ArticleLiterature Review

Dziadek, M. A. & Johnstone, L. S. Biochemical properties and cellular localisation of STIM1 proteins. Cell Calcium 42, 123-132

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract

Human and murine STIM1 were originally discovered as candidate growth regulators in tumours and in the bone marrow stroma, and the structurally related vertebrate family members, STIM2 and the Drosophila homologue D-Stim, were subsequently identified. STIM proteins are ubiquitously expressed type I single-pass transmembrane proteins which have a unique combination of structural motifs within their polypeptide sequences. The extracellular regions contain an N-terminal unpaired EF-hand Ca(2+) binding motif adjacent to an unconventional glycosylated SAM domain, while the cytoplasmic regions contain alpha-helical coiled-coil domains within a region having homology to ERM domains adjacent to the transmembrane region, and phosphorylated proline-rich domains near the C-terminus. STIM1, STIM2 and D-Stim diverge significantly only in their structure C-terminal to the coiled-coil/ERM domains. The STIM structural domains were predicted to function in Ca(2+) binding as well as in mediating interactions between STIM proteins and other proteins, and homotypic STIM1-STIM1 and heterotypic STIM1-STIM2 interactions were demonstrated biochemically. However, the functional significance of the cellular localisation of STIM1 and its domain structure only became evident after recent breakthrough research identified STIM1 as a key regulator of store-operated calcium (SOC) entry into cells. It is now clear that STIM1 is both a sensor of Ca(2+) depletion in the endoplasmic reticulum (ER) lumen and an activator of Orai1-containing SOC channels in the plasma membrane. On the basis of recent functional studies a model can be proposed to explain how the biochemical properties of STIM1 contribute to its precise membrane localisation and its function in regulating SOC entry.

No full-text available

Request Full-text Paper PDF

To read the full-text of this research,
you can request a copy directly from the authors.

... STIM1 was initially described as a protein that induces growth arrest and degeneration of several human tumor cell lines, and is involved in stromal-hematopoietic cell interactions 1,2,3 . When the intraluminal Ca 2+ concentration is reduced, STIM1 [ 671 RKKFPLKIFKKPLKK 685 ] re-localizes within the ER membrane to punctae at ER-plasma membrane junctions, which facilitates its association with members of the Orai-1, E-cadherin, transient receptor potential cation (TRPC), and transient receptor vanilloid potential 4 (TRPV4) cation families 4,5,6,7 . All these proteins contain the acidic motifs (Asp-Asp) [4][5][6][7] . ...
... When the intraluminal Ca 2+ concentration is reduced, STIM1 [ 671 RKKFPLKIFKKPLKK 685 ] re-localizes within the ER membrane to punctae at ER-plasma membrane junctions, which facilitates its association with members of the Orai-1, E-cadherin, transient receptor potential cation (TRPC), and transient receptor vanilloid potential 4 (TRPV4) cation families 4,5,6,7 . All these proteins contain the acidic motifs (Asp-Asp) [4][5][6][7] . ...
... The renin-angiotensin system (RAS) is one of the most well investigated and clinically relevant homoeostatic systems in human physiology 8, 9,10 . As a bioactive peptide of RAS, Ang-(1-7) [angiotensin- (1)(2)(3)(4)(5)(6)(7)] is involved in many biological processes such as neural plasticity, memory and anxiety, and possesses anti-angiogenesis, anti-proliferation, anti-brosis, anti-hypertrophy, anti-thrombosis, and vasodilatation, properties by a direct interaction with its special receptor ACE2 (angiotensin converting enzyme 2) 11,12 . ...
Preprint
Full-text available
The property and signaling mechanisms of the angiotensin produced by ACE2 (angiotensin converting enzyme 2) have been well studied in the renin–angiotensin system. However, less attention has been attracted to the intracellular regulation of ACE2 protein. Using the information which [⁶⁷¹RKKFPLKIFKKPLKK⁶⁸⁵] motif of STIM1(stromal interaction molecule 1 precursor) is able to bind its partner proteins via two acidic amino acids (DD), we recognized that ACE2 contains the short acidic motif binding motif [⁷⁹⁹DD⁸⁰⁰] on its C-terminus. Moreover, we demonstrated that the protein interaction between STIM1 and ACE2 contributes to Ca²⁺ ion release from ER. We recognized that a novel mechanism of ACE2 regulation is modulated through interaction with STIM1, which binds directly to ACE2 as a COVID19 receptor. Further, our findings provide clues why ACE2 helps to release cytoplasmic Ca²⁺ from store-operated Ca²⁺ entry to stimulate viral coat membrane fusion with host plasma membrane, after COVID19 Spike protein binding to ACE2. Our finding also implicates that COVID19 infection enhances cytoplasmic Ca²⁺ ion concentration by releasing Ca²⁺ ion from ER, through its S protein association with its host ACE2 and subsequent STIM1 activation. Thus, our findings may provide the novel target point to prevent COVID19 pandemic recurring and cure it.
... The best examples are Huntington's (Bezprozvanny and Hayden 2004) and Alzheimer's diseases, where abnormal SOCE in neurons was reported by several groups (reviewed by Bojarski et al. 2008). Nonetheless, molecular mechanisms of SOCE in neurons are still not known, and there is little information in the literature about the presence of proteins, which might be involved in SOCE in neuronal cells (reviewed in Dziadek and Johnstone 2007). STIM1 (Stromal Interacting Molecule-1) is a novel regulator of SOCE (Liou et al. 2005. ...
... STIM1 (Stromal Interacting Molecule-1) is a novel regulator of SOCE (Liou et al. 2005. STIM1 is localized in the ER membrane where it serves as a Ca 2+ sensor via an EF-hand Ca 2+ binding site located in the lumen of the ER (Dziadek and Johnstone 2007). Upon Ca 2+ store depletion STIM1 redistributes into punctuate structures, moves closer to the plasma membrane and activates SOC channels (Liou et al. 2005. ...
... They diverge significantly within the C-terminal domain. STIM1 is expressed within the ER and at the cell surface, whereas STIM2 is expressed only in ER (Soboloff et al. 2006b, Dziadek andJohnstone 2007). We showed earlier that STIM1 is present in neurons and it undergoes intracellular translocation that is characteristic for CCE activation in non-excitable cells (Klejman et al. 2009). ...
... e l s e v i e r . c o m / l o c a t e / c e l l s i g C-terminal region [13][14][15]. The understanding of STIM1 regulatory mechanisms might provide important information concerning SOCE activation. ...
... STIM1 has been reported at two different cellular locations, either in the plasma-membrane, with the N-terminal domain facing the extracellular medium, or in the membrane of intracellular Ca 2+ stores, where the N-terminal domain is oriented towards the lumen of the stores [7,[13][14][15]26]. Therefore, we investigated which STIM1 pool was phosphorylated upon platelet stimulation with Tg (200 nM). ...
... Certain STIM1 post-translational modifications have been investigated, including glycosylation in the C-terminal domain and phosphorylation at Ser/Thr residues. For the latter, previous reports have claimed a possible regulatory mechanism in SOCE [13][14][15]38,39]. Here we report for the first time that STIM1 can be phosphorylated in Tyr residues upon Ca 2+ store depletion. ...
... The Orai protein family comprises three human Orai paralogs, namely, Orai1-3 ( Figure 3C) [121,160]. All three isoforms are ubiquitously expressed in many tissues (135,(161)(162)(163). RNA transcripts of Orai1 and Orai2 are found primarily in the spleen, lymph nodes, appendix, bone marrow, and brain, whereas Orai3 is detected more in the prostate, placenta, ovaries, testis, adrenal, urinary bladder, thyroid, endometrium, kidney, liver, and many other tissues [135,[161][162][163]. ...
... The Orai protein family comprises three human Orai paralogs, namely, Orai1-3 ( Figure 3C) [121,160]. All three isoforms are ubiquitously expressed in many tissues (135,(161)(162)(163). RNA transcripts of Orai1 and Orai2 are found primarily in the spleen, lymph nodes, appendix, bone marrow, and brain, whereas Orai3 is detected more in the prostate, placenta, ovaries, testis, adrenal, urinary bladder, thyroid, endometrium, kidney, liver, and many other tissues [135,[161][162][163]. Of note, the expression of Orai3 is restricted, because it is found only in mammals [164]. ...
Article
Full-text available
Simple Summary Cell fate is ultimately determined by the precisely coordinated action of the Ca²⁺-signaling machinery. During carcinogenesis, Ca²⁺ signaling is significantly remodeled due to mutations and/or ectopic expression. Here, we summarize current knowledge on how alterations in Ca²⁺ signaling contribute to the development of different cancer hallmarks. Emphasis is placed on the structure/function relationship of the well-studied store-operated Ca²⁺ channel, i.e., Orai1, and the Ca²⁺-activated K⁺ channel, i.e., SK3, alongside their individual and joint roles in cancer. This review lays out the current state of knowledge of Ca²⁺-signaling effectors and proteins as potential targets for the treatment of certain cancer types, with Orai1 and SK3 presented as emerging therapeutic targets. Abstract Cancer represents a major health burden worldwide. Several molecular targets have been discovered alongside treatments with positive clinical outcomes. However, the reoccurrence of cancer due to therapy resistance remains the primary cause of mortality. Endeavors in pinpointing new markers as molecular targets in cancer therapy are highly desired. The significance of the co-regulation of Ca²⁺-permeating and Ca²⁺-regulated ion channels in cancer cell development, proliferation, and migration make them promising molecular targets in cancer therapy. In particular, the co-regulation of the Orai1 and SK3 channels has been well-studied in breast and colon cancer cells, where it finally leads to an invasion-metastasis cascade. Nevertheless, many questions remain unanswered, such as which key molecular components determine and regulate their interplay. To provide a solid foundation for a better understanding of this ion channel co-regulation in cancer, we first shed light on the physiological role of Ca²⁺ and how this ion is linked to carcinogenesis. Then, we highlight the structure/function relationship of Orai1 and SK3, both individually and in concert, their role in the development of different types of cancer, and aspects that are not yet known in this context.
... The store-operated calcium (SOC) entry (SOCE) is one of the most ubiquitous pathways of calcium influx in mammalian cells, including neurons. SOCE physiologically occurs as a result of the InsP3R-mediated intracellular calcium store depletion and it is controlled by stromal interacting molecules (STIM1 and STIM2) -ER calcium sensors (Dziadek and Johnstone, 2007;Shalygin et al., 2015). The accumulated evidence indicates an important physiological role for neuronal SOCE both in normal and pathogenic conditions (Wegierski and Kuznicki, 2018). ...
... Currently, STIM proteins are established as well-known activators for SOC channels. In mammalians, this family is represented by two proteins: STIM1 and STIM2 (Dziadek and Johnstone, 2007). These proteins act as calcium sensors in the lumen of the ER and mediate the activity of SOC channels upon a loss of calcium binding. ...
Article
Full-text available
Huntington's disease (HD) is a severe autosomal-dominant neurodegenerative disorder caused by a mutation within a gene, encoding huntingtin protein. Here we have used the induced pluripotent stem cell technology to produce patient-specific terminally differentiated GABA-ergic medium spiny neurons modeling a juvenile form of HD (HD76). We have shown that calcium signaling is dramatically disturbed in HD76 neurons, specifically demonstrating higher levels of store-operated and voltage-gated calcium uptakes. However, comparing the HD76 neurons with the previously described low-repeat HD models, we have demonstrated that the severity of calcium signaling alterations does not depend on the length of the polyglutamine tract of the mutant huntingtin. Here we have also observed greater expression of huntingtin and an activator of store-operated calcium channels STIM2 in HD76 neurons. Since shRNA-mediated suppression of STIM2 decreased store-operated calcium uptake, we have speculated that high expression of STIM2 underlies the excessive entry through store-operated calcium channels in HD pathology. Moreover, a previously described potential anti-HD drug EVP4593 has been found to attenuate high levels of both huntingtin and STIM2 that may contribute to its neuroprotective effect. Our results are fully supportive in favor of the crucial role of calcium signaling deregulation in the HD pathogenesis and indicate that the cornerstone of excessive calcium uptake in HD-specific neurons is a calcium sensor and store-operated calcium channels activator STIM2, which should become a molecular target for medical treatment and novel neuroprotective drug development.
... Membranes 2020, 10, 425 4 of 61 STIM1 as well as STIM2 plays an indispensable role in the immune system and are expressed in diverse primary lymphocytes such as T h , T C , and B-cells [56,87]. Nevertheless, they have been detected to be widely spread in a variety of other tissues [82,[88][89][90][91]. High STIM1 levels have been reported for the heart, skeletal muscle, and the central nervous system [1,[92][93][94]. Meanwhile, there is clear evidence that STIM1 proteins occur in a variety of cancer types [92], such a breast, cervical, glioblastoma, and colorectal cancers [92,95]. ...
... Nevertheless, they have been detected to be widely spread in a variety of other tissues [82,[88][89][90][91]. High STIM1 levels have been reported for the heart, skeletal muscle, and the central nervous system [1,[92][93][94]. Meanwhile, there is clear evidence that STIM1 proteins occur in a variety of cancer types [92], such a breast, cervical, glioblastoma, and colorectal cancers [92,95]. ...
Article
Full-text available
Ca2+ ions play a variety of roles in the human body as well as within a single cell. Cellular Ca2+ signal transduction processes are governed by Ca2+ sensing and Ca2+ transporting proteins. In this review, we discuss the Ca2+ and the Ca2+-sensing ion channels with particular focus on the structure-function relationship of the Ca2+ release-activated Ca2+ (CRAC) ion channel, the Ca2+-activated K+ (KCa2+) ion channels, and their modulation via other cellular components. Moreover, we highlight their roles in healthy signaling processes as well as in disease with a special focus on cancer. As KCa2+ channels are activated via elevations of intracellular Ca2+ levels, we summarize the current knowledge on the action mechanisms of the interplay of CRAC and KCa2+ ion channels and their role in cancer cell development.
... Meyer et al. first identified STIM2 as a key actor in basal intracellular Ca 2+ level maintenance (Brandman et al. 2007). This transmembrane protein STIM2 is ubiquitously expressed among human and murine tissues to different degrees and shares 47 % homology with STIM1, the central player in the signaling pathway linking receptormediated release of ER Ca 2+ to SOCE (Dziadek and Johnstone 2007;Liou et al. 2005;Lopez et al. 2012). STIM2 is located at membranes of the ER and acidic stores and, in contrast to STIM1, cannot be detected on cell surface (Lopez et al. 2012;Soboloff et al. 2006). ...
... Since STIM1 and STIM2 are able to form heterooligomers (Dziadek and Johnstone 2007;Soboloff et al. 2006;Stathopulos et al. 2009;Williams et al. 2001), it is plausible that they function as a complex and that STIM2 requires STIM1 to signal to Orai1 channels for both B-SOC-and R-SOC-type Ca 2+ influx. However, STIM1 and STIM2 may also act synergistically only if both are activated following Ca 2+ stores being depleted by strong receptor stimuli, whereas STIM2 may function independently of STIM1 for basal ER Ca 2+ levels or weak receptor stimuli. ...
Article
Full-text available
Tight control of basal cytosolic Ca2+ concentration is essential for cell survival and to fine-tune Ca2+-dependent cell functions. A way to control this basal cytosolic Ca2+ concentration is to regulate membrane Ca2+ channels including store-operated Ca2+ channels and secondary messenger-operated channels linked to G-protein-coupled or tyrosine kinase receptor activation. Orai, with or without its reticular STIM partner and Transient Receptor Potential (TRP) proteins, were considered to be the main Ca2+ channels involved. It is well accepted that, in response to cell stimulation, opening of these Ca2+ channels contributes to Ca2+ entry and the transient increase in cytosolic Ca2+ concentration involved in intracellular signaling. However, in various experimental conditions, Ca2+ entry and/or Ca2+ currents can be recorded at rest, without application of any experimental stimulation. This led to the proposition that some plasma membrane Ca2+ channels are already open/activated in basal condition, contributing therefore to constitutive Ca2+ entry. This article focuses on direct and indirect observations supporting constitutive activity of channels belonging to the Orai and TRP families and on the mechanisms underlying their basal/constitutive activities.
... ER Ca 2+ depletion is coupled to an increase of Ca 2+ entry into the cell. STIM1 and STIM2 are plasma-membrane proteins with a luminal Ca 2+ sensor and interact with Orai1 proteins (108). These tetrameric Ca 2+ channels in the plasma membrane are then responsible for an increased Ca 2+ entry (108). ...
... STIM1 and STIM2 are plasma-membrane proteins with a luminal Ca 2+ sensor and interact with Orai1 proteins (108). These tetrameric Ca 2+ channels in the plasma membrane are then responsible for an increased Ca 2+ entry (108). Ca 2+ signaling is often abnormal in neurodegenerative diseases. ...
Article
Full-text available
Endoplasmic reticulum (ER) and mitochondrial function have both been shown to be critical events in neurodegenerative diseases. The ER mediates protein folding, maturation, sorting as well acts as calcium storage. The unfolded protein response (UPR) is a stress response of the ER that is activated by the accumulation of misfolded proteins within the ER lumen. Although the molecular mechanisms underlying ER stress-induced apoptosis are not completely understood, increasing evidence suggests that ER and mitochondria cooperate to signal cell death. Similarly, calcium-mediated mitochondrial function and dynamics not only contribute to ATP generation and calcium buffering but are also a linchpin in mediating cell fate. Mitochondria and ER form structural and functional networks (mitochondria-associated ER membranes [MAMs]) essential to maintaining cellular homeostasis and determining cell fate under various pathophysiological conditions. Regulated Ca2+ transfer from the ER to the mitochondria is important in maintaining control of pro-survival/pro-death pathways. In this review, we summarize the latest therapeutic strategies that target these essential organelles in the context of neurodegenerative diseases.
... The molecular players mediating SOCE include STIM1 (stromal interaction molecule 1), a transmembrane protein that can function as a calcium sensor in the endoplasmic ER lumen. Upon calcium store depletion, STIM1 is enriched and forms clusters in close proximity to the plasma membrane, where it interacts with and activates the SOC channels (Liou et al., 2005;Dziadek and Johnstone, 2007). ...
... The known ER-resident single-pass proteins STIM1 and STIM2 are homologous molecules that function as Ca 2+ sensors and activators of SOCE (Dziadek and Johnstone, 2007;Gruszczynska-Biegala et al., 2011). The Ca 2+ -binding domains of STIM proteins are identical, except for three residues, but STIM1 is twice as sensitive to Ca 2+ changes in the ER lumen, compared to STIM2 (Brandman et al., 2007), and is therefore regarded as a stronger activator of SOCE. ...
Article
Full-text available
It has been previously reported that N-terminus of mutant huntingtin (product of the 1st exon) is sufficient to cause a Huntington's disease (HD) pathological phenotype. In view of recent data suggesting that improper regulation of store-operated calcium (SOC) channels is involved in neurodegenerative processes, we investigated influence of expression of the mutant huntingtin N-terminal fragment (Htt138Q-1exon) on SOC entry (SOCE) in mouse neuroblastoma cells (Neuro-2a) and in primary culture of medium spiny neurons (MSNs) isolated from mice. The results show that SOCE in these cells is enhanced upon lentiviral expression of the Htt138Q-1exon. Moreover, we demonstrated that RNAi-mediated knockdown of TRPC1, Orai1, or STIM1 proteins leads to dramatic reduction of abnormal SOCE in both Neuro-2a and MSNs, expressing Htt138Q-1exon. Thus, we concluded that abnormal SOCE in these cells is maintained by both TRPC1- and Orai1-containing channels and required STIM1 for its activation. Furthermore, EVP4593 compound previously tested as a potential anti-HD drug in a Drosophila screening system has proved to be capable of reducing SOCE to the normal level in MSNs expressing the Htt138Q-1exon.
... The depleted Ca 2+ stores then induce the opening of Ca 2+ release-activated Ca 2+ (CRAC) channels that permit sustained, store-operated Ca 2+ entry (SOCE) into the cell (Parekh & Putney, 2005;Peinelt et al. 2006;Lewis, 2007;Putney, 2007). Recent work has identified the main molecular players of SOCE connecting Ca 2+ release from stores to the opening of CRAC channels: the ER Ca 2+ sensors stromal cell-interaction molecule (STIM) 1, STIM2 and the Drosophila homologue D-STIM (Williams et al. 2001;Stathopulos et al. 2006;Dziadek & Johnstone, 2007) as well as the CRAC channels Drosophila protein D-Orai (or -CRACM) and its three mammalian homologues, Orai1, Orai2 and Orai3 (or CRACM1, CRACM2 and CRACM3) Prakriya et al. 2006;Vig et al. 2006b;Yeromin et al. 2006;Lis et al. 2007;Gwack et al. 2007). When STIM1 or STIM2 are overexpressed jointly with Orai proteins, they reconstitute large CRAC currents (Mercer et al. 2006;Lis et al. 2007;Parvez et al. 2008) and mutational analysis of Orai1 has revealed several key amino acids that determine the selectivity of CRAC currents Vig et al. 2006a;Yeromin et al. 2006). ...
... When STIM1 or STIM2 are overexpressed jointly with Orai proteins, they reconstitute large CRAC currents (Mercer et al. 2006;Lis et al. 2007;Parvez et al. 2008) and mutational analysis of Orai1 has revealed several key amino acids that determine the selectivity of CRAC currents Vig et al. 2006a;Yeromin et al. 2006). D-STIM, STIM1 and STIM2 are single pass transmembrane proteins in the ER membrane with paired amino-terminal EF-hands located in the luminal side of the ER (Williams et al. 2001;Roos et al. 2005;Dziadek & Johnstone, 2007;Yuan et al. 2009). Upon store depletion, STIM proteins translocate into junctional structures (puncta) close to the plasma membrane (Stathopulos et al. 2006;Wu et al. 2006;Xu et al. 2006;Smyth et al. 2008), where they may bind to and activate CRAC channels Luik et al. 2006;Wu et al. 2007;Lioudyno et al. 2008). ...
Article
Full-text available
Key points Stromal cell‐interaction molecule (STIM) 2 senses Ca ²⁺ levels in the endoplasmic reticulum and activates Ca ²⁺ channels in the plasma membrane upon store depletion. Here we report that STIM2 is preferentially activated by low agonist concentrations that cause mild reductions in endoplasmic reticulum Ca ²⁺ levels. This shows that store‐operated Ca ²⁺ entry is regulated through signal strength, with weak stimuli activating STIM2 and strong stimuli engaging STIM1. The results help us to understand how receptor activation enables differential modulation of Ca ²⁺ entry over a range of agonist concentrations and levels of store depletion. Abstract Agonist‐induced Ca ²⁺ oscillations in many cell types are triggered by Ca ²⁺ release from intracellular stores and driven by store‐operated Ca ²⁺ entry. Stromal cell‐interaction molecule (STIM) 1 and STIM2 serve as endoplasmic reticulum Ca ²⁺ sensors that, upon store depletion, activate Ca ²⁺ release‐activated Ca ²⁺ channels (Orai1–3, CRACM1–3) in the plasma membrane. However, their relative roles in agonist‐mediated Ca ²⁺ oscillations remain ambiguous. Here we report that while both STIM1 and STIM2 contribute to store‐refilling during Ca ²⁺ oscillations in mast cells (RBL), T cells (Jurkat) and human embryonic kidney (HEK293) cells, they do so dependent on the level of store depletion. Molecular silencing of STIM2 by siRNA or inhibition by G418 suppresses store‐operated Ca ²⁺ entry and agonist‐mediated Ca ²⁺ oscillations at low levels of store depletion, without interfering with STIM1‐mediated signals induced by full store depletion. Thus, STIM2 is preferentially activated by low‐level physiological agonist concentrations that cause mild reductions in endoplasmic reticulum Ca ²⁺ levels. We conclude that with increasing agonist concentrations, store‐operated Ca ²⁺ entry is mediated initially by endogenous STIM2 and incrementally by STIM1, enabling differential modulation of Ca ²⁺ entry over a range of agonist concentrations and levels of store depletion.
... Repletion of Ca2+ caused these aggregates to revert back to monomers, demonstrating the reversibility of this Ca2+-dependent conformational change. However, other reports indicate that STIM1–STIM1 and STIM1–STIM2 interactions do occur in Ca2+ store-replete cells because both endogenous and overexpressed STIM1–STIM1 and STIM1–STIM2 complexes can be readily co-immunoprecipitated from store-replete cell lysates [10], [18], [19], [30]. These results were obtained from in vitro experiments, and it is important to note that the conditions in in vitro experiments are not identical to those in living cells. ...
... We found that cells coexpressing VN173-ΔSAM and VC155-ΔSAM (SAM domains deletion mutants) still exhibited high BiFC efficiency. However, cells cotransfected with VN173-ΔC and VC155-ΔC (cytoplasmic coiled-coil/ERM domain deletion mutants) did not exhibit yellow fluorescence, suggesting that the cytoplasmic coiled-coil/ERM domain is necessary for STIM1 oligomerisation in cells in the resting state as has been suggested [17], [18], [30]. Our findings are different from those of Stathopulos, who reported that STIM1 exists only in a monomeric form in the resting state in vitro [15]. ...
Article
Full-text available
Store-operated Ca(2+) channels are a major Ca(2+) entry pathway in nonexcitable cells, which drive various essential cellular functions. Recently, STIM1 and Orai proteins have been identified as the major molecular components of the Ca(2+) release-activated Ca(2+) (CRAC) channel. As the key subunit of the CRAC channel, STIM1 is the ER Ca(2+) sensor and is essential for the recruitment and activation of Orai1. However, the mechanisms in transmission of information of STIM1 to Orai1 still need further investigation. Bimolecular fluorescence complementation (BiFC) is one of the most advanced and powerful tools for studying and visualising protein-protein interactions in living cells. We utilised BiFC and acceptor photobleaching fluorescence resonance energy transfer (FRET) experiments to visualise and determine the state of STIM1 in the living cells in resting state. Our results demonstrate that STIM1 exists in an oligomeric form in resting cells and that rather than the SAM motif, it is the C-terminus (residues 233-474) of STIM1 that is the key domain for the interaction between STIM1s. The STIM1 oligomers (BiFC-STIM1) and wild-type STIM1 colocalised and had a fibrillar distribution in resting conditions. Depletion of ER Ca(2+) stores induced BiFC-STIM1 distribution to become punctate, an effect that could be prevented or reversed by 2-APB. After depletion of the Ca(2+) stores, BiFC-STIM1 has the ability to form puncta that colocalise with wild-type STIM1 or Orai1 near the plasma membrane. Our data also indicate that the function of BiFC-STIM1 was not altered compared with that of wild-type STIM1.
... 16 Our group was one of the first to prove the existence of a SOCE-like process in neurons, which was based on STIM-Orai interactions. 17,18 STIM and Orai proteins are expressed in the human, murine, [18][19][20][21][22] and zebrafish 23 brains. STIM2 is expressed at levels that are comparable to STIM1 in the human brain and is the major isoform in the murine central nervous system, especially in the hippocampus. ...
Article
Stromal interaction molecules (STIMs) are endoplasmic reticulum-resident proteins that regulate Ca2+ homeostasis and signaling by store-operated calcium entry (SOCE). The different properties and functions of STIM1 and STIM2 have been described mostly based on work in vitro. STIM2 knockout mice do not survive until adulthood. Therefore, we generated and characterized stim2a and stim2b double-knockout zebrafish. The (stim2a;stim2b)-/- zebrafish did not have any apparent morphological phenotype. However, RNA sequencing revealed 1424 differentially expressed genes. One of the most upregulated genes was annexin A3a, which is a marker of activated microglia. This corresponded well to an increase in Neutral Red staining in the in vivo imaging of the (stim2a;stim2b)-/- zebrafish brain. The lack of Stim2 decreased zebrafish survival under low oxygen conditions. Behavioral tests, such as the visual-motor response test and dark-light preference test, indicated that (stim2a;stim2b)-/- larvae might have problems with vision. This was consistent with the downregulation of many genes that are related to light perception. The periodic acid-Schiff staining of retina sections from adult zebrafish revealed alterations of the stratum pigmentosum, suggesting the involvement of a Stim2-dependent process in visual perception. Altogether, these data reveal new functions for Stim2 in the nervous system.
... Dorsal horn and spinal cord neurons also express STIM1-2 proteins [145,146]. In the human brain, STIM1-2 are found in all regions [118,147], both in fetal and adult tissues [148], with a high STIM1 expression [136]. ...
Article
Full-text available
The endoplasmic reticulum (ER) is the major intracellular calcium (Ca²⁺) storage compartment in eukaryotic cells. In most instances, the mobilization of Ca²⁺ from this store is followed by a delayed and sustained uptake of Ca²⁺ through Ca²⁺-permeable channels of the cell surface named store-operated Ca²⁺ channels (SOCCs). This gives rise to a store-operated Ca²⁺ entry (SOCE) that has been thoroughly investigated in electrically non-excitable cells where it is the principal regulated Ca²⁺ entry pathway. The existence of this Ca²⁺ route in neurons has long been a matter of debate. However, a growing body of experimental evidence indicates that the recruitment of Ca²⁺ from neuronal ER Ca²⁺ stores generates a SOCE. The present review summarizes the main studies supporting the presence of a depletion-dependent Ca²⁺ entry in neurons. It also addresses the question of the molecular composition of neuronal SOCCs, their expression, pharmacological properties, as well as their physiological relevance.
... However, Stim2 function seems to depend on cell type since two groups showed that the reduction in Stim2 expression only affected SOCE without affecting basal [Ca 2+ ]i where the expression ratio of both Stim proteins seems to determine whether Stim2 controls SOCE, basal [Ca 2+ ]i or [Ca 2+ ]ER (Berna-Erro et al., 2009;Schuhmann et al., 2010). Furthermore, since Stim1 and Stim2 can form hetero-oligomers (Dziadek and Johnstone, 2007;Soboloff et al., 2006;Stathopulos et al., 2009), it is probable that they act as a complex and that Stim2 needs Stim1 to interact with Orai1 channel for both R-SOCE and B-SOCE. However, to act as a complex, both Stims need to be activated which is only possible after Ca 2+ stores depletions while Stim2 can act alone upon weak stimuli or for maintaining basal [Ca 2+ ]ER. ...
Thesis
Since approved by the FDA (Food and Drug Administration) in 1978, platinum doublet therapy remains the current gold standard chemotherapeutic treatment for non-small cell lung cancer (NSCLC). However, resistance to platinum salts evolves rapidly in patients with NSCLC and one of the major reasons behind this resistance is their proved ability to enrich CSC (cancer stem cell) population. Calcium is recognized as an activator of critical signaling pathways, including the ones responsible for resistance to apoptosis. Herein, the calcium channel Orai3 has been recently identified as an important element in the resistance to chemotherapy in breast cancer cells. Furthermore, Orai3 channel is overexpressed in NSCLC tissues, is correlated to a high tumor grade and constitutes a predictive marker of metastasis and survival in resectable NSCLC tumors. In-vitro, Orai3 channel controls cell proliferation in NSCLC cell lines in a calcium dependent manner, via the Akt pathway. In the present work, we investigated the role of Orai3 channel in resistance to Cisplatin in NSCLC and the signaling pathways associated. We found that Orai3 channel becomes overexpressed after treatment with Cisplatin in NSCLC tissues and cell lines. Clinically, Orai3 overexpression after chemotherapy was associated with partial or no tumor regression. In cell lines, Cisplatin treatment increased Orai3 expression in a time-dependent manner and this overexpression was accompanied by an enrichment of CSCs population demonstrated by CD133 staining and an overexpression of the CSCs markers Nanog, SOX-2 and Slug. Moreover, Orai3 silencing or the reduction of extracellular calcium concentration sensitized the cells to Cisplatin and leads to a drastic reduction in the expression of Nanog and SOX-2 and a partial reduction of Slug expression. Furthermore, Orai3 overexpression induced the overexpression of both markers Nanog and SOX-2. Interestingly, we remarked a change in the function of Orai3 upon the treatment with Cisplatin. In basal conditions, Orai3 was found to regulate basal calcium entry in control cells or cells transfected with an empty vector while upon the treatment with Cisplatin or when Orai3 is overexpressed via Orai3 vector transfection, it becomes involved in SOC entry. We also noticed that Orai3 functions as SOC channel in CSCs. Finally, we found that upon the inhibition of the signaling pathway Akt, the expression of the stemness markers didn’t increase and Cisplatin’s efficiency was enhanced. In a conclusion, Orai3 overexpression enables the channel to become a SOC channel and favors the calcium entry. Also, Orai3 channel, via the Akt pathway, promotes the enrichment of CSCs which are insensitive to Cisplatin. Taken together, our results show for the first time that Orai3 channel is able to induce chemoresistance in NSCLC cells by increasing the CSCs population. Our findings provide a new context in the understanding of NSCLC resistance to chemotherapy and present Orai3 as a promising biomarker which could help in the choice of chemotherapeutic drugs for patients with NSCLC
... The stromal interaction molecule 1 (STIM1) is a transmembrane protein of the endoplasmic reticulum (ER) that is important for bone homeostasis and maintenance by regulating Ca 2+ levels in osteoblasts, osteoclasts and osteocytes [30,31] and seems to be involved in chondrogenesis [32,33]. It is a sensor of Ca 2+ levels in the ER, maintains cellular Ca 2+ balance and supports Ca 2+ signaling by initiating the store-operated Ca 2+ entry process following store depletion [34,35]. The tight regulation of the intracellular calcium concentration is crucial for the actin cytoskeleton, and the fact that STIM-1 has been shown to directly interact with TSP-1 and COMP [36] makes it an attractive candidate that could provide a link between the actin cytoskeleton and the cartilage ECM. ...
Article
Full-text available
Osteoarthritis (OA) is a multifactorial disease which is characterized by a change in the homeostasis of the extracellular matrix (ECM). The ECM is essential for the function of the articular cartilage and plays an important role in cartilage mechanotransduction. To provide a better understanding of the interaction between the ECM and the actin cytoskeleton, we investigated the localization and expression of the Ca2+-dependent proteins cartilage oligomeric matrix protein (COMP), thrombospondin-1 (TSP-1), plastin 3 (PLS3) and stromal interaction molecule 1 (STIM1). We investigated 16 patients who suffered from varus knee OA and performed a topographical analysis of the cartilage from the medial and lateral compartment of the proximal tibial plateau. In a varus knee, OA is more pronounced in the medial compared to the lateral compartment as a result of an overloading due to the malalignment. We detected a location-dependent staining of PLS3 and STIM1 in the articular cartilage tissue. The staining intensity for both proteins correlated with the degree of cartilage degeneration. The staining intensity of TSP-1 was clearly reduced in the cartilage of the more affected medial compartment, an observation that was confirmed in cartilage extracts by immunoblotting. The total amount of COMP was unchanged; however, slight changes were detected in the localization of the protein. Our results provide novel information on alterations in OA cartilage suggesting that Ca2+-dependent mechanotransduction between the ECM and the actin cytoskeleton might play an essential role in the pathomechanism of OA.
... STIM proteins (in humans 1 and 2) are single-pass membrane proteins located in the ER membrane that, after ER Ca 2+ depletion, regulate the Orai channel (Hou et al., 2020). Structurally, all STIM1 monomers contain an N-terminal signal peptide, a canonical Ca 2+ -binding EF 1 hand, a non-Ca 2+binding EF 2 hand, and a sterile α-motif (SAM) domain in the ER luminal region (known as ER-SAM) (Dziadek and Johnstone, 2007;Schober et al., 2019). The C-terminus domain located in the cytosolic side of the ER membrane is characterized by the coiled-coil 1 (CC1) segments, a STIM-Orai-activating region (SOAR) or calcium release-activated calcium (CRAC) activation domain (CAD), and the motif S/TxIP which are the components of the Orai1 activation small fragment Stathopulos et al., 2013;Rathner et al., 2018; Figure 2). ...
Article
Full-text available
Membrane contact sites (MCS) are typically defined as areas of proximity between heterologous or homologous membranes characterized by specific proteins. The study of MCS is considered as an emergent field that shows how crucial organelle interactions are in cell physiology. MCS regulate a myriad of physiological processes such as apoptosis, calcium, and lipid signaling, just to name a few. The membranal interactions between the endoplasmic reticulum (ER)–mitochondria, the ER–plasma membrane, and the vesicular traffic have received special attention in recent years, particularly in cancer research, in which it has been proposed that MCS regulate tumor metabolism and fate, contributing to their progression. However, as the therapeutic or diagnostic potential of MCS has not been fully revisited, in this review, we provide recent information on MCS relevance on calcium and lipid signaling in cancer cells and on its role in tumor progression. We also describe some proteins associated with MCS, like CERT, STIM1, VDAC, and Orai, that impact on cancer progression and that could be a possible diagnostic marker. Overall, these information might contribute to the understanding of the complex biology of cancer cells.
... Ca 2+ overload. In total, STIM2.2 is 61 amino acids longer than STIM1 (Figure 1) [93]. It has also been reported that STIM2.2 forms heterodimers with STIM1, which increases the latter's recruitment to ER-PM junctions and facilitates its activation [94,95]. ...
Article
Full-text available
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.
... The Ca 2+ sensor STIM1 in the ER is one of the main players in SOCE machinery (Dziadek and Johnstone, 2007;Prakriya and Lewis, 2015;Putney, 2017). STIM1 protein was previously shown to be required for SOCE activation in SK-N-SH HTT138Q (Vigont et al., 2014). ...
Article
Full-text available
Huntington’s disease (HD) is a hereditary neurodegenerative disease that is caused by polyglutamine expansion within the huntingtin (HTT) gene. One of the cellular activities that is dysregulated in HD is store-operated calcium entry (SOCE), a process by which Ca2+ release from the endoplasmic reticulum (ER) induces Ca2+ influx from the extracellular space. HTT-associated protein-1 (HAP1) is a binding partner of HTT. The aim of the present study was to examine the role of HAP1A protein in regulating SOCE in YAC128 mice, a transgenic model of HD. After Ca2+ depletion from the ER by the activation of inositol-(1,4,5)triphosphate receptor type 1 (IP3R1), we detected an increase in the activity of SOC channels when HAP1 protein isoform HAP1A was overexpressed in medium spiny neurons (MSNs) from YAC128 mice. A decrease in the activity of SOC channels in YAC128 MSNs was observed when HAP1 protein was silenced. In YAC128 MSNs that overexpressed HAP1A, an increase in activity of IP3R1 was detected while the ionomycin-sensitive ER Ca2+ pool decreased. 6-Bromo-N-(2-phenylethyl)-2,3,4,9-tetrahydro-1H-carbazol-1-amine hydrochloride (C20H22BrClN2), identified in our previous studies as a SOCE inhibitor, restored the elevation of SOCE in YAC128 MSN cultures that overexpressed HAP1A. The IP3 sponge also restored the elevation of SOCE and increased the release of Ca2+ from the ER in YAC128 MSN cultures that overexpressed HAP1A. The overexpression of HAP1A in the human neuroblastoma cell line SK-N-SH (i.e., a cellular model of HD (SK-N-SH HTT138Q)) led to the appearance of a pool of constitutively active SOC channels and an increase in the expression of STIM2 protein. Our results showed that HAP1A causes the activation of SOC channels in HD models by affecting IP3R1 activity.
... This suggestion is in good agreement with the data that mutant huntingtin could potentiate the receptor for IP 3 and cause alterations in calcium content in the endoplasmic reticulum (Tang et al., 2003). Since STIM1 and STIM2 proteins regulating SOC channels are sensitive to calcium concentration in the lumen of endoplasmic reticulum (Dziadek and Johnstone, 2007), more complete store depletion can result in stronger activation of these proteins and, therefore, in increasing calcium entry through the SOC channels. It should also be noted that STIM1 and STIM2 have different affinity to calcium ions. ...
Article
Full-text available
Neurodegenerative pathologies are among the most serious and socially significant problems of modern medicine, along with cardiovascular and oncological diseases. Several attempts have been made to prevent neuronal death using novel drugs targeted to the cell calcium signaling machinery, but the lack of adequate models for screening markedly impairs the development of relevant drugs. A potential breakthrough in this field is offered by the models of hereditary neurodegenerative pathologies based on endogenous expression of mutant proteins in neurons differentiated from patient-specific induced pluripotent stem cells (iPSCs). Here, we study specific features of store-operated calcium entry (SOCE) using an iPSCs-based model of Huntington’s disease (HD) and analyze the pharmacological effects of a specific drug targeted to the calcium channels. We show that SOCE in gamma aminobutyric acid-ergic striatal medium spiny neurons (GABA MSNs) was mediated by currents through at least two different channel groups, ICRAC and ISOC. Both of these groups were upregulated in HD neurons compared with the wild-type neurons. Thapsigargin-induced intracellular calcium store depletion in GABA MSNs resulted in predominant activation of either ICRAC or ISOC. The potential anti-HD drug EVP4593, which was previously shown to have neuroprotective activity in different HD models, affected both ICRAC and ISOC.
... SOCE was first found to regulate a number of functions in non-excitable cells [13,14], but several studies have recently demonstrated its importance in excitable cells, including neurons [15][16][17][18][19][20]. STIMs and ORAIs are ubiquitously expressed throughout the human and murine brain [20][21][22][23][24]. STIM2, a more Ca 2+ -sensitive homologue [25], is expressed at levels comparable to STIM1 in the human brain and is the predominant isoform in the murine central nervous system. ...
Article
Full-text available
STIM1 is an endoplasmic reticulum calcium sensor that is involved in several processes in neurons, including store-operated calcium entry. STIM1 also inhibits voltage-gated calcium channels, such as Cav1.2 and Cav3.1, and is thus considered a multifunctional protein. The aim of this work was to investigate the ways in which transgenic neuronal overexpression of STIM1 in FVB/NJ mice affects animal behavior and the electrophysiological properties of neurons in acute hippocampal slices. We overexpressed STIM1 from the Thy1.2 promoter and verified neuronal expression by quantitative reverse-transcription polymerase chain reaction, Western blot, and immunohistochemistry. Mature primary hippocampal cultures expressed STIM1 but exhibited no changes in calcium homeostasis. Basal synaptic transmission efficiency and short-term plasticity were comparable in slices that were isolated from transgenic mice, similarly as the magnitude of long-term potentiation. However, long-term depression that was induced by the glutamate receptor 1/5 agonist (S)-3,5-dihydroxyphenylglycine was impaired in STIM1 slices. Interestingly, transgenic mice exhibited a decrease in anxiety-like behavior and improvements in contextual learning. In summary, our data indicate that STIM1 overexpression in neurons in the brain perturbs metabotropic glutamate receptor signaling, leading to impairments in long-term depression and alterations in animal behavior. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
... For intracellular stores replenishment by means of sarco/endoplasmatic reticulum Ca 2+ -ATPase (SERCA) pumps located in ER membrane, the store-operated Ca 2+ entry (SOCE) is required [183]. The Stromal interacting protein 1 (STIM1) is the chief component in SOCE, STIM1 represents the Ca sensor responsible for communicating the depleted state of intracellular Ca 2+ compartments to store-operated Ca 2+ channels, and found ubiquitously in ER [184]. Ultimately, congregated STIM1 activates members of the Orai family of store-operated Ca 2+ channels, which result in plasma membrane Ca 2+ release-activated Ca 2+ (CRAC) channels opening and Ca 2+ influx into the cell [185]. ...
Article
The mast cells are integral part of immune system and they have pleiotropic physiological functions in our body. Any type of abnormal stimuli causes the mast cells receptors to spur the otherwise innocuous mast cells to degranulate and release inflammatory mediators like histamine, cytokines, chemokines and prostaglandins. These mediators are involved in various diseases like allergy, asthma, mastocytosis, cardiovascular disorders, etc. Herein, we describe the receptors involved in degranulation of mast cells and are broadly divided into four categories: G-protein coupled receptors, ligand gated ion channels, immunoreceptors and pattern recognition receptors. Although, activation of pattern recognition receptors do not cause mast cell degranulation, but result in cytokines production. Degranulation itself is a complex process involving cascade of events like membrane fusion events and various proteins like VAMP, Syntaxins, DOCK5, SNAP-23, MARCKS. Furthermore, we described these mast cell receptors antagonists or agonists useful in treatment of myriad diseases. Like, omalizumab anti-IgE antibody is highly effective in asthma, allergic disorders treatment and recently mechanistic insight of IgE uncovered; matrix mettaloprotease inhibitor marimistat is under phase III trial for inflammation, muscular dystrophy diseases; ZPL-389 (H4 receptor antagonist) is in Phase 2a Clinical Trial for atopic dermatitis and psoriasis; JNJ3851868 an oral H4 receptor antagonist is in phase II clinical development for asthma, rheumatoid arthritis. Therefore, research is still in inchoate stage to uncover mast cell biology, mast cell receptors, their therapeutic role in myriad diseases.
... STIM1 is a single-pass transmembrane protein, which resides mainly in ER membrane. Until now several domains within the STIM1 structure, which are important for its function and for the interaction with other SOCIC components, have been identified ( Lee et al. 2009a;Dziadek and Johnstone 2007). The STIM1 N-terminal is facing the ER lumen and contains classical and hidden Ca 2+ binding EF-hands (67-96) and a sterile a-motif (SAM, 132-200). ...
Chapter
Full-text available
Store Operated Ca2+ Entry (SOCE), the main Ca2+ influx mechanism in non-excitable cells, is implicated in the immune response and has been reported to be affected in several pathologies including cancer. The basic molecular constituents of SOCE are Orai, the pore forming unit, and STIM, a multidomain protein with at least two principal functions: one is to sense the Ca2+ content inside the lumen of the endoplasmic reticulum(ER) and the second is to activate Orai channels upon depletion of the ER. The link between Ca2+ depletion inside the ER and Ca2+ influx from extracellular media is through a direct association of STIM and Orai, but for this to occur, both molecules have to interact and form clusters where ER and plasma membrane (PM) are intimately apposed. In recent years a great number of components have been identified as participants in SOCE regulation, including regions of plasma membrane enriched in cholesterol and sphingolipids, the so called lipid rafts, which recruit a complex platform of specialized microdomains, which cells use to regulate spatiotemporal Ca2+ signals.
... Invertebrates lack this adaptation; however, SOCE is present and invertebrate STIM participates in this process. [50][51][52][53][54] This is consonant with the retention of STIM1 function, although at a reduced level, in SHR-A3 lacking the same C-terminal domain absent in invertebrates. Our findings do not implicate the TRPC channel regulation in the control of renal water handling in the CD. ...
Article
Full-text available
Store-operated calcium entry (SOCE) is the mechanism by which extracellular signals elicit prolonged intracellular calcium elevation to drive changes in fundamental cellular processes. Here, we investigated the role of SOCE in the regulation of renal water reabsorption, using the inbred rat strain SHR-A3 as an animal model with disrupted SOCE. We found that SHR-A3, but not SHR-B2, have a novel truncating mutation in the gene encoding stromal interaction molecule 1 (STIM1), the endoplasmic reticulum calcium (Ca(2+)) sensor that triggers SOCE. Balance studies revealed increased urine volume, hypertonic plasma, polydipsia, and impaired urinary concentrating ability accompanied by elevated circulating arginine vasopressin (AVP) levels in SHR-A3 compared with SHR-B2. Isolated, split-open collecting ducts (CD) from SHR-A3 displayed decreased basal intracellular Ca(2+) levels and a major defect in SOCE. Consequently, AVP failed to induce the sustained intracellular Ca(2+) mobilization that requires SOCE in CD cells from SHR-A3. This effect decreased the abundance of aquaporin 2 and enhanced its intracellular retention, suggesting impaired sensitivity of the CD to AVP in SHR-A3. Stim1 knockdown in cultured mpkCCDc14 cells reduced SOCE and basal intracellular Ca(2+) levels and prevented AVP-induced translocation of aquaporin 2, further suggesting the effects in SHR-A3 result from the expression of truncated STIM1. Overall, these results identify a novel mechanism of nephrogenic diabetes insipidus and uncover a role of SOCE in renal water handling.
... STIM1 is a single-pass transmembrane protein, which resides mainly in ER membrane. Until now several domains within the STIM1 structure, which are important for its function and for the interaction with other SOCIC components, have been identified (Lee et al. 2009a;Dziadek and Johnstone 2007). The STIM1 N-terminal is facing the ER lumen and contains classical and hidden Ca 2+ binding EF-hands (67-96) and a sterile a-motif (SAM,. ...
Article
Full-text available
Store-operated calcium entry (SOCE) occurs at specialized regions where the endoplasmic reticulum and plasma membranes are closely apposed. Several molecules converge in these junctions to form a complex that spatiotemporally circumscribes SOCE signaling. We have named recently this complex as SOCIC (Store Operated Calcium Influx Complex). There is a growing list of SOCIC members, including the Ca2+ sensor and channel activator STIM1, the Orai and TRPC1 channels, SOCE regulators as CaM and CRACR2A, and SOCE-regulated proteins as SERCA and adenylyl cyclases. Considering that under physiological conditions Ca2+ entry is transient, SOCIC should be a dynamic structure that goes through assembly and disassembly cycles depending on cell requirements, and on the depleted state of intracellular Ca2+ stores. Moreover SOCIC seems to assembly at specialized regions of plasma membrane known as lipid rafts. In this chapter we discuss the evidence supporting the idea that SOCE occurs at microdomains and introduce the SOCIC components known so far. Then we illustrate some ideas on how this complex is assembled and disassembled. Finally we address the evidence of physiological and pathological implications of the microdomain organization of SOCE.
... Stim1 and Stim2, as well as Orai1 and Orai2, are widely expressed in rodent and human brains (Dziadek and Johnstone, 2007;Gross et al., 2007;Gwack et al., 2007;Berna-Erro et al., 2009;Gruszczynska-Biegala et al., 2011;Steinbeck et al., 2011;Hartmann et al., 2014). Stim1 and Stim2 proteins are uniformly distributed throughout mouse brain. ...
Article
Full-text available
Stim1 and Orai1 are ubiquitous proteins that have long been known to mediate Ca2+ release-activated Ca2+ (CRAC) current (ICRAC) and store-operated Ca2+ entry (SOCE) only in non-excitable cells. SOCE is activated following the depletion of the endogenous Ca2+ stores, which are mainly located within the endoplasmic reticulum (ER), to replete the intracellular Ca2+ reservoir and engage specific Ca2+-dependent processes, such as proliferation, migration, cytoskeletal remodelling, and gene expression. Their paralogues, Stim2, Orai2 and Orai3, support SOCE in heterologous expression systems, but their physiological role is still obscure. Ca2+ inflow in neurons has long been exclusively ascribed to voltage-operated and receptor-operated channels. Nevertheless, recent work has unveiled that Stim1-2 and Orai1-2, but not Orai3, proteins are also expressed and mediate SOCE in neurons. Herein, we survey current knowledge about the neuronal distribution of Stim and Orai proteins in rodent and human brains; we further discuss that Orai2 is the main pore-forming subunit of CRAC channels in central neurons, in which it may be activated by either Stim1 or Stim2 depending on species, brain region and physiological stimuli. We examine the functions regulated by SOCE in neurons, where this pathway is activated under resting conditions to refill the ER, control spinogenesis and regulate gene transcription. Besides, we highlighted the possibility that SOCE also controls neuronal excitation and regulate synaptic plasticity. Finally, we evaluate the involvement of Stim and Orai proteins in severe neurodegenerative and neurological disorders, such as Alzheimer’s Disease and epilepsy.
... Orai proteins are expressed in neuronal cells and are operational. Studies by [68] and our group showed that STIM1 is present in neuronal cells in the brain [69,70], together with Orai2 [71]. More information was subsequently obtained about the neuronal expression of STIM1, STIM2, and three Orai proteins and their involvement in SOCE, ...
Article
Full-text available
In this review we describe the present knowledge about store operated Ca2 + entry (SOCE) in neurons and the proteins involved in this process: STIM, as well as Orai and TRP channels. We address the issue of whether SOCE is used only to refill Ca2 + in the ER or whether Ca2 + that enters the neuronal cell during SOCE also performs signaling functions. We collected the data indicating that SOCE and its components participate in the important processes in neurons. This has implications for identifying new drug targets for the treatment of brain diseases. Evidence indicates that in neurodegenerative diseases Ca2 + homeostasis and SOCE components become dysregulated. Thus, different targets and strategies might be identified for the potential treatment of these diseases. This article is part of a Special Issue entitled: 13th European Symposium on Calcium.
... This suggests that the DPB compounds would be useful tools to selectively probe the actions of STIM2 in store-operated Ca 2+ signaling responses. However, in most cell systems both STIM1 and STIM2 are expressed and it would be unclear what effect DPBs would exert on heterologously expressed STIM1 and STIM2 which would include mixed STIM1/STIM2 aggregates [67] In the earlier study [33], it was suggested that in cells expressing STIM1, the action of 2-APB might be to prevent STIM1aggregation. However, we reveal here that the inhibitory action of DPB162 is observed on the cytosolic STIM1ct and SOAR fragments which do not undergo store-dependent aggregation. ...
Article
The coupling of ER Ca2+-sensing STIM proteins and PM Orai Ca2+ entry channels generates “store-operated” Ca2+ signals crucial in controlling responses in many cell types. The dimeric derivative of 2-aminoethoxydiphenyl borinate (2-APB), DPB162-AE, blocks functional coupling between STIM1 and Orai1 with an IC50 (200 nM) 100-fold lower than 2-APB. Unlike 2-APB, DPB162-AE does not affect L-type or TRPC channels or Ca2+ pumps at maximal STIM1-Orai1 blocking levels. DPB162-AE blocks STIM1-induced Orai1 or Orai2, but does not block Orai3 or STIM2-mediated effects. We narrowed the DPB162-AE site of action to the STIM-Orai activating region (SOAR) of STIM1. DPB162-AE does not prevent the SOAR-Orai1 interaction but potently blocks SOAR-mediated Orai1 channel activation, yet its action is not as an Orai1 channel pore blocker. Using the SOAR-F394H mutant which prevents both physical and functional coupling to Orai1, we reveal DPB162-AE rapidly restores SOAR-Orai binding but only slowly restores Orai1 channel-mediated Ca2+ entry. With the same SOAR mutant, 2-APB induces rapid physical and functional coupling to Orai1, but channel activation is transient. We infer that the actions of both 2-APB and DPB162-AE are directed toward the STIM1-Orai1 coupling interface. Compared to 2-APB, DPB162-AE is a much more potent and specific STIM1/Orai1 functional uncoupler. DPB162-AE provides an important pharmacological tool and a useful mechanistic probe for the function and coupling between STIM1 and Orai1 channels.
... It is particularly abundant in the cerebellum and the hippocampus (Klejman et al., 2009;Lein et al., 2007), both brain regions that are also enriched in group I mGluRs and TRPC channels (Lein et al., 2007). On the cellular level, expression of STIM1 is particularly strong in PNs (Dziadek and Johnstone, 2007;Klejman et al., 2009;Skibinska-Kijek et al., 2009;Stiber et al., 2008). STIM1 localizes primarily to the membrane of the ER (Liou et al., 2005;Roos et al., 2005), where its luminal N terminus functions as a Ca 2+ sensor (Williams et al., 2001(Williams et al., , 2002. ...
Article
In central mammalian neurons, activation of metabotropic glutamate receptor type1 (mGluR1) evokes a complex synaptic response consisting of IP3 receptor-dependent Ca(2+) release from internal Ca(2+) stores and a slow depolarizing potential involving TRPC3 channels. It is largely unclear how mGluR1 is linked to its downstream effectors. Here, we explored the role of stromal interaction molecule 1 (STIM1) in regulating neuronal Ca(2+) signaling and mGluR1-dependent synaptic transmission. By analyzing mouse cerebellar Purkinje neurons, we demonstrate that STIM1 is an essential regulator of the Ca(2+) level in neuronal endoplasmic reticulum Ca(2+) stores. Both mGluR1-dependent synaptic potentials and IP3 receptor-dependent Ca(2+) signals are strongly attenuated in the absence of STIM1. Furthermore, the Purkinje neuron-specific deletion of Stim1 causes impairments in cerebellar motor behavior. Together, our results demonstrate that in the mammalian nervous system STIM1 is a key regulator of intracellular Ca(2+) signaling, metabotropic glutamate receptor-dependent synaptic transmission, and motor coordination.
... The aggregation of STIM1 puncta upon SOCE activation that we observed was consistent with previous reports in non-neuronal cells, where resting cells displayed uniform distribution of STIM1 on endoplasmic reticulum (ER) membranes. In these cells, store depletion caused rapid STIM1 redistribution into punctate clusters within areas of the ER membrane located close to the plasma membrane Dziadek and Johnstone 2007). ...
Article
J. Neurochem. (2012) 122, 1155–1166. Coordinated calcium signalling is vital for neuronal growth cone function and axon pathfinding. Although store-operated calcium entry (SOCE) has been suggested to be an important source of calcium in growth cone navigation, the mechanisms that regulate calcium signalling, particularly the regulation of internal calcium stores within growth cones, are yet to be fully determined. Stromal Interaction Molecule 1 (STIM1) is a calcium-sensing protein localized in the endoplasmic reticulum membrane that interacts with Orai proteins in the plasma membrane to initiate SOCE and refilling of intracellular calcium stores. We hypothesize that STIM1- and Orai1/2-mediated SOCE are necessary for growth cone turning responses to extracellular guidance cues. We show that STIM1 and Orai reorganize into puncta upon store depletion and during growth cone turning with STIM1 localization biased towards the turning side (high calcium side) of the growth cone. Importantly, STIM1 knock-down perturbed growth cone turning responses to the guidance cues brain-derived neurotrophic factor and semaphorin-3a (Sema-3a), as well as abolishing Sema-3a-induced growth cone collapse. Furthermore, STIM1 knock-down abolished SOCE induced by brain-derived neurotrophic factor, but not Sema-3a. Our data suggest that STIM1 is essential for correct growth cone navigation, playing multiple roles in growth cone motility, including the activation of SOCE.
... A key component in SOCE is the stromal interacting protein 1 (STIM1; Roos et al., 2005), that represents the Ca 2+ sensor responsible for communicating the depleted state of intracellular Ca 2+ compartments to store-operated Ca 2+ channels. In quiescent cells with ER filled with Ca 2+ , STIM1 is distributed homogenously throughout ER (Dziadek and Johnstone, 2007) but relocates upon release of Ca 2+ from ER stores to distinct puncta on ER in close proximity to the plasma membrane ( Liou et al., 2005). Aggregated STIM1 activates members of the Orai family of store-operated Ca 2+ channels, resulting in opening of plasma membrane Ca 2+ release-activated Ca 2+ (CRAC) channels and Ca 2+ influx into the cell ( Prakriya et al., 2006). ...
Article
Full-text available
Mast cell activation mediated by the high affinity receptor for IgE (FcεRI) is a key event in allergic response and inflammation. Other receptors on mast cells, as c-Kit for stem cell factor and G protein-coupled receptors (GPCRs) synergistically enhance the FcεRI-mediated release of inflammatory mediators. Activation of various signaling pathways in mast cells results in changes in cell morphology, adhesion to substrate, exocytosis, and migration. Reorganization of cytoskeleton is pivotal in all these processes. Cytoskeletal proteins also play an important role in initial stages of FcεRI and other surface receptors induced triggering. Highly dynamic microtubules formed by αβ-tubulin dimers as well as microfilaments build up from polymerized actin are affected in activated cells by kinases/phosphatases, Rho GTPases and changes in concentration of cytosolic Ca²⁺. Also important are nucleation proteins; the γ-tubulin complexes in case of microtubules or Arp 2/3 complex with its nucleation promoting factors and formins in case of microfilaments. The dynamic nature of microtubules and microfilaments in activated cells depends on many associated/regulatory proteins. Changes in rigidity of activated mast cells reflect changes in intermediate filaments build up from vimentin. This review offers a critical appraisal of current knowledge on the role of cytoskeleton in mast cells signaling.
... When next took advantage of the fact that mutation of an aspartate residue to alanine (D76A) in the EF domain of STIM1 results in localization of the protein into punctae, which may then directly or indirectly trigger Ca 2+ influx [17,18]. The ERM domain, which mediates STIM1 aggregation and interaction with Orai and/or TRPC channels, is also required for SOCE activation [14,18,19]. Thus, we infected NRVMs with adenoviral constructs coding for constitutively active (D76A, Ca-STIM1) or dominant-negative (ΔERM, Dn-STIM1) STIM1 proteins. ...
Article
Alterations in intracellular Ca(2+) homeostasis are an important trigger of pathological cardiac remodeling; however, mechanisms governing context-dependent changes in Ca(2+) influx are poorly understood. Store-operated Ca(2+) entry (SOCE) is a major mechanism regulating Ca(2+) trafficking in numerous cell types, yet its prevalence in adult heart and possible role in physiology and disease are each unknown. The Ca(2+)-binding protein, stromal interaction molecule 1 (STIM1), is a Ca(2+) sensor in the sarcoplasmic reticulum (SR), capable of triggering SOCE by interacting with plasma membrane Ca(2+) channels. We report that SOCE is abundant and robust in neonatal cardiomyocytes; however, SOCE is absent from adult cardiomyocytes. Levels of STIM1 transcript and protein correlate with the amplitude of SOCE, and manipulation of STIM1 protein levels (via shRNA) or activity (via expression of constitutively active or dominant-negative mutants) reveals a critical role for STIM1 in activating SOCE in cardiac myocytes. In neonatal hearts a recently identified STIM1 splice variant (STIM1L) is predominant but diminishes with maturation, only to reemerge with agonist- or afterload-induced cardiac stress. To test for pathophysiological relevance, we evaluated both in vitro and in vivo models of cardiac hypertrophy, finding that STIM1 expression is re-activated by pathological stress to trigger significant SOCE-dependent Ca(2+) influx. STIM1 amplifies agonist-induced hypertrophy via activation of the calcineurin-NFAT pathway. Importantly, inhibition of STIM1 suppresses agonist-triggered hypertrophy, pointing to a requirement for SOCE in this remodeling response. Stress-triggered STIM1 re-expression, and consequent SOCE activation, are critical elements in the upstream, Ca(2+)-dependent control of pathological cardiac hypertrophy.
... STIM1 has a single-pass transmembrane domain and resides mainly in the ER membrane (21). Its N-terminal is facing the ER lumen and contains an EF-hand, which binds calcium with a low affinity (200-600 nM), that causes calcium to be bound to STIM1 when the ER is full (near 1 mM) and unbound when stores are depleted (22,23). In the ER-full state, STIM1 distributes homoge-neously at the ER-membrane, while under ER depletion STIM1 oligomerizes, forming what has been described as puncta (24)(25)(26). ...
Article
Full-text available
There is a specialized form of calcium influx that involves a close communication between endoplasmic reticulum and the channels at the plasma membrane. In one side store depletion activates channels known as store-operated channels (SOC), which are responsible of the well-studied store-operated calcium entry (SOCE). SOC comprises two different types of channels. Orai, which is exclusively activated by store depletion being the channel responsible of the calcium release-activated calcium current, and transient receptor potential canonical channel, which in contrast, is activated by store depletion only under specific conditions and carries nonselective cationic currents. On the other hand, it has been recently shown that store depletion also inhibits calcium channels. The first member identified, of what we named as store-inhibited channels (SIC), is the L-type voltage-gated calcium channel. Stores control both SOC and SIC by means of the multifunctional protein STIM1. The identification of SOC and SIC opens a new scenario for the role of store depletion in the modulation of different calcium entry pathways, which may satisfy different cellular processes.
... It is believed to function as the sensor of luminal Ca 2ϩ and probably transduces the store depletion signal to the plasma membrane through translocation within the ER into areas closely juxtaposed with the plasma membrane (7, 7-9, 36). As described above, the protein is expressed not only in the ER but also in the plasma membrane (23)(24)(25)(26)(27), although its role in this membrane is not yet clear. Within the ER membrane, STIM1 has a luminal facing Ca 2ϩ -sensing EF-hand motif. ...
Article
Full-text available
Store-operated channels (SOCs) mediate Ca2+ entry signals in response to endoplasmic reticulum (ER) Ca2+ depletion in most cells. STIM1 senses decreased ER luminal Ca2+ through its EF-hand Ca2+-binding motif and aggregates in near-plasma membrane (PM) ER junctions to activate PM Orai1, the functional SOC. STIM1 is also present in the PM, although its role there is unknown. STIM1-mediated coupling was examined using the stable EF20 HEK293 cell line expressing the STIM1-D76A/E87A EF-hand mutant (STIM1EF) deficient in Ca2+ binding. EF20 cells were viable despite constitutive Ca2+ entry, allowing study of SOC activation without depleting ER Ca2+. STIM1EF was exclusively in stable near-PM junctions, 3.5-fold larger than formed with STIM1WT. STIMEF-expressing cells had normal ER Ca2+ levels but substantially reduced ER Ca2+ leak. Expression of antiapoptotic Bcl-2 proteins (BCl-2, MCL-1, BCL-XL) were increased 2-fold in EF20 cells, probably reflecting survival of EF20 cells but not accounting for decreased ER Ca2+ leak. Surface biotinylation and streptavidin pull-down of cells expressing STIM1WT or STIM1EF revealed strong PM interactions of both proteins. Although surface expression of STIM1WT was clearly detectable, STIM1EF was undetectable at the cell surface. Thus, the Ca2+ binding-defective STIM1EF mutant exists exclusively in aggregates within near-PM junctions but, unlike STIM1WT, is not trafficked to the PM. Although not inserted in the PM, external application of a monoclonal anti-N-terminal STIM1 antibody blocked constitutive STIMEF-mediated Ca2+ entry, but only in cells expressing endogenous STIM1WT and not in DT40 STIM1 knock-out cells devoid of STIMWT. This suggests that PM-STIM1 may play a regulatory role in SOC activation.
... STIM1 and 2 reside in the endoplasmic reticulum where they function as sensors of endoplasmic reticulum Ca 2þ content. Both proteins have a Ca 2þ -binding EF-hand motif in the Nterminus, directed toward the lumen of the endoplasmic reticulum (Dziadek and Johnstone 2007). A drop in endoplasmic reticulum Ca 2þ content causes Ca 2þ to dissociate from STIM1. ...
Article
Full-text available
Calcium signaling results from a complex interplay between activation and inactivation of intracellular and extracellular calcium permeable channels. This complexity is obvious from the pattern of calcium signals observed with modest, physiological concentrations of calcium-mobilizing agonists, which typically present as sequential regenerative discharges of stored calcium, a process referred to as calcium oscillations. In this review, we discuss recent advances in understanding the underlying mechanism of calcium oscillations through the power of mathematical modeling. We also summarize recent findings on the role of calcium entry through store-operated channels in sustaining calcium oscillations and in the mechanism by which calcium oscillations couple to downstream effectors.
... The involvement of canonical transient receptor potential channels (Ong et al., 2007) or the TRPV1 channel (Liu et al., 2003) in storeregulated Ca 2ϩ influx has been proposed, but their participation in SOCE under the control of STIM1 is unlikely (DeHaven et al., 2009). Previous studies of neuronal tissues have identified both STIM1 and Orai1 in the brain, especially the cerebellum (Klejman et al., 2009), and have located STIM1 in the fetal peripheral nervous system, including the DRG (Dziadek and Johnstone, 2007;Gasperini et al., 2009). Our new data demonstrate the expression of both STIM1 and Orai1 at the transcript and protein levels in adult sensory neurons, thereby establishing that the molecular hardware for SOCE activity is present in these cells. ...
Article
Full-text available
Painful nerve injury disrupts levels of cytoplasmic and stored Ca(2+) in sensory neurons. Since influx of Ca(2+) may occur through store-operated Ca(2+) entry (SOCE) as well as voltage- and ligand-activated pathways, we sought confirmation of SOCE in sensory neurons from adult rats and examined whether dysfunction of SOCE is a possible pathogenic mechanism. Dorsal root ganglion neurons displayed a fall in resting cytoplasmic Ca(2+) concentration when bath Ca(2+) was withdrawn, and a subsequent elevation of cytoplasmic Ca(2+) concentration (40 ± 5 nm) when Ca(2+) was reintroduced, which was amplified by store depletion with thapsigargin (1 μm), and was significantly reduced by blockers of SOCE, but was unaffected by antagonists of voltage-gated membrane Ca(2+) channels. We identified the underlying inwardly rectifying Ca(2+)-dependent I(CRAC) (Ca(2+) release activated current), as well as a large thapsigargin-sensitive inward current activated by withdrawal of bath divalent cations, representing SOCE. Molecular components of SOCE, specifically STIM1 and Orai1, were confirmed in sensory neurons at both the transcript and protein levels. Axonal injury by spinal nerve ligation (SNL) elevated SOCE and I(CRAC). However, SOCE was comparable in injured and control neurons when stores were maximally depleted by thapsigargin, and STIM1 and Orai1 levels were not altered by SNL, showing that upregulation of SOCE after SNL is driven by store depletion. Blockade of SOCE increased neuronal excitability in control and injured neurons, whereas injured neurons showed particular dependence on SOCE for maintaining levels of cytoplasmic and stored Ca(2+), which indicates a compensatory role for SOCE after injury.
... Stromal interaction molecule 1 (STIM1) plays a critical role in activating the plasma membrane (PM)-localized SOCs of the ORAI family (2)(3)(4). Localized predominantly on the endoplasmic reticulum (ER), STIM1 spans the membrane with the N terminus confined to the ER lumen and the longer C terminus facing the cytosol (5). Control over STIM1 activation is mediated by ER Ca 2ϩ store depletion; dissociation of Ca 2ϩ from the N-terminal EF hand Ca 2ϩ binding domain induces oligomerization, which drives STIM1 to precise locations on the ER juxtaposed to the PM (6). ...
Article
Full-text available
Calcium (Ca(2+)) influx through the plasma membrane store-operated Ca(2+) channel ORAI1 is controlled by Ca(2+) sensors of the stromal interaction molecule (STIM) family. STIM1 responds to endoplasmic reticulum (ER) Ca(2+) store depletion by redistributing and activating ORAI1 from regions of the ER juxtaposed to the plasma membrane. Unlike STIM1, STIM2 can regulate ORAI1 in a store-dependent and store-independent manner, but the mechanism by which this is achieved is unknown. Here we find that STIM2 is translated from a highly conserved methionine residue and is directed to the ER by an incredibly long 101-amino acid signal peptide. We find that although the majority of the total STIM2 population resides on the ER membrane, a second population escapes ER targeting to accumulate as a full-length preprotein in the cytosol, signal peptide intact. Unlike STIM2, preSTIM2 localizes to the inner leaflet of the plasma membrane where it interacts with ORAI1 to regulate basal Ca(2+) concentration and Ca(2+)-dependent gene transcription in a store-independent manner. Furthermore, a third protein comprising a fragment of the STIM2 signal peptide is released from the ER membrane into the cytosol where it regulates gene transcription in a Ca(2+)-independent manner. This study establishes a new model for STIM2-mediated regulation of ORAI1 in which two distinct proteins, STIM2 and preSTIM2, control store-dependent and store-independent modes of ORAI1 activation.
... Within weeks of one another, two laboratories independently published the results of screens of limited siRNA libraries, both revealing the essential role of STIM proteins in store operated Ca 2+ entry [50,51]. STIM1, or stromal interacting molecule-1, had been known for some time as a plasma membrane and endoplasmic reticulum single spanning transmembrane protein [52]. Vertebrates make two STIMs, designated STIM1 and STIM2 while invertebrates have only one. ...
Article
Store-operated Ca(2+) entry is a process whereby the depletion of intracellular Ca(2+) stores signals the opening of plasma membrane Ca(2+) channels. It has long been thought that the main function of store-operated Ca(2+) entry was the replenishment of intracellular Ca(2+) stores following their discharge during intracellular Ca(2+) signaling. Recent results, however, suggest that the primary function of these channels may be to provide direct Ca(2+) signals to recipients localized to spatially restricted areas close to the sites of Ca(2+) entry in order to initiate specific signaling pathways.
Article
Coiled coils, consisting of at least two α-helices, have important roles in the regulation of transcription, cell differentiation, and cell growth. Peptides composed of d-amino acids (d-peptides) have received great attention for their potential in biomedical applications, because they give large diversity for the design of peptidyl drug and are more resistant to proteolytic digestion than l-peptides. However, the interactions between l-peptides/l-protein and d-peptides in the formation of complex are poorly understood. In this study, stereoisomer-specific peptides were constructed corresponding to regions of the basic-leucine-zipper domains of Jun and Fos proteins. basic-leucine-zipper domains consist of an N-terminal basic domain, which is responsible for DNA binding, and a C-terminal domain that enables homodimerization or heterodimerization via formation of a coiled-coil. By combining peptides with different stereochemistries, the d-l heterochiral Jun-Fos heterodimer formation induced DNA binding by the basic domains of Jun-Fos. Our study provides new insight into the interaction between l-peptide and d-peptide enantiomers for developing d-peptide materials and drugs. Copyright © 2017 European Peptide Society and John Wiley & Sons, Ltd.
Chapter
Oxidative stress has been implicated in a number of pathologies including ischemia–reperfusion. During ischemia–reperfusion, excess amounts of different types of reactive oxygen species accumulate and can cause tissue damage. In the coronary artery smooth muscle, one of the proteins readily affected by reactive oxygen species is the sarco/endoplasmic reticulum Ca2+ pump. This leads to an inability of cells to sequester Ca2+ into the sarco/endoplasmic reticulum and release it from this organelle for cell activation during signal transduction. Here, we present the hypothesis that the damage by reactive oxygen species to sarco/endoplasmic reticulum Ca2+ pump affects the Ca2+ refilling into the SER. The role of Na+–Ca2+ exchanger and store-depletion-dependent channels is discussed in this context.
Chapter
The junctions between the endoplasmic reticulum and the plasma membrane are essential platforms for the activation of store operated Ca2+ influx. These junctions have specific dimensions and are non-uniformly distributed in polarized cells. The mechanisms involved in the formation of the junctions are currently unknown. Some cell types display stationary junctions, whilst in other cells new junctions can form rapidly following the reduction of the Ca2+ concentration in the lumen of the endoplasmic reticulum. The proteins involved in the activation of the Ca2+ influx could be involved the formation of the junctions. The architecture and the dynamics of the junctions are important for the regulation of Ca2+ signaling cascade and its downstream events.
Article
Full-text available
We have shown that the expression of full-length mutated huntingtin in human neuroblastoma cells (SK-N-SH) leads to an abnormal increase in calcium entry through store-operated channels. In this paper, the expression of the N-terminal fragment of mutated huntingtin (Htt138Q-1exon) is shown to be enough to provide an actual model for Huntington's disease. We have shown that Htt138Q-1exon expression causes increased store-operated calcium entry, which is mediated by at least two types of channels in SK-N-SH cells with different reversal potentials. Calcium sensor, STIM1, is required for activation of store-operated calcium entry in these cells. The results provide grounds for considering the proteins responsible for the activation and maintenance of the store-operated calcium entry as promising targets for developing novel therapeutics for neurodegenerative diseases.
Article
Activation of glutamate receptors and followed increase of intracellular calcium concentration is a key pathological mechanism involved in secondary neuronal injury after traumatic brain injury (TBI). Stromal interaction molecule (STIM) proteins are considered to be important players in regulating neuronal Ca(2+) homeostasis under normal aging and pathological conditions. Here, we investigated the role of STIM1 in regulating metabotropic glutamate receptor 1 (mGluR1)-related Ca(2+) signaling and neuronal survival by using an in vitro traumatic neuronal injury (TNI) model. The expression of STIM1 was significantly increased at both mRNA and protein levels after TNI. Down-regulation of STIM1 by specific small interfere RNA significantly preserved neuronal viability, decreased lactate dehydrogenase release, and inhibited apoptotic cell death after traumatic injury. Moreover, knockdown of STIM1 significantly alleviated the mGluR1-related increase of cytoplasmic Ca(2+) levels after TNI. By analyzing Ca(2+) imaging in Ca(2+)-free conditions, we demonstrated that the mGluR1-dependent inositol trisphosphate receptor and/or ryanodine receptor-mediated Ca(2+) release from the endoplasmic reticulum after TNI is strongly attenuated in the absence of STIM1. Together, our results demonstrate that in the mammalian nervous system, STIM1 is a key regulator of mGluR1-dependent Ca(2+) signaling and knockdown of STIM1 might be an effective intervention target in TBI.
Article
Full-text available
Neuroglial cells are homeostatic neural cells. Generally, they are electrically non-excitable and their activation is associated with the generation of complex intracellular Ca(2+) signals that define the "Ca(2+) excitability" of glia. in mammalian glial cells the major source of Ca(2+) for this excitability is the lumen of the endoplasmic reticulum (ER), which is ultimately (re)filled from the extracellular space. This occurs via store-operated Ca(2+) entry (SoCE) which is supported by a specific signaling system connecting the ER with plasmalemmal Ca(2+) entry. Here, emptying of the ER Ca(2+) store is necessary and sufficient for the activation of SOCE, and without Ca(2+) influx via SOCE the ER store cannot be refilled. The molecular arrangements underlying SOCE are relatively complex and include plasmalemmal channels, ER Ca(2+) sensors, such as stromal interaction molecule, and possibly ER Ca(2+) pumps (of the SERCA type). There are at least two sets of plasmalemmal channels mediating SOCE, the Ca(2+)-release activated channels, Orai, and transient receptor potential (TRP) channels. The molecular identity of neuroglial SOCE has not been yet identified unequivocally. However, it seems that Orai is predominantly expressed in microglia, whereas astrocytes and oligodendrocytes rely more on TRP channels to produce SoCE. in physiological conditions the SoCE pathway is instrumental for the sustained phase of the Ca(2+) signal observed following stimulation of metabotropic receptors on glial cells.
Article
Calcium influx through plasma membrane store-operated Ca2+ (SOC) channels is triggered when the endoplasmic reticulum (ER) Ca2+ store is depleted — a homeostatic Ca2+ signalling mechanism that remained enigmatic for more than two decades. RNA-interference (RNAi) screening and molecular and cellular physiological analysis recently identified STIM1 as the mechanistic 'missing link' between the ER and the plasma membrane. STIM proteins sense the depletion of Ca2+ from the ER, oligomerize, translocate to junctions adjacent to the plasma membrane, organize Orai or TRPC (transient receptor potential cation) channels into clusters and open these channels to bring about SOC entry.
Article
Full-text available
Background: Endothelial progenitor cells (EPCs) may be recruited from bone marrow to sustain tumor vascularisation and promote the metastatic switch. Understanding the molecular mechanisms driving EPC proliferation and tubulogenesis could outline novel targets for alternative anti-angiogenic treatments. Store-operated Ca(2+) entry (SOCE), which is activated by a depletion of the intracellular Ca(2+) pool, regulates the growth of human EPCs, where is mediated by the interaction between the endoplasmic reticulum Ca(2+)-sensor, Stim1, and the plasmalemmal Ca(2+) channel, Orai1. As oncogenesis may be associated to the capability of tumor cells to grow independently on Ca(2+) influx, it is important to assess whether SOCE regulates EPC-dependent angiogenesis also in tumor patients. Methodology/principal findings: The present study employed Ca(2+) imaging, recombinant sub-membranal and mitochondrial aequorin, real-time polymerase chain reaction, gene silencing techniques and western blot analysis to investigate the expression and the role of SOCE in EPCs isolated from peripheral blood of patients affected by renal cellular carcinoma (RCC; RCC-EPCs) as compared to control EPCs (N-EPCs). SOCE, activated by either pharmacological (i.e. cyclopiazonic acid) or physiological (i.e. ATP) stimulation, was significantly higher in RCC-EPCs and was selectively sensitive to BTP-2, and to the trivalent cations, La(3+) and Gd(3+). Furthermore, 2-APB enhanced thapsigargin-evoked SOCE at low concentrations, whereas higher doses caused SOCE inhibition. Conversely, the anti-angiogenic drug, carboxyamidotriazole (CAI), blocked both SOCE and the intracellular Ca(2+) release. SOCE was associated to the over-expression of Orai1, Stim1, and transient receptor potential channel 1 (TRPC1) at both mRNA and protein level The intracellular Ca(2+) buffer, BAPTA, BTP-2, and CAI inhibited RCC-EPC proliferation and tubulogenesis. The genetic suppression of Stim1, Orai1, and TRPC1 blocked CPA-evoked SOCE in RCC-EPCs. Conclusions: SOCE is remodelled in EPCs from RCC patients and stands out as a novel molecular target to interfere with RCC vascularisation due to its ability to control proliferation and tubulogenesis.
Chapter
Definition of the DomainIntroduction and Historical PerspectivesStructure and Dynamics of the ERER ProteomeER: A Multifunctional OrganelleConclusion AcknowledgmentsAbbreviationsReferences
Article
The possible role of STIM1 protein in the regulation of activity of receptor- and store-operated Ca2+ channels in non-excitable cells has been studied. Receptor- and store-operated Ca2+ influxes have been measured using the fluorescent method of detection of cytosolic Ca2+ concentration and the electrophysiological methods of whole-cell and single-channel current recordings in the control HEK293 cells and in HEK293 cells with suppressed expression of STIM1. The experiments have shown that STIM1 suppression results in a reduction of the amplitudes of both receptor- and store-operated inward calcium currents. The decrease of total Ca2+ influx of in response to an agonist or to passive depletion of calcium stores upon STIM1 suppression was due to the decrease or total absence of the activity of high-conductance channels Imax and non-selective channels Ins in HEK293 cells. A decrease in the STIM1 amount also altered the activity regulation of low-conductance Imin channels that changed from exclusively agonist-operated into store-dependent channels in HEK293 cells. Key wordsSTIM1-receptor-operated influx-store-operated influx-calcium-HEK293
Article
Full-text available
Activation of mast cells by aggregation of the high-affinity IgE receptors (FcεRI) initiates signaling events leading to the release of inflammatory and allergic mediators stored in cytoplasmic granules. A key role in this process play changes in concentrations of intracellular Ca(2+) controlled by store-operated Ca(2+) entry (SOCE). Although microtubules are also involved in the process leading to degranulation, the molecular mechanisms that control microtubule rearrangement during activation are largely unknown. In this study, we report that activation of bone marrow-derived mast cells (BMMCs) induced by FcεRI aggregation or treatment with pervanadate or thapsigargin results in generation of protrusions containing microtubules (microtubule protrusions). Formation of these protrusions depended on the influx of extracellular Ca(2+). Changes in cytosolic Ca(2+)concentration also affected microtubule plus-end dynamics detected by microtubule plus-end tracking protein EB1. Experiments with knockdown or reexpression of STIM1, the key regulator of SOCE, confirmed the important role of STIM1 in the formation of microtubule protrusions. Although STIM1 in activated cells formed puncta associated with microtubules in protrusions, relocation of STIM1 to a close proximity of cell membrane was independent of growing microtubules. In accordance with the inhibition of Ag-induced Ca(2+) response and decreased formation of microtubule protrusions in BMMCs with reduced STIM1, the cells also exhibited impaired chemotactic response to Ag. We propose that rearrangement of microtubules in activated mast cells depends on STIM1-induced SOCE, and that Ca(2+) plays an important role in the formation of microtubule protrusions in BMMCs.
Article
Full-text available
Article
Full-text available
We have used specific inhibitors of oligosaccharide processing enzymes as probes to determine the involvement of oligosaccharide residues in the biosynthesis and function of insulin and insulin-like growth factor-I receptors. In a previous study (Duronio, V., Jacobs, S., and Cuatrecasas, P. (1986) J. Biol. Chem. 261, 970-975) swainsonine was used to inhibit mannosidase II, resulting in the production of receptors containing only hybrid-type oligosaccharides. These receptors had a slightly lower molecular weight and were much more sensitive to endoglycosidase H, but otherwise behaved identically to normal receptors. In this study, we used two compounds that inhibit oligosaccharide processing at earlier steps: (i) N-methyl-1-deoxynojirimycin (MedJN), which inhibits glucosidases I and II and yields glucosylated, high mannose oligosaccharides, and (ii) manno-1-deoxynojirimycin (MandJN), which inhibits mannosidase I and yields high mannose oligosaccharides. In the presence of MandJN, HepG2 cells synthesized receptors of lower molecular weight, which were cleaved into alpha and beta subunits and were able to bind hormone and autophosphorylate. These receptors were as sensitive to endoglycosidase H as receptors made in the presence of swainsonine. In the presence of MedJN, receptors of only slightly lower molecular weight than normal were synthesized and were shown to contain some glucosylated high mannose oligosaccharides. These receptors were able to bind hormone and retained hormone-sensitive autophosphorylation activity. In both cases, the incompletely processed receptors could be detected at the cell surface by cross-linking of iodinated hormone and susceptibility to trypsin digestion, although less receptor was present in cells treated with MedJN. Studies of receptor synthesis using pulse-chase labeling showed that the receptor precursors synthesized in the presence of MedJN were cleaved into alpha and beta subunits at a slower rate than normal receptors or those made in the presence of MandJN. Inhibition of oligosaccharide processing had no effect on the association of the receptor subunits into disulfide-linked oligomeric complexes.
Article
Full-text available
Our understanding of lympho-hematopoietic microenvironments is incomplete, and a new cloning strategy was developed to identify molecules that bind to B lineage lymphocyte precursors. A cell sorting procedure was used for initial enrichment of cDNAs from stromal cell mRNA that contained signal sequences and were therefore likely to encode transmembrane or secreted proteins. A second step involved expression of the library as soluble Ig fusion proteins. Finally, pools representing these proteins were screened for the ability to recognize pre-B cells. This approach resulted in the cloning of biglycan, syndecan 4, collagen type I, clusterin, matrix glycoprotein sc1, osteonectin, and one unknown molecule (designated SIM). The full-length cDNA of SIM revealed that it is a type I transmembrane protein, and its intracellular domain has weak homology with myosin heavy chain and related proteins. Staining of established cell lines and freshly isolated hematopoietic cells with the Ig fusion proteins revealed distinct patterns of reactivity and differential dependence on divalent cations. Biglycan-, sc1-, and SIM-Ig fusion proteins selectively increased interleukin 7-dependent proliferation of pre-B cells. Overexpression of the entire SIM protein affected the morphology of 293T cells, while expression of just the extracellular portion was without effect. Thus, a series of stromal cell surface molecules has been identified that interact with blood cell precursors. Three of them promoted the survival and/or proliferation of pre-B cells in culture, and all merit further study in relation to lympho-hematopoiesis.
Article
Full-text available
The expression of GOK, a gene recently identified at 11p15.5, was studied in breast cancer, rhabdomyosarcoma, and rhabdoid tumor cell lines. In these neoplasms, deletions at 11p15 and suppression of tumorigenicity induced by a normal human chromosome 11 were previously demonstrated. Whereas breast cancer cell lines express readily detectable levels of GOK mRNA, expression is absent in rhabdomyosarcoma and rhabdoid tumor cell lines. This is in contrast with the high expression of GOK in skeletal muscle, the normal tissue of origin of rhabdomyosarcomas, suggesting that down-regulation of GOK expression could be involved in tumor development. In agreement with this hypothesis, transfection of GOK cDNA into G401 derived from a rhabdoid tumor and RD cells derived from a rhabdomyosarcoma that do not express detectable levels of GOK mRNA, induced cell death. Because GOK expression is not compatible with growth of these tumor cells, these results support the hypothesis that loss of GOK expression plays a role in tumor establishment or progression and suggest that GOK may act as a recessive tumor suppressor gene in rhabdomyosarcomas and rhabdoid tumors. On the contrary, transfection of GOK cDNA into the breast cancer cell line HBL100 produced no detectable effects, indicating that the growth-suppressive effect of GOK in RD and G401 cells was specific. Because rhabdomyosarcomas have been observed in cases of Beckwith-Wiedemann syndrome, a genetic disorder linked to 11p15, a role of GOK in this disease cannot be excluded.
Article
Full-text available
Transmembrane glycoproteins with type 1 topology can be retrieved to the endoplasmic reticulum (ER) by a retrieval signal containing a di-lysine (KK) motif near the C terminus. To investigate the structural requirements for ER retrieval, we have constructed mutants of the simian immunodeficiency virus (SIV) envelope (Env) protein with cytoplasmic tails of different lengths and containing a KK motif at the -3 and -4 positions. Such proteins were found to be retained intracellularly when the signal was located 18 amino acids or more away from the membrane spanning domain. The retrieval signal was found to be functional even when placed at the distal end of the wild-type SIV Env protein with 164 amino acids in the cytoplasmic tail, as shown by the lack of proteolytic processing and lack of cell surface expression of the mutant proteins. However, proteins with a cytoplasmic tail length of 13 amino acids or less having the di-lysine motif at the -3 and -4 positions were not retrieved to the ER since they were found to be processed and transported to the cell surface. The surface-expressed proteins were found to be functional in inducing cell fusion, whereas the proteins retained intracellularly were defective in fusion activity. We also found that the KK motif introduced near an amphipathic helical region in the cytoplasmic tail was not functional. These results demonstrate that the ability of the KK motif to cause protein retrieval and retention in the endoplasmic reticulum depends on the length and structure of the cytoplasmic domain. The ER retrieval of the mutant proteins was found to correlate with increased intracellular binding to beta COP proteins.
Article
Full-text available
STIM1 (where STIM is stromal interaction molecule) is a candidate tumour suppressor gene that maps to human chromosome 11p15.5, a region implicated in a variety of cancers, particularly embryonal rhabdomyosarcoma. STIM1 codes for a transmembrane phosphoprotein whose structure is unrelated to that of any other known proteins. The precise pathway by which STIM1 regulates cell growth is not known. In the present study we screened gene databases for STIM1-related sequences, and have identified and characterized cDNA sequences representing a single gene in humans and other vertebrates, which we have called STIM2. We identified a single STIM homologue in Drosophila melanogaster (D-Stim) and Caenorhabditis elegans, but no homologues in yeast. STIM1, STIM2 and D-Stim have a conserved genomic organization, indicating that the vertebrate family of two STIM genes most probably arose from a single ancestral gene. The three STIM proteins each contain a single SAM (sterile alpha-motif) domain and an unpaired EF hand within the highly conserved extracellular region, and have coiled-coil domains that are conserved in structure and position within the cytoplasmic region. However, the STIM proteins diverge significantly within the C-terminal half of the cytoplasmic domain. Differential levels of phosphorylation appear to account for two molecular mass isoforms (105 and 115 kDa) of STIM2. We demonstrate by mutation analysis and protein sequencing that human STIM2 initiates translation exclusively from a non-AUG start site in vivo. STIM2 is expressed ubiquitously in cell lines, and co-precipitates with STIM1 from cell lysates. This association into oligomers in vivo indicates a possible functional interaction between STIM1 and STIM2. The structural similarities between STIM1, STIM2 and D-STIM suggest conserved biological functions.
Article
Full-text available
ERM (ezrin/radixin/moesin) proteins recognize the cytoplasmic domains of adhesion molecules in the formation of the membrane-associated cytoskeleton. Here we report the crystal structure of the radixin FERM (4.1 and ERM) domain complexed with the ICAM-2 cytoplasmic peptide. The non-polar region of the ICAM-2 peptide contains the RxxTYxVxxA sequence motif to form a beta-strand followed by a short 3(10)-helix. It binds the groove of the phosphotyrosine-binding (PTB)-like subdomain C mediated by a beta-beta association and several side-chain interactions. The binding mode of the ICAM-2 peptide to the FERM domain is distinct from that of the NPxY motif-containing peptide binding to the canonical PTB domain. Mutation analyses based on the crystal structure reveal the determinant elements of recognition and provide the first insights into the physical link between adhesion molecules and ERM proteins.
Article
Full-text available
Store-operated Ca2+ (SOC) channels regulate many cellular processes, but the underlying molecular components are not well defined. Using an RNA interference (RNAi)-based screen to identify genes that alter thapsigargin (TG)-dependent Ca2+ entry, we discovered a required and conserved role of Stim in SOC influx. RNAi-mediated knockdown of Stim in Drosophila S2 cells significantly reduced TG-dependent Ca2+ entry. Patch-clamp recording revealed nearly complete suppression of the Drosophila Ca2+ release-activated Ca2+ (CRAC) current that has biophysical characteristics similar to CRAC current in human T cells. Similarly, knockdown of the human homologue STIM1 significantly reduced CRAC channel activity in Jurkat T cells. RNAi-mediated knockdown of STIM1 inhibited TG- or agonist-dependent Ca2+ entry in HEK293 or SH-SY5Y cells. Conversely, overexpression of STIM1 in HEK293 cells modestly enhanced TG-induced Ca2+ entry. We propose that STIM1, a ubiquitously expressed protein that is conserved from Drosophila to mammalian cells, plays an essential role in SOC influx and may be a common component of SOC and CRAC channels.
Article
Full-text available
For thyroid hormone synthesis, thyroid peroxidase (TPO) molecules must be transported from the endoplasmic reticulum via the Golgi complex to be delivered at the cell surface to catalyze iodination of secreted thyroglobulin. Like other glycoproteins, TPO molecules in transit to the cell surface have the potential to acquire endoglycosidase H resistance as a consequence of Golgi-based modification of their N-linked carbohydrates, and measurement of the intracellular distribution of TPO has often relied on this assumption. To examine TPO surface distribution in thyrocyte cell lines, we prepared new antibodies against rat TPO. Antibody reactivity was first established upon expression of recombinant rat (r) TPO in 293 cells, which were heterogeneous for surface expression as determined by flow cytometry. By cell fractionation, surface rTPO fractionated distinctly from internal pools of TPO (that co-fractionate with calnexin), yet surface TPO molecules remained endoglycosidase H (endo H)-sensitive. Although the FRTL5 (and PC Cl3) rat thyrocyte cell line also exhibits almost no endo H-resistant TPO, much of the endogenous rTPO is localized to the cell surface by immunofluorescence. Similar results were obtained by fractionation of FRTL5 cell membranes on sucrose gradients. We conclude that in FRTL5 cells, a large fraction of rTPO is delivered to the plasma membrane yet does not acquire Golgi-type processing of its N-glycans. Rat and mouse thyroid tissue TPO also shows little or no endo H resistance, although cell fractionation still needs to be optimized for these tissues.
Article
Full-text available
Ca(2+) signaling in nonexcitable cells is typically initiated by receptor-triggered production of inositol-1,4,5-trisphosphate and the release of Ca(2+) from intracellular stores. An elusive signaling process senses the Ca(2+) store depletion and triggers the opening of plasma membrane Ca(2+) channels. The resulting sustained Ca(2+) signals are required for many physiological responses, such as T cell activation and differentiation. Here, we monitored receptor-triggered Ca(2+) signals in cells transfected with siRNAs against 2,304 human signaling proteins, and we identified two proteins required for Ca(2+)-store-depletion-mediated Ca(2+) influx, STIM1 and STIM2. These proteins have a single transmembrane region with a putative Ca(2+) binding domain in the lumen of the endoplasmic reticulum. Ca(2+) store depletion led to a rapid translocation of STIM1 into puncta that accumulated near the plasma membrane. Introducing a point mutation in the STIM1 Ca(2+) binding domain resulted in prelocalization of the protein in puncta, and this mutant failed to respond to store depletion. Our study suggests that STIM proteins function as Ca(2+) store sensors in the signaling pathway connecting Ca(2+) store depletion to Ca(2+) influx.
Article
Full-text available
As the sole Ca2+ entry mechanism in a variety of non-excitable cells, store-operated calcium (SOC) influx is important in Ca2+ signalling and many other cellular processes. A calcium-release-activated calcium (CRAC) channel in T lymphocytes is the best-characterized SOC influx channel and is essential to the immune response, sustained activity of CRAC channels being required for gene expression and proliferation. The molecular identity and the gating mechanism of SOC and CRAC channels have remained elusive. Previously we identified Stim and the mammalian homologue STIM1 as essential components of CRAC channel activation in Drosophila S2 cells and human T lymphocytes. Here we show that the expression of EF-hand mutants of Stim or STIM1 activates CRAC channels constitutively without changing Ca2+ store content. By immunofluorescence, EM localization and surface biotinylation we show that STIM1 migrates from endoplasmic-reticulum-like sites to the plasma membrane upon depletion of the Ca2+ store. We propose that STIM1 functions as the missing link between Ca2+ store depletion and SOC influx, serving as a Ca2+ sensor that translocates upon store depletion to the plasma membrane to activate CRAC channels.
Article
Full-text available
Focal adhesion kinase (FAK) is a non-receptor tyrosine kinase that localizes to focal adhesions in adherent cells. Through phosphorylation of proteins assembled at the cytoplasmic tails of integrins, FAK promotes signaling events that modulate cellular growth, survival, and migration. The amino-terminal region of FAK contains a region of sequence homology with band 4.1 and ezrin/radixin/moesin (ERM) proteins termed a FERM domain. FERM domains are found in a variety of signaling and cytoskeletal proteins and are thought to mediate intermolecular interactions with partner proteins and phospholipids at the plasma membrane and intramolecular regulatory interactions. Here we report two crystal structures of an NH2-terminal fragment of avian FAK containing the FERM domain and a portion of the regulatory linker that connects the FERM and kinase domains. The tertiary folds of the three subdomains (F1, F2, and F3) are similar to those of known FERM structures despite low sequence conservation. Differences in the sequence and relative orientation of the F3 subdomain alters the nature of the interdomain interface, and the phosphoinositide binding site found in ERM family FERM domains is not present in FAK. A putative protein interaction site on the F3 lobe is masked by the proximal region of the linker. Additionally, in one structure the adjacent Src SH3 and SH2 binding sites in the linker associate with the surfaces of the F3 and F1 lobes, respectively. These structural features suggest the possibility that protein interactions of the FAK FERM domain can be regulated by binding of Src kinases to the linker segment.
Article
Full-text available
The neurofibromatosis type2 (NF2) tumor suppressor gene product, merlin, is structurally related to the ezrin-radixin-moesin (ERM) family of proteins that anchor the actin cytoskeleton to specific membrane proteins and participate in cell signaling. However, the basis of the tumor suppressing activity of merlin is not well understood. Previously, we identified a role of merlin as an inhibitor of the Ras-ERK signaling pathway. Recent studies have suggested that phosphorylation of merlin, as of other ERM proteins, may regulate its function. To determine whether phosphorylation of merlin affects its suppression of Ras-ERK signaling, we generated plasmids expressing full-length merlin with substitutions of serine 518, a potential phosphorylation site. A substitution that mimics constitutive phosphorylation (S518D) abrogated the ability of merlin to suppress effects of the Ras-ERK signaling pathway such as Ras-induced SRE transactivation, Elk-mediated SRE transactivation, Ras-induced ERK phosphorylation and Ras-induced focus formation. On the other hand, an S518A mutant, which mimics nonphosphorylated merlin, acted like wild type merlin. These observations show that mimicking merlin phosphorylation impairs not only growth suppression by merlin but also its inhibitory action on the Ras-ERK signaling pathway.
Article
Full-text available
Receptor-induced Ca²⁺ signals are key to the function of all cells and involve release of Ca²⁺ from endoplasmic reticulum (ER) stores, triggering Ca²⁺ entry through plasma membrane (PM) “store-operated channels” (SOCs). The identity of SOCs and their coupling to store depletion remain molecular and mechanistic mysteries. The single transmembrane-spanning Ca²⁺-binding protein, STIM1, is necessary in this coupling process and is proposed to function as an ER Ca²⁺ sensor to provide the trigger for SOC activation. Here we reveal that, in addition to being an ER Ca²⁺ sensor, STIM1 functions within the PM to control operation of the Ca²⁺ entry channel itself. Increased expression levels of STIM1 correlate with a gain in function of Ca²⁺ release-activated Ca²⁺ (CRAC) channel activity. Point mutation of the N-terminal EF hand transforms the CRAC channel current (I CRAC) into a constitutively active, Ca²⁺ store-independent mode. Mutants in the EF hand and cytoplasmic C terminus of STIM1 alter operational parameters of CRAC channels, including pharmacological profile and inactivation properties. Last, Ab externally applied to the STIM1 N-terminal EF hand blocks both I CRAC in hematopoietic cells and SOC-mediated Ca²⁺ entry in HEK293 cells, revealing that STIM1 has an important functional presence within the PM. The results reveal that, in addition to being an ER Ca²⁺ sensor, STIM1 functions within the PM to exert control over the operation of SOCs. As a cell surface signaling protein, STIM1 represents a key pharmacological target to control fundamental Ca²⁺-regulated processes including secretion, contraction, metabolism, cell division, and apoptosis. • calcium signaling • calcium channel • patch-clamp • mast cells • T lymphocytes
Article
Full-text available
Depletion of intracellular calcium stores activates store-operated calcium entry across the plasma membrane in many cells. STIM1, the putative calcium sensor in the endoplasmic reticulum, and the calcium release-activated calcium (CRAC) modulator CRACM1 (also known as Orai1) in the plasma membrane have recently been shown to be essential for controlling the store-operated CRAC current (I(CRAC)). However, individual overexpression of either protein fails to significantly amplify I(CRAC). Here, we show that STIM1 and CRACM1 interact functionally. Overexpression of both proteins greatly potentiates I(CRAC), suggesting that STIM1 and CRACM1 mutually limit store-operated currents and that CRACM1 may be the long-sought CRAC channel.
Article
Full-text available
Recent studies by our group and others demonstrated a required and conserved role of Stim in store-operated Ca²⁺ influx and Ca²⁺ release-activated Ca²⁺ (CRAC) channel activity. By using an unbiased genome-wide RNA interference screen in Drosophila S2 cells, we now identify 75 hits that strongly inhibited Ca²⁺ influx upon store emptying by thapsigargin. Among these hits are 11 predicted transmembrane proteins, including Stim, and one, olf186-F, that upon RNA interference-mediated knockdown exhibited a profound reduction of thapsigargin-evoked Ca²⁺ entry and CRAC current, and upon overexpression a 3-fold augmentation of CRAC current. CRAC currents were further increased to 8-fold higher than control and developed more rapidly when olf186-F was cotransfected with Stim. olf186-F is a member of a highly conserved family of four-transmembrane spanning proteins with homologs from Caenorhabditis elegans to human. The endoplasmic reticulum (ER) Ca²⁺ pump sarco-/ER calcium ATPase (SERCA) and the single transmembrane-soluble N-ethylmaleimide-sensitive (NSF) attachment receptor (SNARE) protein Syntaxin5 also were required for CRAC channel activity, consistent with a signaling pathway in which Stim senses Ca²⁺ depletion within the ER, translocates to the plasma membrane, and interacts with olf186-F to trigger CRAC channel activity. • capacitative calcium entry (CCE) • genome-wide screen • CRAC channel • RNA interference • store-operated calcium (SOC) influx
Article
Full-text available
The two membrane proteins, STIM1 and Orai1, have each been shown to be essential for the activation of store-operated channels (SOC). Yet, how these proteins functionally interact is not known. Here, we reveal that STIM1 and Orai1 expressed together reconstitute functional SOCs. Expressed alone, Orai1 strongly reduces store-operated Ca2+ entry (SOCE) in human embryonic kidney 293 cells and the Ca2+ release-activated Ca2+ current (ICRAC) in rat basophilic leukemia cells. However, expressed along with the store-sensing STIM1 protein, Orai1 causes a massive increase in SOCE, enhancing the rate of Ca2+entry by up to 103-fold. This entry is entirely store-dependent since the same coexpression causes no measurable store-independent Ca2+ entry. The entry is completely blocked by the SOC blocker, 2-aminoethoxydiphenylborate. Orai1 and STIM1 coexpression also caused a large gain in CRAC channel function in rat basophilic leukemia cells. The close STIM1 homologue, STIM2, inhibited SOCE when expressed alone but coexpressed with Orai1 caused substantial constitutive (store-independent) Ca2+ entry. STIM proteins are known to mediate Ca2+ store-sensing and endoplasmic reticulum-plasma membrane coupling with no intrinsic channel properties. Our results revealing a powerful gain in SOC function dependent on the presence of both Orai1 and STIM1 strongly suggest that Orai1 contributes the PM channel component responsible for Ca2+ entry. The suppression of SOC function by Orai1 overexpression likely reflects a required stoichiometry between STIM1 and Orai1.
Article
Full-text available
The molecular nature of store-operated Ca2+-selective channels has remained an enigma, due largely to the continued inability to convincingly demonstrate Ca2+-selective store-operated currents resulting from exogenous expression of known genes. Recent findings have implicated two proteins, Stim1 and Orai1, as having essential roles in store-operated Ca2+ entry across the plasma membrane. However, transient overexpression of these proteins on their own results in little or no increase in store-operated entry. Here we demonstrate dramatic synergism between these two mediators; co-transfection of HEK293 cells with Stim1 and Orai1 results in an approximate 20-fold increase in store-operated Ca2+ entry and Ca2+-selective current. This demonstrates that these two proteins are limiting for both the signaling and permeation mechanisms for Ca2+-selective store-operated Ca2+ entry. There are three mammalian homologs of Orai1, and in expression experiments they all produced or augmented store-operated Ca2+ entry with efficacies in the order Orai1 > Orai2 > Orai3. Stim1 apparently initiates the signaling process by acting as a Ca2+ sensor in the endoplasmic reticulum. This results in rearrangement of Stim1 within the cell and migration toward the plasma membrane to regulate in some manner Orai1 located in the plasma membrane. However, we demonstrate that Stim1 does not incorporate in the surface membrane, and thus likely regulates or interacts with Orai1 at sites of close apposition between the plasma membrane and an intracellular Stim1-containing organelle.
Article
Full-text available
STIM1 (stromal interaction molecule 1) has recently been proposed to communicate the intracellular Ca2+ stores with the plasma membrane to mediate store-operated Ca2+ entry. Here we describe for the first time that Ca2+ store depletion stimulates rapid STIM1 surface expression and association with endogenously expressed human canonical TRP1 (hTRPC1) independently of rises in cytosolic free Ca2+ concentration. These events require the support of the actin cytoskeleton in human platelets, as reported for the coupling between type II inositol 1,4,5-trisphosphate receptor in the Ca2+ stores and hTRPC1 in the plasma membrane, which has been suggested to underlie the activation of store-operated Ca2+ entry in these cells. Electrotransjection of cells with anti-STIM1 antibody, directed toward the N-terminal sequence that includes the Ca2+-binding region, prevented the migration of STIM1 toward the plasma membrane, the interaction between STIM1 and hTRPC1, the coupling between hTRPC1 and type II inositol 1,4,5-trisphosphate receptor, and reduced store-operated Ca2+ entry. These findings provide evidence for a role of STIM1 in the activation of store-operated Ca2+ entry probably acting as a Ca2+ sensor.
Article
Full-text available
Recent RNA interference screens have identified several proteins that are essential for store-operated Ca2+ influx and Ca2+ release-activated Ca2+ (CRAC) channel activity in Drosophila and in mammals, including the transmembrane proteins Stim (stromal interaction molecule) and Orai. Stim probably functions as a sensor of luminal Ca2+ content and triggers activation of CRAC channels in the surface membrane after Ca2+ store depletion. Among three human homologues of Orai (also known as olf186-F), ORAI1 on chromosome 12 was found to be mutated in patients with severe combined immunodeficiency disease, and expression of wild-type Orai1 restored Ca2+ influx and CRAC channel activity in patient T cells. The overexpression of Stim and Orai together markedly increases CRAC current. However, it is not yet clear whether Stim or Orai actually forms the CRAC channel, or whether their expression simply limits CRAC channel activity mediated by a different channel-forming subunit. Here we show that interaction between wild-type Stim and Orai, assessed by co-immunoprecipitation, is greatly enhanced after treatment with thapsigargin to induce Ca2+ store depletion. By site-directed mutagenesis, we show that a point mutation from glutamate to aspartate at position 180 in the conserved S1-S2 loop of Orai transforms the ion selectivity properties of CRAC current from being Ca2+-selective with inward rectification to being selective for monovalent cations and outwardly rectifying. A charge-neutralizing mutation at the same position (glutamate to alanine) acts as a dominant-negative non-conducting subunit. Other charge-neutralizing mutants in the same loop express large inwardly rectifying CRAC current, and two of these exhibit reduced sensitivity to the channel blocker Gd3+. These results indicate that Orai itself forms the Ca2+-selectivity filter of the CRAC channel.
Article
Full-text available
Stromal interaction molecule 1 (STIM1) has recently been identified as a key player in store-operated Ca2+ entry. Endoplasmic reticulum (ER) luminal Ca2+ depletion results in STIM1 redistribution from ER membrane homogeneity to distinctly localized aggregates near the plasma membrane; these changes precede and are linked to cytoplasmic Ca2+ influx via Ca2+ release-activated channels (CRACs). The molecular mechanisms initiating ER STIM1 redistribution and plasma membrane CRAC activity are not well understood. We recombinantly expressed the Ca2+-sensing region of STIM1 consisting of the EF-hand together with the sterile alpha-motif (SAM) domain (EF-SAM) to investigate its Ca2+-related conformational and biochemical features. We demonstrate that Ca2+-loaded EF-SAM (holo) contains high alpha-helicity, whereas EF-SAM in the absence of Ca2+ (apo) is much less compact. Accordingly, the melting temperature (Tm) of the holoform is approximately 25 degrees C higher than apoform; heat and urea-derived thermodynamic parameters indicate a Ca2+-induced stabilization of 3.2 kcal mol(-1). We show that holoEF-SAM exists as a monomer, whereas apoEF-SAM readily forms a dimer and/or oligomer, and that oligomer to monomer transitions and vice versa are at least in part mediated by changes in surface hydrophobicity. Additionally, we find that the Ca2+ binding affinity of EF-SAM is relatively low with an apparent dissociation constant (Kd) of approximately 0.2-0.6 mM and a binding stoichiometry of 1. Our results suggest that EF-SAM actively participates in and is the likely the molecular trigger initiating STIM1 punctae formation via large conformational changes. The low Ca2+ affinity of EF-SAM is reconciled with the confirmed role of STIM1 as an ER Ca2+ sensor.
Article
Full-text available
Depletion of intracellular calcium (Ca²⁺) stores induces store-operated Ca²⁺ (SOC) entry across the plasma membrane (PM). STIM1, a putative Ca²⁺ sensor in the endoplasmic reticulum (ER), has been recently shown to be necessary for SOC channel activation. Here we show that STIM1 dynamically moves in tubulovesicular shape on the ER and its subcompartment in resting living cells, whereas, upon Ca²⁺ store depletion, it is rapidly redistributed into discrete puncta that are located underneath, but not inserted into the PM. Normal constitutive movement of STIM1 is mediated through the coiled-coil and Ser/Thr-rich C-terminal domains in the cytoplasmic region of STIM1, whereas subsequent inducible puncta formation further requires the sterile α motif domain protruding into the ER lumen. Each of these three domains (coiled-coil, Ser/Thr-rich, and sterile α motif) was essential for activating SOC channels. Hence, our findings based on structure–function experiments suggest that constitutive dynamic movement of STIM1 in the ER and its subcompartment is obligatory for subsequent depletion-dependent redistribution of STIM1 into puncta underneath the PM and activation of SOC channels. • B cell receptor • calcium signaling • DT40 • store-operated calcium
Article
Full-text available
In all cells Ca2+ signals are key to controlling a spectrum of cellular responses. Ca2+ signals activated by phospholipase C-coupled receptors have two components-rapid Ca2+ release from ER stores followed by slower Ca2+ entry from outside the cell. The coupling process between ER and PM to mediate this "store-operated" Ca2+ entry process has remained a molecular and mechanistic mystery. Through a combination of high throughput screening and molecular physiological approaches, the machinery and mechanism of this process have been elucidated. Two proteins are key to the coupling process. STIM1, a single spanning membrane protein with an unpaired Ca2+ binding EF-hand functions as the sensor of ER luminal Ca2+ and through redistribution in the ER transduces information directly to the PM. Orai1, a tetra-spanning PM protein, functions as the highly Ca2+ selective channel in the PM that is gated through interactions with the store-activated ER Ca2+ sensor. This molecular pas-de-deux between ER and PM components represents not only a crucial signaling pathway, but also a new paradigm in inter-organelle communication.
Article
The release of Ca2+ from intracellular stores by sub-optimal doses of inositol trisphosphate has been shown to be dose-related ('quantal'), and a simple model is proposed here to account for this phenomenon. It is suggested that there is a regulatory Ca2(+)-binding site on, or associated with, the luminal domain of the InsP3 receptor, which allosterically controls Ca2+ efflux, and the affinity for Ca2+ of that site is modulated by InsP3 binding to the cytoplasmic domain of the receptor; a similar mechanism applied to the ryanodine receptor might also explain some aspects of Ca2(+)-induced Ca2+ release. The stimulated entry of Ca2+ into a cell which occurs upon activation of inositide-linked receptors has been variously and confusingly proposed to be regulated by InsP3, InsP4, and/or a 'capacitative' Ca2+ pool; the mechanism of InsP3 receptor action suggested here is shown to lead to a potential reconciliation of all these conflicting proposals.
Article
EF-hand proteins undergo conformational changes upon binding of Ca2+. This event is a crucial step in many Ca2+-dependent cellular processes. Recent advances in the three-dimensional structural analysis of various EF-hand proteins have led to new insights into the structure and functional relationship of this large family of Ca2+-binding proteins.
Article
A novel gene (GOK) has been cloned from human chromosome region 11p15.5 that is believed to contain a gene or genes associated with a number of pediatric malignancies, including Wilms tumor. A 4-kb cDNA has been cloned and it encodes a predicted protein of approximately 84 kDa that could be translated in vitro. Computer analysis predicted that the protein had a signal peptide and may contain a transmembrane helix. Restriction mapping by pulsed-field electrophoresis indicates that GOK is located 1.7 kb telomeric of RRM1, and both genes are transcribed in the same direction. GOK displays high evolutionary conservation: cloning and partial sequencing of a mouse genomic clone revealed 90% identity with the human gene at both the nucleotide and the predicted amino acid levels.
Article
ERM (ezrin/radixin/moesin) proteins crosslink actin filaments with plasma membranes. The carboxyl termini of these proteins bind actin filaments, while the amino termini bind plasma membranes using a binding partner, such as CD44. Specific signals activate ERM proteins to bind actin filaments and the plasma membrane; these include phosphoinositides and/or phosphorylation mechanisms, which might be located downstream from the Rho-dependent pathway.
Article
Ezrin, radixin and moesin, collectively known as the ERM proteins, are a group of closely related membrane-cytoskeleton linkers that regulate cell adhesion and cortical morphogenesis. ERM proteins can self-associate through intra- and inter-molecular interactions, and these interactions mask several binding sites on the proteins. ERM activation involves unfolding of the molecule, and allows the protein to bind to plasma membrane components either directly, or indirectly through linker proteins. The discovery that the tumour-suppressor NF2, also known as merlin/schwannomin, is related to ERM proteins has added a new impetus to investigations of their roles. This review discusses current understanding of the structure and function of members of the ERM family of proteins.
Article
The elusive coupling between endoplasmic reticulum (ER) Ca2+ stores and plasma membrane (PM) "store-operated" Ca2+ entry channels was probed through a novel combination of cytoskeletal modifications. Whereas coupling was unaffected by disassembly of the actin cytoskeleton, in situ redistribution of F-actin into a tight cortical layer subjacent to the PM displaced cortical ER and prevented coupling between ER and PM Ca2+ entry channels, while not affecting inositol 1,4,5-trisphosphate-mediated store release. Importantly, disassembly of the induced cortical actin layer allowed ER to regain access to the PM and reestablish coupling of Ca2+ entry channels to Ca2+ store depletion. Coupling is concluded to be mediated by a physical "secretion-like" mechanism involving close but reversible interactions between the ER and the PM.
Article
STIM1 is a novel candidate growth suppressor gene mapping to the human chromosome region 11p15.5 that is associated with several malignancies. STIM1 overexpression studies in G401 rhabdoid tumour, rhabdomyosarcoma and rodent myoblast cell lines causes growth arrest, consistent with a potential role as a tumour growth suppressor. We used highly specific antibodies to show by immunofluorescence and cell surface biotinylation studies that STIM1 is located at the cell surface of K562 cells. Western blot analysis revealed that the 90-kDa STIM1 protein is ubiquitously expressed in various human primary cells and tumour cell lines. STIM1 is not secreted from cells and does not appear to undergo proteolytic processing. We show evidence of post-translational modification of STIM1, namely phosphorylation and N-linked glycosylation. Phosphorylation of STIM1 in vivo occurs predominantly on serine residues. Thus, STIM1, the putative tumour growth suppressor gene is ubiquitously expressed and has features of a regulatory cell-surface phosphoprotein.
Article
Stromal interaction molecule 1 (STIM1) is a cell surface transmembrane glycoprotein implicated in tumour growth control and stromal-haematopoietic cell interactions. A single sterile alpha motif (SAM) protein-protein interaction domain is modelled within its extracellular region, a subcellular localisation not previously described for other SAM domain-containing proteins. We have defined the transmembrane topology of STIM1 by determining the sites of N-linked glycosylation. We have confirmed that STIM1 is modified by N-linked glycosylation at two sites within the SAM domain itself, deduced as asparagine residues N131 and N171, demonstrating that STIM1 is translocated across the membrane of the endoplasmic reticulum such that the SAM domain resides within the endoplasmic reticulum (ER) lumen. Both N-linked oligosaccharides remain endoglycosidase H-sensitive, indicating absence of full processing within the ER and Golgi. This immature modification is nevertheless sufficient and critical for cell surface expression of STIM1. We show that STIM1-STIM1 homotypic interactions are mediated via the cytoplasmic rather than the extracellular region of STIM1, excluding an essential role for the SAM domain in these protein interactions. These studies provide the first evidence for an extracellular localisation of a SAM domain within any protein, and the first example of a SAM domain modified by N-linked glycosylation.
Article
Capacitative calcium entry is a process whereby the depletion of Ca(2+) from intracellular stores (likely endoplasmic or sarcoplasmic reticulum) activates plasma membrane Ca(2+) channels. Current research has focused on identification of capacitative calcium entry channels and the mechanism by which Ca(2+) store depletion activates the channels. Leading candidates for the channels are members of the transient receptor potential (TRP) superfamily, although no single gene or gene product has been definitively proven to mediate capacitative calcium entry. The mechanism for activation of the channels is not known; proposals fall into two general categories, either a diffusible signal released from the Ca(2+) stores when their Ca(2+) levels become depleted, or a more direct protein-protein interaction between constituents of the endoplasmic reticulum and the plasma membrane channels. Capacitative calcium entry is a major mechanism for regulated Ca(2+) influx in non-excitable cells, but recent research has indicated that this pathway plays an important role in the function of neuronal cells, and may be important in a number of neuropathological conditions. This review will summarize some of these more recent findings regarding the role of capacitative calcium entry in normal and pathological processes in the nervous system.
Article
ERM (ezrin/radixin/moesin) proteins provide a regulated linkage between membrane-associated proteins and the actin cytoskeleton. Previous work has shown that ezrin can exist in a dormant monomeric state in which the N-terminal FERM domain is tightly associated with the C-ERMAD (carboxyl-terminal ERM association domain), masking binding sites for at least some ligands, including F-actin and the scaffolding protein EBP50. Activation of ezrin requires relief of the intramolecular association, and this is believed to involve phosphorylation of threonine 567. Studies have therefore employed the T567D phosphomimetic mutant to explore the consequences of ezrin activation in vivo. Ezrin also exists as a stable dimer, in which the orientation of the two subunits is unknown, but might involve the central alpha-helical region predicted to form a coiled-coil. By characterization of ezrin mutants, we show that relief of the intramolecular association in the monomer results in unmasking of ligand binding sites and a significant conformational change, that the T567D mutation has a small effect on the biochemical activation of ezrin, and that the predicted coiled-coil region does not drive dimer formation. These results provide strong support for the conformational activation model of ezrin, elucidate the basis for dimer formation, and reveal that a mutant generally considered to be fully activated is not.
Article
Protein-protein interactions are essential for the assembly, regulation, and localization of functional protein complexes in the cell. SAM domains are among the most abundant protein-protein interaction motifs in organisms from yeast to humans. Although SAM domains adopt similar folds, they are remarkably versatile in their binding properties. Some identical SAM domains can interact with each other to form homodimers or polymers. In other cases, SAM domains can bind to other related SAM domains, to non-SAM domain-containing proteins, and even to RNA. Such versatility earns them functional roles in myriad biological processes, from signal transduction to transcriptional and translational regulation. In this review, we describe the structural basis of SAM domain interactions and highlight their roles in the scaffolding of protein complexes in normal and pathological processes.
Article
Protein-protein interactions occurring via the recognition of short peptide sequences by modular interaction domains play a central role in the assembly of signalling protein complexes and larger protein networks that regulate cellular behaviour. In addition to spatial and temporal factors, the specificity of signal transduction is intimately associated with the specificity of many co-operative, pairwise binding events upon which various pathways are built. Although protein interaction domains are usually identified via the recognition code, the consensus sequence motif, to which they selectively bind, they are highly versatile and play diverse roles in the cell. For example, a given interaction domain can bind to multiple sequences that exhibit no apparent identity, and, on the other hand, domains of the same class or different classes may favour a given consensus motif. This promiscuity in ligand selection is typified by the SH3 (Src homology 3) domain and several other interaction modules that commonly recognize proline-rich sequences. Furthermore, interaction domains are highly adaptable, a property that is essential for the evolution of novel pathways and modulation of signalling dynamics. The ability of certain interaction domains to perform multiple tasks, however, poses a challenge for the cell to control signalling specificity when cross-talk between pathways is undesired. Extensive structural and biochemical analysis of many interaction domains in recent years has started to shed light on the molecular basis underlying specific compared with diverse binding events that are mediated by interaction domains and the role affinity plays in affecting domain specificity and regulating cellular signal transduction.
Article
The coupling mechanism between endoplasmic reticulum (ER) Ca(2+) stores and plasma membrane (PM) store-operated channels (SOCs) remains elusive [1-3]. STIM1 was shown to play a crucial role in this coupling process [4-7]; however, the role of the closely related STIM2 protein remains undetermined. We reveal that STIM2 is a powerful SOC inhibitor when expressed in HEK293, PC12, A7r5, and Jurkat T cells. This contrasts with gain of SOC function in STIM1-expressing cells. While STIM1 is expressed in both the ER and plasma membrane, STIM2 is expressed only intracellularly. Store depletion induces redistribution of STIM1 into distinct "puncta." STIM2 translocates into puncta upon store depletion only when coexpressed with STIM1. Double labeling shows coincidence of STIM1 and STIM2 within puncta, and immunoprecipitation reveals direct interactions between STIM1 and STIM2. Independent of store depletion, STIM2 colocalizes with and blocks the function of a STIM1 EF-hand mutant that preexists in puncta and is constitutively coupled to activate SOCs. Thus, whereas STIM1 is a required mediator of SOC activation, STIM2 is a powerful inhibitor of this process, interfering with STIM1-mediated SOC activation at a point downstream of puncta formation. The opposing functions of STIM1 and STIM2 suggest they may play a coordinated role in controlling SOC-mediated Ca(2+) entry signals.
Article
Receptor-evoked Ca2+ signalling involves Ca2+ release from the endoplasmic reticulum, followed by Ca2+ influx across the plasma membrane. Ca2+ influx is essential for many cellular functions, from secretion to transcription, and is mediated by Ca2+-release activated Ca2+ (I(crac)) channels and store-operated calcium entry (SOC) channels. Although the molecular identity and regulation of I(crac) and SOC channels have not been precisely determined, notable recent findings are the identification of STIM1, which has been indicated to regulate SOC and I(crac) channels by functioning as an endoplasmic reticulum Ca2+ sensor, and ORAI1 (ref. 7) or CRACM1 (ref. 8)--both of which may function as I(crac) channels or as an I(crac) subunit. How STIM1 activates the Ca2+ influx channels and whether STIM1 contributes to the channel pore remains unknown. Here, we identify the structural features that are essential for STIM1-dependent activation of SOC and I(crac) channels, and demonstrate that they are identical to those involved in the binding and activation of TRPC1. Notably, the cytosolic carboxyl terminus of STIM1 is sufficient to activate SOC, I(crac) and TRPC1 channels even when native STIM1 is depleted by small interfering RNA. Activity of STIM1 requires an ERM domain, which mediates the selective binding of STIM1 to TRPC1, 2 and 4, but not to TRPC3, 6 or 7, and a cationic lysine-rich region, which is essential for gating of TRPC1. Deletion of either region in the constitutively active STIM1(D76A) yields dominant-negative mutants that block native SOC channels, expressed TRPC1 in HEK293 cells and I(crac) in Jurkat cells. These observations implicate STIM1 as a key regulator of activity rather than a channel component, and reveal similar regulation of SOC, I(crac) and TRPC channel activation by STIM1.
Article
Store-operated Ca2+ entry (SOCE) mediates much of the Ca2+ entry evoked by receptors that stimulate phospholipase C. However, for 20 years, the plasma membrane Ca2+ channel and the signal linking its activation to loss of Ca2+ from the endoplasmic reticulum (ER) have eluded detection. But the search might now be over. Two proteins, STIM1 (the ER Ca2+ sensor) and Orai1 (the Ca2+ channel), have recently been identified as the missing links in SOCE.
Article
The highly polarized nature of epithelial cells in exocrine glands necessitates targeting, assembly into complexes and confinement of the molecules comprising the Ca(2+) signaling apparatus, to cellular microdomains. Such high degree of polarized localization has been shown for all Ca(2+) signaling molecules tested, including G protein coupled receptors and their associated proteins, Ca(2+) pumps, Ca(2+) influx channels at the plasma membrane and Ca(2+) release channels in the endoplasmic reticulum. Although the physiological significance of polarized Ca(2+) signaling is clear, little is known about the mechanism of targeting, assembly and retention of Ca(2+) signaling complexes in cellular microdomains. The present review attempts to summarize the evidence in favor of polarized expression of Ca(2+) signaling proteins at the apical pole of secretory cells with emphasis on the role of scaffolding proteins in the assembly and function of the Ca(2+) signaling complexes. The consequence of polarized enrichment of Ca(2+) signaling complexes at the apical pole is generation of an apical to basal pole gradient of cell responsiveness that, at low physiological agonist concentrations, limits Ca(2+) spikes to the apical pole, and when a Ca(2+) wave occurs, it always propagates from the apical to the basal pole. Our understanding of Ca(2+) signaling in microdomains is likely to increase rapidly with the application of techniques to controllably and selectively disrupt components of the complexes and apply high resolution recording techniques, such as TIRF microscopy to this problem.
Article
STIM1 and Orai1 have recently been identified to be crucial in the regulation of store-operated Ca(2+) entry. However, it remains to be established how STIM1 couples store depletion to the functioning of Orai1 in the plasma membrane. Using quantitative measurement, we find little STIM1 on the surface membrane which is not increased by store depletion. We further demonstrate that Orai1 assembles into clusters that co-localize with STIM1 aggregations upon store depletion. The clustering of Orai1 is only seen when Oari1 are co-expressed with STIM1, but not when expressed alone. Moreover, ER retreat from cell periphery leads to mismatching of Orai1 and STIM1 puncta. Therefore, we propose that store depletion causes aggregation and translocation of STIM1 in close apposition to the plasma membrane, which in turn recruits Orai1 in the plasma membrane to the sites of STIM1 aggregates to assemble functional units of CRAC channels in a stoichiometric manner.
Article
Recent studies have indicated a critical role for STIM (stromal interacting molecule) proteins in the regulation of the store-operated mode of receptor-activated Ca2+ entry. Current models emphasize the role of STIM located in the endoplasmic reticulum membrane, where a Ca2+-binding EF-hand domain within the N-terminal of the protein lies within the lumen and is thought to represent the sensor for the depletion of intracellular Ca2+ stores. Dissociation of Ca2+ from this domain induces the aggregation of STIM to regions of the ER immediately adjacent to the plasma membrane where it acts to regulate the activity of store-operated Ca2+ channels. However, the possible effects of STIM on other modes of receptor-activated Ca2+ entry have not been examined. Here we show that STIM1 also regulates the arachidonic-acid-regulated Ca2+-selective (ARC) channels - receptor-activated Ca2+ entry channels whose activation is entirely independent of store depletion. Regulation of the ARC channels by STIM1 does not involve dissociation of Ca2+ from the EF-hand, or any translocation of STIM1. Instead, a critical role of STIM1 resident in the plasma membrane is indicated. Thus, exposure of intact cells to an antibody targeting the extracellular N-terminal domain of STIM1 inhibits ARC channel activity without significantly affecting the store-operated channels. A similar specific inhibition of the ARC channels is seen in cells expressing a STIM1 construct in which the N-linked glycosylation sites essential for the constitutive cell surface expression of STIM1, were mutated. We conclude that, in contrast to store-operated channels, regulation of ARC channels by STIM1 depends exclusively on the pool of STIM1 constitutively residing in the plasma membrane. These data demonstrate that STIM1 is a more universal regulator of Ca2+ entry pathways than previously thought, and appears to have multiple modes of action.
The FERM domain: a unique module involved in the linkage of cytoplasmic proteins to the membrane
  • Chishti
ERM proteins in cell adhesion and membrane dynamics
  • Mangeat
Identification and characterization of the STIM (stromal interaction molecule) gene family: coding for a novel class of transmembrane proteins
  • Williams