharmacology and Toxicology, Pre-clinical Center for Molecular Signalling (PZMS), Saarland University, 66421 Homburg, Germany.
| |
Collapse
|
7
|
Lemos FO, Bultynck G, Parys JB. A comprehensive overview of the complex world of the endo- and sarcoplasmic reticulum Ca 2+-leak channels. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119020. [PMID: 33798602 DOI: 10.1016/j.bbamcr.2021.119020] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 12/11/2022]
Abstract
Inside cells, the endoplasmic reticulum (ER) forms the largest Ca2+ store. Ca2+ is actively pumped by the SERCA pumps in the ER, where intraluminal Ca2+-binding proteins enable the accumulation of large amount of Ca2+. IP3 receptors and the ryanodine receptors mediate the release of Ca2+ in a controlled way, thereby evoking complex spatio-temporal signals in the cell. The steady state Ca2+ concentration in the ER of about 500 μM results from the balance between SERCA-mediated Ca2+ uptake and the passive leakage of Ca2+. The passive Ca2+ leak from the ER is often ignored, but can play an important physiological role, depending on the cellular context. Moreover, excessive Ca2+ leakage significantly lowers the amount of Ca2+ stored in the ER compared to normal conditions, thereby limiting the possibility to evoke Ca2+ signals and/or causing ER stress, leading to pathological consequences. The so-called Ca2+-leak channels responsible for Ca2+ leakage from the ER are however still not well understood, despite over 20 different proteins have been proposed to contribute to it. This review has the aim to critically evaluate the available evidence about the various channels potentially involved and to draw conclusions about their relative importance.
Collapse
Affiliation(s)
- Fernanda O Lemos
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium.
| |
Collapse
|
8
|
Huo J, Dong H. Gating and regulation of the calcium release-activated calcium channel: Recent progress from experiments and molecular modeling. Biopolymers 2021; 111:e23392. [PMID: 33460071 DOI: 10.1002/bip.23392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 11/08/2022]
Abstract
Calcium release-activated calcium (CRAC) channels are highly calcium ion (Ca2+)-selective channels in the plasma membrane. The transient drop of endoplasmic reticulum Ca2+ level activates its calcium sensor stromal interaction molecule (STIM) and then triggers the gating of the CRAC channel pore unit Orai. This process involves a variety of activities of the immune system. Therefore, understanding how the activation and regulation of the CRAC channel can be accomplished is essential. Here we briefly summarize the recent progress on Orai gating and its regulation by 2-aminoethoxydiphenylborate (2-APB) obtained from structural biology studies, biochemical and electrophysiological measurements, as well as molecular modeling. Indeed, integration between experiments and computations has further deepened our understanding of the channel gating and regulation.
Collapse
Affiliation(s)
- Jun Huo
- Kuang Yaming Honors School, Nanjing University, Nanjing, China
| | - Hao Dong
- Kuang Yaming Honors School, Nanjing University, Nanjing, China.,Institute for Brain Sciences, Nanjing University, Nanjing, China
| |
Collapse
|
9
|
Transcriptomic Profiling of Ca2+ Transport Systems During the Formation of the Cerebral Cortex in Mice. Cells 2020; 9:cells9081800. [PMID: 32751129 PMCID: PMC7465657 DOI: 10.3390/cells9081800] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/20/2020] [Accepted: 07/24/2020] [Indexed: 01/05/2023] Open
Abstract
Cytosolic calcium (Ca2+) transients control key neural processes, including neurogenesis, migration, the polarization and growth of neurons, and the establishment and maintenance of synaptic connections. They are thus involved in the development and formation of the neural system. In this study, a publicly available whole transcriptome sequencing (RNA-Seq) dataset was used to examine the expression of genes coding for putative plasma membrane and organellar Ca2+-transporting proteins (channels, pumps, exchangers, and transporters) during the formation of the cerebral cortex in mice. Four ages were considered: embryonic days 11 (E11), 13 (E13), and 17 (E17), and post-natal day 1 (PN1). This transcriptomic profiling was also combined with live-cell Ca2+ imaging recordings to assess the presence of functional Ca2+ transport systems in E13 neurons. The most important Ca2+ routes of the cortical wall at the onset of corticogenesis (E11–E13) were TACAN, GluK5, nAChR β2, Cav3.1, Orai3, transient receptor potential cation channel subfamily M member 7 (TRPM7) non-mitochondrial Na+/Ca2+ exchanger 2 (NCX2), and the connexins CX43/CX45/CX37. Hence, transient receptor potential cation channel mucolipin subfamily member 1 (TRPML1), transmembrane protein 165 (TMEM165), and Ca2+ “leak” channels are prominent intracellular Ca2+ pathways. The Ca2+ pumps sarco/endoplasmic reticulum Ca2+ ATPase 2 (SERCA2) and plasma membrane Ca2+ ATPase 1 (PMCA1) control the resting basal Ca2+ levels. At the end of neurogenesis (E17 and onward), a more numerous and diverse population of Ca2+ uptake systems was observed. In addition to the actors listed above, prominent Ca2+-conducting systems of the cortical wall emerged, including acid-sensing ion channel 1 (ASIC1), Orai2, P2X2, and GluN1. Altogether, this study provides a detailed view of the pattern of expression of the main actors participating in the import, export, and release of Ca2+. This work can serve as a framework for further functional and mechanistic studies on Ca2+ signaling during cerebral cortex formation.
Collapse
|
10
|
Arachidonic Acid Attenuates Cell Proliferation, Migration and Viability by a Mechanism Independent on Calcium Entry. Int J Mol Sci 2020; 21:ijms21093315. [PMID: 32392840 PMCID: PMC7247542 DOI: 10.3390/ijms21093315] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 12/13/2022] Open
Abstract
Arachidonic acid (AA) is a phospholipase A2 metabolite that has been reported to mediate a plethora of cellular mechanisms involved in healthy and pathological states such as platelet aggregation, lymphocyte activation, and tissue inflammation. AA has been described to activate Ca2+ entry through the arachidonate-regulated Ca2+-selective channels (ARC channels). Here, the analysis of the changes in the intracellular Ca2+ homeostasis revealed that, despite MDA-MB-231 cells expressing the ARC channel components Orai1, Orai3, and STIM1, AA does not evoke Ca2+ entry in these cells. We observed that AA evokes Ca2+ entry in MDA-MB-231 cells transiently expressing ARC channels. Nevertheless, MDA-MB-231 cell treatment with AA reduces cell proliferation and migration while inducing cell death through apoptosis. The latter mostly likely occurs via mitochondria membrane depolarization and the activation of caspases-3, -8, and -9. Altogether, our results indicate that AA exerts anti-tumoral effects on MDA-MB-231 cells, without having any effect on non-tumoral breast epithelial cells, by a mechanism that is independent on the activation of Ca2+ influx via ARC channels.
Collapse
|
11
|
Kappel S, Kilch T, Baur R, Lochner M, Peinelt C. The Number and Position of Orai3 Units within Heteromeric Store-Operated Ca 2+ Channels Alter the Pharmacology of I CRAC. Int J Mol Sci 2020; 21:ijms21072458. [PMID: 32252254 PMCID: PMC7178029 DOI: 10.3390/ijms21072458] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/24/2020] [Accepted: 03/30/2020] [Indexed: 12/21/2022] Open
Abstract
Store-operated heteromeric Orai1/Orai3 channels have been discussed in the context of aging, cancer, and immune cell differentiation. In contrast to homomeric Orai1 channels, they exhibit a different pharmacology upon application of reactive oxygen species (ROS) or 2-aminoethoxydiphenyl borate (2-APB) in various cell types. In endogenous cells, subunit composition and arrangement may vary and cannot be defined precisely. In this study, we used patch-clamp electrophysiology to investigate the 2-APB profile of store-operated and store-independent homomeric Orai1 and heteromeric Orai1/Orai3 concatenated channels with defined subunit compositions. As has been shown previous, one or more Orai3 subunit(s) within the channel result(s) in decreased Ca2+ release activated Ca2+ current (ICRAC). Upon application of 50 µM 2-APB, channels with two or more Orai3 subunits exhibit large outward currents and can be activated by 2-APB independent from storedepletion and/or the presence of STIM1. The number and position of Orai3 subunits within the heteromeric store-operated channel change ion conductivity of 2-APB-activated outward current. Compared to homomeric Orai1 channels, one Orai3 subunit within the channel does not alter 2-APB pharmacology. None of the concatenated channel constructs were able to exactly simulate the complex 2-APB pharmacology observed in prostate cancer cells. However, 2-APB profiles of prostate cancer cells are similar to those of concatenated channels with Orai3 subunit(s). Considering the presented and previous results, this indicates that distinct subtypes of heteromeric SOCE channels may be selectively activated or blocked. In the future, targeting distinct heteromeric SOCE channel subtypes may be the key to tailored SOCE-based therapies.
Collapse
Affiliation(s)
- Sven Kappel
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland; (S.K.); (R.B.); (M.L.)
| | | | - Roland Baur
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland; (S.K.); (R.B.); (M.L.)
| | - Martin Lochner
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland; (S.K.); (R.B.); (M.L.)
| | - Christine Peinelt
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bühlstrasse 28, 3012 Bern, Switzerland; (S.K.); (R.B.); (M.L.)
- Correspondence: ; Tel.: +41-31-631-3415
| |
Collapse
|
12
|
Morgan AJ, Yuan Y, Patel S, Galione A. Does lysosomal rupture evoke Ca 2+ release? A question of pores and stores. Cell Calcium 2019; 86:102139. [PMID: 31881482 DOI: 10.1016/j.ceca.2019.102139] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/05/2019] [Accepted: 12/05/2019] [Indexed: 02/04/2023]
Abstract
Lysosomotropic agents have been used to permeabilize lysosomes and thereby implicate these organelles in diverse cellular processes. Since lysosomes are Ca2+ stores, this rupturing action, particularly that induced by GPN, has also been used to rapidly release Ca2+ from lysosomes. However, a recent study has questioned the mechanism of action of GPN and concluded that, acutely, it does not permeabilize lysosomes but releases Ca2+ directly from the ER instead. We therefore appraise these provocative findings in the context of the existing literature. We suggest that further work is required to unequivocally rule out lysosomes as contributors to GPN-evoked Ca2+ signals.
Collapse
Affiliation(s)
- Anthony J Morgan
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom.
| | - Yu Yuan
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Sandip Patel
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Antony Galione
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, United Kingdom
| |
Collapse
|
13
|
Ovando-Zambrano JC, Arias-Montaño JA, Boucard AA. Alternative splicing event modifying ADGRL1/latrophilin-1 cytoplasmic tail promotes both opposing and dual cAMP signaling pathways. Ann N Y Acad Sci 2019; 1456:168-185. [PMID: 31339586 DOI: 10.1111/nyas.14198] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/11/2019] [Accepted: 07/02/2019] [Indexed: 12/31/2022]
Abstract
The adhesion G protein-coupled receptor ADGRL1/latrophilin-1 (LPHN1) stabilizes synapse formation through heterophilic interactions. A growing consensus is pointing to the role of LPHN1 in modulating intracellular levels of cAMP, although conflicting data exist. Variants of LPHN1 resulting from alternative splicing differ at multiple sites, two of which, designated as SSA and SSB, modify extracellular and intracellular receptor regions, respectively. While SSA splicing modulates receptor-ligand affinity, the function of SSB splicing remains elusive. Here, we explored the role of SSB in an attempt to unify current findings on LPHN1 signaling pathways by testing SSB-containing and SSB-deficient receptor variants in signaling paradigms involving interaction with their ligands neurexin and FLRT. cAMP competitive binding assays revealed that cells expressing either receptor variant exhibited a ligand-dependent decrease in the forskolin-induced cAMP accumulation. Surprisingly, the expression of SSB-containing LPHN1 promoted both constitutive and ligand-dependent cAMP production, whereas SSB-deficient LPHN1 did not. Pertussis toxin treatment unveiled a constitutive coupling to Gαi/o for SSB-containing LPHN1 while abrogating the ligand-mediated activation of Gαs . Importantly, neither receptor variant increased the intracellular concentration of Ca2+ nor MAP kinase activation in the presence of ligands. These results suggest that SSB splicing selectively affects the duality of LPHN1 signaling toward opposing cAMP pathways.
Collapse
Affiliation(s)
- José-Carlos Ovando-Zambrano
- Departamento de Fisiología, Biofísica, y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
| | - José-Antonio Arias-Montaño
- Departamento de Fisiología, Biofísica, y Neurociencias, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
| | - Antony A Boucard
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Ciudad de México, México
| |
Collapse
|
14
|
The Trans Golgi Region is a Labile Intracellular Ca 2+ Store Sensitive to Emetine. Sci Rep 2018; 8:17143. [PMID: 30464185 PMCID: PMC6249204 DOI: 10.1038/s41598-018-35280-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 09/10/2018] [Indexed: 01/22/2023] Open
Abstract
The Golgi apparatus (GA) is a bona fide Ca2+ store; however, there is a lack of GA-specific Ca2+ mobilizing agents. Here, we report that emetine specifically releases Ca2+ from GA in HeLa and HL-1 atrial myocytes. Additionally, it has become evident that the trans-Golgi is a labile Ca2+ store that requires a continuous source of Ca2+ from either the external milieu or from the ER, to enable it to produce a detectable transient increase in cytosolic Ca2+. Our data indicates that the emetine-sensitive Ca2+ mobilizing mechanism is different from the two classical Ca2+ release mechanisms, i.e. IP3 and ryanodine receptors. This newly discovered ability of emetine to release Ca2+ from the GA may explain why chronic consumption of ipecac syrup has muscle side effects.
Collapse
|
15
|
Bhattacharya A, Kumar J, Hermanson K, Sun Y, Qureshi H, Perley D, Scheidegger A, Singh BB, Dhasarathy A. The calcium channel proteins ORAI3 and STIM1 mediate TGF-β induced Snai1 expression. Oncotarget 2018; 9:29468-29483. [PMID: 30034631 PMCID: PMC6047677 DOI: 10.18632/oncotarget.25672] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/09/2018] [Indexed: 12/11/2022] Open
Abstract
Calcium influx into cells via plasma membrane protein channels is tightly regulated to maintain cellular homeostasis. Calcium channel proteins in the plasma membrane and endoplasmic reticulum have been linked to cancer, specifically during the epithelial-mesenchymal transition (EMT), a cell state transition process implicated in both cancer cell migration and drug resistance. The transcription factor SNAI1 (SNAIL) is upregulated during EMT and is responsible for gene expression changes associated with EMT, but the calcium channels required for Snai1 expression remain unknown. In this study, we show that blocking store-operated calcium entry (SOCE) with 2-aminoethoxydiphenylborane (2APB) reduces cell migration but, paradoxically, increases the level of TGF-β dependent Snai1 gene activation. We determined that this increased Snai1 transcription involves signaling through the AKT pathway and subsequent binding of NF-κB (p65) at the Snai1 promoter in response to TGF-β. We also demonstrated that the calcium channel protein ORAI3 and the stromal interaction molecule 1 (STIM1) are required for TGF-β dependent Snai1 transcription. These results suggest that calcium channels differentially regulate cell migration and Snai1 transcription, indicating that each of these steps could be targeted to ensure complete blockade of cancer progression.
Collapse
Affiliation(s)
- Atrayee Bhattacharya
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Janani Kumar
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
- Present address: MD Anderson Cancer Center, Mitchell Basic Sciences Research Building, TX, USA
| | - Kole Hermanson
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Yuyang Sun
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
- Present address: UT Health Science Center, San Antonio, San Antonio, TX, USA
| | - Humaira Qureshi
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
- Present address: Habib University, University Avenue, Gulistan-e-Jauhar, Karachi, Pakistan
| | - Danielle Perley
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Adam Scheidegger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| | - Brij B. Singh
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
- Present address: UT Health Science Center, San Antonio, San Antonio, TX, USA
| | - Archana Dhasarathy
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA
| |
Collapse
|
16
|
López E, Gómez-Gordo L, Cantonero C, Bermejo N, Pérez-Gómez J, Granados MP, Salido GM, Rosado Dionisio JA, Redondo Liberal PC. Stanniocalcin 2 Regulates Non-capacitative Ca 2+ Entry and Aggregation in Mouse Platelets. Front Physiol 2018; 9:266. [PMID: 29628897 PMCID: PMC5876523 DOI: 10.3389/fphys.2018.00266] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 03/08/2018] [Indexed: 12/11/2022] Open
Abstract
Stanniocalcin 2 (STC2) is a fish protein that controls body Ca2+ and phosphate metabolism. STC2 has also been described in mammals, and as platelet function highly depends on both extracellular and intracellular Ca2+, we have explored its expression and function in these cells. STC2−/− mice exhibit shorter tail bleeding time than WT mice. Platelets from STC2-deficient mice showed enhanced aggregation, as well as enhanced Ca2+ mobilization in response to the physiological agonist thrombin (Thr) and the diacylglycerol analog, OAG, a selective activator of the non-capacitative Ca2+ entry channels. Interestingly, platelets from STC2−/− mice exhibit attenuated interaction between STIM1 and Orai1 in response to Thr, thus suggesting that STC2 is required for Thr-evoked STIM1-Orai1 interaction and the subsequent store-operated Ca2+ entry (SOCE). We have further assessed possible changes in the expression of the most relevant channels involved in non-capacitative Ca2+ entry in platelets. Then, protein expression of Orai3, TRPC3 and TRPC6 were evaluated by Western blotting, and the results revealed that while the expression of Orai3 was enhanced in the STC2-deficient mice, others like TRPC3 and TRPC6 remains almost unaltered. Summarizing, our results provide for the first time evidence for a role of STC2 in platelet physiology through the regulation of agonist-induced Ca2+ entry, which might be mediated by the regulation of Orai3 channel expression.
Collapse
Affiliation(s)
- Esther López
- Department of Physiology (PHYCELL) of the Veterinary Faculty, University of Extremadura, Cáceres, Spain
| | - L Gómez-Gordo
- Department of Animal Medicine, Veterinary Faculty University of Extremadura, Cáceres, Spain
| | - Carlos Cantonero
- Department of Physiology (PHYCELL) of the Veterinary Faculty, University of Extremadura, Cáceres, Spain
| | - Nuria Bermejo
- Hematology Unit, San Pedro de Alcantara Hospital, Cáceres, Spain
| | - Jorge Pérez-Gómez
- Faculty of Sport Sciences, University of Extremadura, Cáceres, Spain
| | - María P Granados
- Aldea Moret Health Center, Extremadura Health Service, Cáceres, Spain
| | - Gines M Salido
- Institute of Molecular Pathology Biomarkers, Cáceres, Spain
| | - Juan A Rosado Dionisio
- Department of Physiology (PHYCELL) of the Veterinary Faculty, University of Extremadura, Cáceres, Spain
| | - Pedro C Redondo Liberal
- Department of Physiology (PHYCELL) of the Veterinary Faculty, University of Extremadura, Cáceres, Spain
| |
Collapse
|
17
|
Kania E, Roest G, Vervliet T, Parys JB, Bultynck G. IP 3 Receptor-Mediated Calcium Signaling and Its Role in Autophagy in Cancer. Front Oncol 2017; 7:140. [PMID: 28725634 PMCID: PMC5497685 DOI: 10.3389/fonc.2017.00140] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/19/2017] [Indexed: 01/09/2023] Open
Abstract
Calcium ions (Ca2+) play a complex role in orchestrating diverse cellular processes, including cell death and survival. To trigger signaling cascades, intracellular Ca2+ is shuffled between the cytoplasm and the major Ca2+ stores, the endoplasmic reticulum (ER), the mitochondria, and the lysosomes. A key role in the control of Ca2+ signals is attributed to the inositol 1,4,5-trisphosphate (IP3) receptors (IP3Rs), the main Ca2+-release channels in the ER. IP3Rs can transfer Ca2+ to the mitochondria, thereby not only stimulating core metabolic pathways but also increasing apoptosis sensitivity and inhibiting basal autophagy. On the other hand, IP3-induced Ca2+ release enhances autophagy flux by providing cytosolic Ca2+ required to execute autophagy upon various cellular stresses, including nutrient starvation, chemical mechanistic target of rapamycin inhibition, or drug treatment. Similarly, IP3Rs are able to amplify Ca2+ signals from the lysosomes and, therefore, impact autophagic flux in response to lysosomal channels activation. Furthermore, indirect modulation of Ca2+ release through IP3Rs may also be achieved by controlling the sarco/endoplasmic reticulum Ca2+ ATPases Ca2+ pumps of the ER. Considering the complex role of autophagy in cancer development and progression as well as in response to anticancer therapies, it becomes clear that it is important to fully understand the role of the IP3R and its cellular context in this disease. In cancer cells addicted to ER–mitochondrial Ca2+ fueling, IP3R inhibition leads to cancer cell death via mechanisms involving enhanced autophagy or mitotic catastrophe. Moreover, IP3Rs are the targets of several oncogenes and tumor suppressors and the functional loss of these genes, as occurring in many cancer types, can result in modified Ca2+ transport to the mitochondria and in modulation of the level of autophagic flux. Similarly, IP3R-mediated upregulation of autophagy can protect some cancer cells against natural killer cells-induced killing. The involvement of IP3Rs in the regulation of both autophagy and apoptosis, therefore, directly impact cancer cell biology and contribute to the molecular basis of tumor pathology.
Collapse
Affiliation(s)
- Elzbieta Kania
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kankerinstituut, KU Leuven, Leuven, Belgium
| | - Gemma Roest
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kankerinstituut, KU Leuven, Leuven, Belgium
| | - Tim Vervliet
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kankerinstituut, KU Leuven, Leuven, Belgium
| | - Jan B Parys
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kankerinstituut, KU Leuven, Leuven, Belgium
| | - Geert Bultynck
- Laboratory for Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kankerinstituut, KU Leuven, Leuven, Belgium
| |
Collapse
|
18
|
Ivanova H, Kerkhofs M, La Rovere RM, Bultynck G. Endoplasmic Reticulum-Mitochondrial Ca 2+ Fluxes Underlying Cancer Cell Survival. Front Oncol 2017; 7:70. [PMID: 28516062 PMCID: PMC5413502 DOI: 10.3389/fonc.2017.00070] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/28/2017] [Indexed: 11/17/2022] Open
Abstract
Calcium ions (Ca2+) are crucial, ubiquitous, intracellular second messengers required for functional mitochondrial metabolism during uncontrolled proliferation of cancer cells. The mitochondria and the endoplasmic reticulum (ER) are connected via “mitochondria-associated ER membranes” (MAMs) where ER–mitochondria Ca2+ transfer occurs, impacting the mitochondrial biology related to several aspects of cellular survival, autophagy, metabolism, cell death sensitivity, and metastasis, all cancer hallmarks. Cancer cells appear addicted to these constitutive ER–mitochondrial Ca2+ fluxes for their survival, since they drive the tricarboxylic acid cycle and the production of mitochondrial substrates needed for nucleoside synthesis and proper cell cycle progression. In addition to this, the mitochondrial Ca2+ uniporter and mitochondrial Ca2+ have been linked to hypoxia-inducible factor 1α signaling, enabling metastasis and invasion processes, but they can also contribute to cellular senescence induced by oncogenes and replication. Finally, proper ER–mitochondrial Ca2+ transfer seems to be a key event in the cell death response of cancer cells exposed to chemotherapeutics. In this review, we discuss the emerging role of ER–mitochondrial Ca2+ fluxes underlying these cancer-related features.
Collapse
Affiliation(s)
- Hristina Ivanova
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kanker Instituut (LKI), KU Leuven, Leuven, Belgium
| | - Martijn Kerkhofs
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kanker Instituut (LKI), KU Leuven, Leuven, Belgium
| | - Rita M La Rovere
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kanker Instituut (LKI), KU Leuven, Leuven, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine, Leuven Kanker Instituut (LKI), KU Leuven, Leuven, Belgium
| |
Collapse
|