ijms-logo

Journal Browser

Journal Browser

STIMulating Ca2+ Homeostasis

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Biochemistry".

Deadline for manuscript submissions: closed (30 November 2020) | Viewed by 32270

Special Issue Editors


E-Mail Website
Guest Editor
JKU Life Science Center, Johannes Kepler University Linz, Linz, Austria
Interests: calcium; CRAC; STIM; orai; Ca2+-dependent transcription; cancer; intracellular signalling
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Johannes Kepler University Linz, Linz, Austria
Interests: Calcium, SOCE, CRAC, structure - function of STIM + Orai
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

We cordially invite you to submit experimental papers, review articles, or commentaries for an upcoming issue of IJMS, which Marc and I are supporting as guest editors.

There is no doubt, essential cellular processes are crucially dependent on correct Ca2+ signaling through Ca2+ release-activated Ca2+ (CRAC) channels. In the past decade, important advances have been made in the characterization of the CRAC channel key players, STIM1 and Orai1; their impact on human diseases; and accessory proteins supporting the Ca2+ signaling cascade. Much effort has gone into understanding the processes underlying physiological Ca2+ signaling.

This Issue of IJMS (“STIMulating Ca2+ Homeostasis”) will cover a selection of novel research and review articles in the field of Ca2+ signaling through CRAC channels. It will focus on Ca2+ homeostasis within cells; store-operated Ca2+ entry; CRAC channel proteins; STIM/Orai proteins including their isoforms, their structure/activation/interaction, and gating, accessory proteins; diseases related to altered Ca2+ homeostasis; MD simulations on Ca2+ signaling proteins; and the use of CRISPR/Cas9 techniques in this field.

Best regards,

Dr. Irene Frischauf
Dr. Marc Fahrner
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • STIM
  • Orai
  • CRAC
  • Ca2+ signalling
  • structure–function
  • Ca2+-related diseases
  • MD simulation

Published Papers (9 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

23 pages, 3810 KiB  
Article
Synthesis and Characterization of Store-Operated Calcium Entry Inhibitors Active in the Submicromolar Range
by Camille Le Guilcher, Tomas Luyten, Jan B. Parys, Mathieu Pucheault and Olivier Dellis
Int. J. Mol. Sci. 2020, 21(24), 9777; https://doi.org/10.3390/ijms21249777 - 21 Dec 2020
Cited by 2 | Viewed by 2750
Abstract
The store-operated calcium entry, better known as SOCE, forms the main Ca2+ influx pathway in non-excitable cells, especially in leukocytes, where it is required for cell activation and the immune response. During the past decades, several inhibitors were developed, but they lack [...] Read more.
The store-operated calcium entry, better known as SOCE, forms the main Ca2+ influx pathway in non-excitable cells, especially in leukocytes, where it is required for cell activation and the immune response. During the past decades, several inhibitors were developed, but they lack specificity or efficacy. From the non-specific SOCE inhibitor 2-aminoethyl diphenylborinate (2-APB), we synthetized 16 new analogues by replacing/modifying the phenyl groups. Among them, our compound P11 showed the best inhibitory capacity with a Ki ≈ 75 nM. Furthermore, below 1 µM, P11 was devoid of any inhibitory activity on the two other main cellular targets of 2-APB, the IP3 receptors, and the SERCA pumps. Interestingly, Jurkat T cells secrete interleukin-2 under phytohemagglutinin stimulation but undergo cell death and stop IL-2 synthesis when stimulated in the presence of increasing P11 concentrations. Thus, P11 could represent the first member of a new and potent family of immunosuppressors. Full article
(This article belongs to the Special Issue STIMulating Ca2+ Homeostasis)
Show Figures

Graphical abstract

25 pages, 4234 KiB  
Article
PGRMC1 Inhibits Progesterone-Evoked Proliferation and Ca2+ Entry Via STIM2 in MDA-MB-231 Cells
by Carlos Cantonero, Ginés M. Salido, Juan A. Rosado and Pedro C. Redondo
Int. J. Mol. Sci. 2020, 21(20), 7641; https://doi.org/10.3390/ijms21207641 - 15 Oct 2020
Cited by 14 | Viewed by 3072
Abstract
Progesterone receptor membrane component 1 (PGRMC1) has been shown to regulate some cancer hallmarks. Progesterone (P4) evokes intracellular calcium (Ca2+) changes in the triple-negative breast cancer cell lines (MDA-MB-231, MDA-MB-468, and BT-20) and in other breast cancer cell lines [...] Read more.
Progesterone receptor membrane component 1 (PGRMC1) has been shown to regulate some cancer hallmarks. Progesterone (P4) evokes intracellular calcium (Ca2+) changes in the triple-negative breast cancer cell lines (MDA-MB-231, MDA-MB-468, and BT-20) and in other breast cancer cell lines like the luminal MCF7 cells. PGRMC1 expression is elevated in MDA-MB-231 and MCF7 cells as compared to non-tumoral MCF10A cell line, and PGRMC1 silencing enhances P4-evoked Ca2+ mobilization. Here, we found a new P4-dependent Ca2+ mobilization pathway in MDA-MB-231 cells and other triple-negative breast cancer cells, as well as in MCF7 cells that involved Stromal interaction molecule 2 (STIM2), Calcium release-activated calcium channel protein 1 (Orai1), and Transient Receptor Potential Channel 1 (TRPC1). Stromal interaction molecule 1 (STIM1) was not involved in this novel Ca2+ pathway, as evidenced by using siRNA STIM1. PGRMC1 silencing reduced the negative effect of P4 on cell proliferation and cell death in MDA-MB-231 cells. In line with the latter observation, Nuclear Factor of Activated T-Cells 1 (NFAT1) nuclear accumulation due to P4 incubation for 48 h was enhanced in cells transfected with the small hairpin siRNA against PGRMC1 (shPGRMC1). These results provide evidence for a novel P4-evoked Ca2+ entry pathway that is downregulated by PGRMC1. Full article
(This article belongs to the Special Issue STIMulating Ca2+ Homeostasis)
Show Figures

Graphical abstract

28 pages, 5651 KiB  
Article
Synthesis and Pharmacological Characterization of 2-Aminoethyl Diphenylborinate (2-APB) Derivatives for Inhibition of Store-Operated Calcium Entry (SOCE) in MDA-MB-231 Breast Cancer Cells
by Achille Schild, Rajesh Bhardwaj, Nicolas Wenger, Dominic Tscherrig, Palanivel Kandasamy, Jan Dernič, Roland Baur, Christine Peinelt, Matthias A. Hediger and Martin Lochner
Int. J. Mol. Sci. 2020, 21(16), 5604; https://doi.org/10.3390/ijms21165604 - 5 Aug 2020
Cited by 19 | Viewed by 4424
Abstract
Calcium ions regulate a wide array of physiological functions including cell differentiation, proliferation, muscle contraction, neurotransmission, and fertilization. The endoplasmic reticulum (ER) is the major intracellular Ca2+ store and cellular events that induce ER store depletion (e.g., activation of inositol 1,4,5-triphosphate (IP [...] Read more.
Calcium ions regulate a wide array of physiological functions including cell differentiation, proliferation, muscle contraction, neurotransmission, and fertilization. The endoplasmic reticulum (ER) is the major intracellular Ca2+ store and cellular events that induce ER store depletion (e.g., activation of inositol 1,4,5-triphosphate (IP3) receptors) trigger a refilling process known as store-operated calcium entry (SOCE). It requires the intricate interaction between the Ca2+ sensing stromal interaction molecules (STIM) located in the ER membrane and the channel forming Orai proteins in the plasma membrane (PM). The resulting active STIM/Orai complexes form highly selective Ca2+ channels that facilitate a measurable Ca2+ influx into the cytosol followed by successive refilling of the ER by the sarcoplasmic/endoplasmic reticulum calcium ATPase (SERCA). STIM and Orai have attracted significant therapeutic interest, as enhanced SOCE has been associated with several cancers, and mutations in STIM and Orai have been linked to immunodeficiency, autoimmune, and muscular diseases. 2-Aminoethyl diphenylborinate (2-APB) is a known modulator and depending on its concentration can inhibit or enhance SOCE. We have synthesized several novel derivatives of 2-APB, introducing halogen and other small substituents systematically on each position of one of the phenyl rings. Using a fluorometric imaging plate reader (FLIPR) Tetra-based calcium imaging assay we have studied how these structural changes of 2-APB affect the SOCE modulation activity at different compound concentrations in MDA-MB-231 breast cancer cells. We have discovered 2-APB derivatives that block SOCE at low concentrations, at which 2-APB usually enhances SOCE. Full article
(This article belongs to the Special Issue STIMulating Ca2+ Homeostasis)
Show Figures

Figure 1

15 pages, 3209 KiB  
Article
Luminal STIM1 Mutants that Cause Tubular Aggregate Myopathy Promote Autophagic Processes
by Matthias Sallinger, Adéla Tiffner, Tony Schmidt, Daniel Bonhenry, Linda Waldherr, Irene Frischauf, Victoria Lunz, Isabella Derler, Romana Schober and Rainer Schindl
Int. J. Mol. Sci. 2020, 21(12), 4410; https://doi.org/10.3390/ijms21124410 - 21 Jun 2020
Cited by 21 | Viewed by 2961
Abstract
Stromal interaction molecule 1 (STIM1) is a ubiquitously expressed Ca2+ sensor protein that induces permeation of Orai Ca2+ channels upon endoplasmic reticulum Ca2+-store depletion. A drop in luminal Ca2+ causes partial unfolding of the N-terminal STIM1 domains and [...] Read more.
Stromal interaction molecule 1 (STIM1) is a ubiquitously expressed Ca2+ sensor protein that induces permeation of Orai Ca2+ channels upon endoplasmic reticulum Ca2+-store depletion. A drop in luminal Ca2+ causes partial unfolding of the N-terminal STIM1 domains and thus initial STIM1 activation. We compared the STIM1 structure upon Ca2+ depletion from our molecular dynamics (MD) simulations with a recent 2D NMR structure. Simulation- and structure-based results showed unfolding of two α-helices in the canonical and in the non-canonical EF-hand. Further, we structurally and functionally evaluated mutations in the non-canonical EF-hand that have been shown to cause tubular aggregate myopathy. We found these mutations to cause full constitutive activation of Ca2+-release-activated Ca2+ currents (ICRAC) and to promote autophagic processes. Specifically, heterologously expressed STIM1 mutations in the non-canonical EF-hand promoted translocation of the autophagy transcription factors microphthalmia-associated transcription factor (MITF) and transcription factor EB (TFEB) into the nucleus. These STIM1 mutations additionally stimulated an enhanced production of autophagosomes. In summary, mutations in STIM1 that cause structural unfolding promoted Ca2+ down-stream activation of autophagic processes. Full article
(This article belongs to the Special Issue STIMulating Ca2+ Homeostasis)
Show Figures

Figure 1

13 pages, 2325 KiB  
Article
The Number and Position of Orai3 Units within Heteromeric Store-Operated Ca2+ Channels Alter the Pharmacology of ICRAC
by Sven Kappel, Tatiana Kilch, Roland Baur, Martin Lochner and Christine Peinelt
Int. J. Mol. Sci. 2020, 21(7), 2458; https://doi.org/10.3390/ijms21072458 - 2 Apr 2020
Cited by 12 | Viewed by 2624
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. [...] Read more.
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. Full article
(This article belongs to the Special Issue STIMulating Ca2+ Homeostasis)
Show Figures

Figure 1

Review

Jump to: Research

34 pages, 5634 KiB  
Review
The Orai Pore Opening Mechanism
by Adéla Tiffner, Lena Maltan, Sarah Weiß and Isabella Derler
Int. J. Mol. Sci. 2021, 22(2), 533; https://doi.org/10.3390/ijms22020533 - 7 Jan 2021
Cited by 16 | Viewed by 3481
Abstract
Cell survival and normal cell function require a highly coordinated and precise regulation of basal cytosolic Ca2+ concentrations. The primary source of Ca2+ entry into the cell is mediated by the Ca2+ release-activated Ca2+ (CRAC) channel. Its action is [...] Read more.
Cell survival and normal cell function require a highly coordinated and precise regulation of basal cytosolic Ca2+ concentrations. The primary source of Ca2+ entry into the cell is mediated by the Ca2+ release-activated Ca2+ (CRAC) channel. Its action is stimulated in response to internal Ca2+ store depletion. The fundamental constituents of CRAC channels are the Ca2+ sensor, stromal interaction molecule 1 (STIM1) anchored in the endoplasmic reticulum, and a highly Ca2+-selective pore-forming subunit Orai1 in the plasma membrane. The precise nature of the Orai1 pore opening is currently a topic of intensive research. This review describes how Orai1 gating checkpoints in the middle and cytosolic extended transmembrane regions act together in a concerted manner to ensure an opening-permissive Orai1 channel conformation. In this context, we highlight the effects of the currently known multitude of Orai1 mutations, which led to the identification of a series of gating checkpoints and the determination of their role in diverse steps of the Orai1 activation cascade. The synergistic action of these gating checkpoints maintains an intact pore geometry, settles STIM1 coupling, and governs pore opening. We describe the current knowledge on Orai1 channel gating mechanisms and summarize still open questions of the STIM1–Orai1 machinery. Full article
(This article belongs to the Special Issue STIMulating Ca2+ Homeostasis)
Show Figures

Figure 1

29 pages, 1129 KiB  
Review
More Than Just Simple Interaction between STIM and Orai Proteins: CRAC Channel Function Enabled by a Network of Interactions with Regulatory Proteins
by Sascha Berlansky, Christina Humer, Matthias Sallinger and Irene Frischauf
Int. J. Mol. Sci. 2021, 22(1), 471; https://doi.org/10.3390/ijms22010471 - 5 Jan 2021
Cited by 20 | Viewed by 5483
Abstract
The calcium-release-activated calcium (CRAC) channel, activated by the release of Ca2+ from the endoplasmic reticulum (ER), is critical for Ca2+ homeostasis and active signal transduction in a plethora of cell types. Spurred by the long-sought decryption of the molecular nature of [...] Read more.
The calcium-release-activated calcium (CRAC) channel, activated by the release of Ca2+ from the endoplasmic reticulum (ER), is critical for Ca2+ homeostasis and active signal transduction in a plethora of cell types. Spurred by the long-sought decryption of the molecular nature of the CRAC channel, considerable scientific effort has been devoted to gaining insights into functional and structural mechanisms underlying this signalling cascade. Key players in CRAC channel function are the Stromal interaction molecule 1 (STIM1) and Orai1. STIM1 proteins span through the membrane of the ER, are competent in sensing luminal Ca2+ concentration, and in turn, are responsible for relaying the signal of Ca2+ store-depletion to pore-forming Orai1 proteins in the plasma membrane. A direct interaction of STIM1 and Orai1 allows for the re-entry of Ca2+ from the extracellular space. Although much is already known about the structure, function, and interaction of STIM1 and Orai1, there is growing evidence that CRAC under physiological conditions is dependent on additional proteins to function properly. Several auxiliary proteins have been shown to regulate CRAC channel activity by means of direct interactions with STIM1 and/or Orai1, promoting or hindering Ca2+ influx in a mechanistically diverse manner. Various proteins have also been identified to exert a modulatory role on the CRAC signalling cascade although inherently lacking an affinity for both STIM1 and Orai1. Apart from ubiquitously expressed representatives, a subset of such regulatory mechanisms seems to allow for a cell-type-specific control of CRAC channel function, considering the rather restricted expression patterns of the specific proteins. Given the high functional and clinical relevance of both generic and cell-type-specific interacting networks, the following review shall provide a comprehensive summary of regulators of the multilayered CRAC channel signalling cascade. It also includes proteins expressed in a narrow spectrum of cells and tissues that are often disregarded in other reviews of similar topics. Full article
(This article belongs to the Special Issue STIMulating Ca2+ Homeostasis)
Show Figures

Graphical abstract

18 pages, 1993 KiB  
Review
STIM Proteins: An Ever-Expanding Family
by Herwig Grabmayr, Christoph Romanin and Marc Fahrner
Int. J. Mol. Sci. 2021, 22(1), 378; https://doi.org/10.3390/ijms22010378 - 31 Dec 2020
Cited by 25 | Viewed by 3595
Abstract
Stromal interaction molecules (STIM) are a distinct class of ubiquitously expressed single-pass transmembrane proteins in the endoplasmic reticulum (ER) membrane. Together with Orai ion channels in the plasma membrane (PM), they form the molecular basis of the calcium release-activated calcium (CRAC) channel. An [...] Read more.
Stromal interaction molecules (STIM) are a distinct class of ubiquitously expressed single-pass transmembrane proteins in the endoplasmic reticulum (ER) membrane. Together with Orai ion channels in the plasma membrane (PM), they form the molecular basis of the calcium release-activated calcium (CRAC) channel. An intracellular signaling pathway known as store-operated calcium entry (SOCE) is critically dependent on the CRAC channel. The SOCE pathway is activated by the ligand-induced depletion of the ER calcium store. STIM proteins, acting as calcium sensors, subsequently sense this depletion and activate Orai ion channels via direct physical interaction to allow the influx of calcium ions for store refilling and downstream signaling processes. This review article is dedicated to the latest advances in the field of STIM proteins. New results of ongoing investigations based on the recently published functional data as well as structural data from nuclear magnetic resonance (NMR) spectroscopy and molecular dynamics (MD) simulations are reported and complemented with a discussion of the latest developments in the research of STIM protein isoforms and their differential functions in regulating SOCE. Full article
(This article belongs to the Special Issue STIMulating Ca2+ Homeostasis)
Show Figures

Figure 1

21 pages, 1319 KiB  
Review
Altered Ca2+ Homeostasis in Immune Cells during Aging: Role of Ion Channels
by Dorina Zöphel, Chantal Hof and Annette Lis
Int. J. Mol. Sci. 2021, 22(1), 110; https://doi.org/10.3390/ijms22010110 - 24 Dec 2020
Cited by 12 | Viewed by 3168
Abstract
Aging is an unstoppable process and begins shortly after birth. Each cell of the organism is affected by the irreversible process, not only with equal density but also at varying ages and with different speed. Therefore, aging can also be understood as an [...] Read more.
Aging is an unstoppable process and begins shortly after birth. Each cell of the organism is affected by the irreversible process, not only with equal density but also at varying ages and with different speed. Therefore, aging can also be understood as an adaptation to a continually changing cellular environment. One of these very prominent changes in age affects Ca2+ signaling. Especially immune cells highly rely on Ca2+-dependent processes and a strictly regulated Ca2+ homeostasis. The intricate patterns of impaired immune cell function may represent a deficit or compensatory mechanisms. Besides, altered immune function through Ca2+ signaling can profoundly affect the development of age-related disease. This review attempts to summarize changes in Ca2+ signaling due to channels and receptors in T cells and beyond in the context of aging. Full article
(This article belongs to the Special Issue STIMulating Ca2+ Homeostasis)
Show Figures

Figure 1

Back to TopTop