A rise in cytosolic Ca2+ is used as a key signalling messenger in eukaryotic cells. The Ca2+ signal drives life and death and controls myriad responses in between. Inherent in the use of such a multifarious signal is the danger of disease, arising from dysregulated Ca2+ signalling. One ancient, highly conserved and widespread Ca2+ entry pathway is the store-operated Ca2+ release-activated Ca2+ (CRAC) channel. Mutations in STIM1 and ORAI1, the genes that encode the functional channel, are tightly linked to a CRAC channelopathy in humans, which encompasses severe combined immune deficiency, myopathy and anhidrotic ectodermal dysplasia. Moreover, sustained Ca2+ entry through the channels leads to a range of systemic disorders, including acute pancreatitis, asthma and inflammatory bowel disease. In this review, we describe how aberrant CRAC channel activity causes a range of diseases, highlighting commonalities between these diverse pathologies.

Hundreds of hormones, neurotransmitters, paracrine signals and mechanical forces bombard the surface of a mammalian cell each day. These primary stimuli trigger a wide range of responses across an enormous temporal bandwidth, from sub-milliseconds to days. Remarkably, this diverse array of responses is co-ordinated through a surprisingly small number of intracellular second messengers. Of the known second messengers, cytosolic Ca2+ is a universal signal used by cells throughout the phylogenetic tree [1,2]. Balanced finely like a tightrope walker, cytosolic Ca2+ is both the harbinger of vitality and the spectre of demise. A rise in cytosolic Ca2+ is essential for fertilisation of the egg, driving the beginning of life [3]. Throughout life, cytosolic Ca2+ regulates neurotransmitter release, muscle contraction, energy production, hormone secretion, cell motility and cell growth and differentiation [3]. However, cytosolic Ca2+ can also cause cell death through apoptosis or necrosis [2,3]. An inherent risk of relying on a single signal to control such fundamental physiological processes is that any distortion or compromise of the signal can result in diseases with often debilitating consequences. This review focuses on disorders that arise from aberrant Ca2+ signals through one widely distributed Ca2+ channel, the store-operated Ca2+ release-activated Ca2+ (CRAC) channel, drawing on examples from different systems to highlight commonalities.

The distribution of Ca²+ across and within cells is summarised in Figure 1A. The extracellular concentration of free Ca²+ is approximately 1.2 mM, while resting cytosolic Ca²+ is typically maintained at 50–100 nM. This low cytosolic concentration is maintained by P-type Ca²+ ATPase pumps and electrogenic Na+-Ca²+ exchangers in the plasma membrane. Most cells also have a negative membrane potential, which, combined with the steep Ca²+ concentration gradient across the membrane, creates a significant electrochemical gradient that drives Ca²+ influx. Because the phospholipid bilayer is impermeable to ions, Ca²+ movement occurs through various Ca²+-permeable ion channel proteins distributed in the plasma membrane. These channels are gated by a range of factors, including voltage, mechanical forces, temperature and extracellular or intracellular ligands (Figure 1B).

Distribution of calcium across and within cells.

Figure 1:
Distribution of calcium across and within cells.

(A) A cartoon compares the distribution of Ca2+ across and within a typical eukaryotic cell. Values shown are for unstimulated conditions. Upon stimulation, Ca2+ rises in the cytosol, nucleoplasm and mitochondrial matrix and, depending on the stimulus, falls within the endoplasmic reticulum, lysosome and Golgi apparatus. (B) Types of plasma membrane Ca2+-permeable ion channels. P2X is an ATP-gated non-selective cation channel representative of the ligand-gated family. The figure was generated using BioRender. TRP, Transient Receptor Potential; VGCC, voltage-gated calcium channel. Values adjacent to organelles represent concentration within the organelle.

Figure 1:
Distribution of calcium across and within cells.

(A) A cartoon compares the distribution of Ca2+ across and within a typical eukaryotic cell. Values shown are for unstimulated conditions. Upon stimulation, Ca2+ rises in the cytosol, nucleoplasm and mitochondrial matrix and, depending on the stimulus, falls within the endoplasmic reticulum, lysosome and Golgi apparatus. (B) Types of plasma membrane Ca2+-permeable ion channels. P2X is an ATP-gated non-selective cation channel representative of the ligand-gated family. The figure was generated using BioRender. TRP, Transient Receptor Potential; VGCC, voltage-gated calcium channel. Values adjacent to organelles represent concentration within the organelle.

Close modal

Several organelles function as mobilisable Ca²+ stores, including the endoplasmic reticulum, Golgi apparatus and lysosomes [3]. These organelles release Ca²+ into the cytosol through Ca²+-permeable ion channels in response to specific second messengers. Mitochondria are also heavily involved in cytosolic Ca²+ dynamics, taking up Ca²+ through the mitochondrial Ca²+ uniporter (MCU) and releasing it via Na+-Ca²+ exchange [4]. However, mitochondrial Ca²+ release occurs only after the organelle has accumulated Ca²+ through the MCU in response to a prior rise in cytosolic Ca²+.

Although Ca²+ release from organelles contributes to cytosolic Ca²+ signalling, the limited storage capacity of these organelles means that influx from the large extracellular Ca²+ reservoir is essential to sustain Ca²+ signalling. Furthermore, the substantial electrochemical gradient for Ca²+ entry makes an increase in membrane permeability following the opening of a Ca²+ ion channel highly effective in rapidly raising cytosolic Ca²+. One major type of Ca²+ channel is the store-operated CRAC channel.

CRAC channels in the plasma membrane comprise an evolutionarily conserved and widespread route for Ca2+ entry [5,6]. These channels are robustly expressed in electrically non-excitable cells, such as immune cells, epithelia, hepatocytes, endothelia and glia but are also found in neurons and muscle, although their function in excitable cells remains less well established.

The defining feature of CRAC channels is their dependence on the Ca2+ content of the endoplasmic reticulum (ER) [7]. When the Ca2+ store is full, the channels are closed, but when Ca2+ within the store falls, the channels open. Regardless of how stores are depleted of Ca2+, the channels activate. Physiologically, this is generally accomplished following activation of G protein-coupled receptors that link to Gq heterotrimeric GTP-binding proteins and phospholipase Cß isoforms or growth factor receptors that activate phospholipase Cγ enzymes. In both cases, phospholipase C hydrolyses the membrane phospholipid phosphatidyl inositol 4,5-bisphosphate to generate the second messengers inositol 1,4,5-trisphosphate (InsP3) and diacylglycerol. Diacylglycerol stimulates protein kinase C, whereas InsP3 diffuses into the cytosol and rapidly releases Ca2+ by binding to and opening InsP3-gated Ca2+ channels in the ER membrane [3].

The CRAC channel machinery is a multi-component system comprising two key players: Orai and STIM proteins. Orai proteins, located in the plasma membrane, form the pore-lining subunits of CRAC channels [8-11] (Figure 2). STIM proteins, embedded in the ER membrane, act as Ca2+ sensors that regulate CRAC channel gating and activity [13,14] (Figure 2). These proteins have a canonical Ca²+-binding EF hand on the N-terminus facing the ER lumen. In humans, three Orai paralogs (Orai1, Orai2 and Orai3) and two STIM paralogs (STIM1 and STIM2) are present, which, along with several splice variants, alternative translation initiation forms of Orai1, and differences between STIM1 and STIM2 in Ca2+ affinities, enable more precise fine-tuning of store-operated Ca2+ entry [5,6]. Following a physiological stimulus, Ca2+ levels in the ER lumen drop as a result of InsP3-dependent Ca²+ release. The fall in ER Ca2+ content is detected by STIM1 and STIM2 through Ca2+ unbinding from the EF hand domains. This process induces a conformational change that unmasks cytosolic domains, resulting in exposure of channel-activating regions (Figure 2). STIM proteins then translocate across the ER to specialised regions that are located just below the plasma membrane. Here, the exposed channel-activating domain, also called STIM Orai-activating region of the activated STIM proteins, interacts with the Orai subunits to assemble and gate open the pore, allowing the influx of extracellular Ca2+ through CRAC channels [15,16] (Figure 2).

Molecular basis of the Ca2+ release-activated Ca2+ (CRAC) channel.

Figure 2:
Molecular basis of the Ca2+ release-activated Ca2+ (CRAC) channel.

In resting cells with endoplasmic reticulum (ER) replete with Ca2+ (~400 µM), STIM1 dimers and Orai1 channels are in inactive states in the ER and plasma membrane, respectively (1). Stimulation of cell-surface receptors increases InsP3 (2), and this leads to Ca2+ store depletion (3), where ER Ca2+ content falls significantly. The drop in lumen Ca2+ results in Ca2+ disassociation from the EF hand of STIM1 (4a). Unbinding of Ca2+ activates STIM1 dimers by promoting aggregation and interaction of EF-SAM domains within the ER lumen (4a), extending the coiled coil 1 (CC1) domain and exposing the locked STIM1 cytosolic channel-activating domain/STIM Orai-activating region (CAD/SOAR) region and the PI(4,5)P2 binding lysine (K)-rich domain (4b). STIM1 forms oligomers, which bind the phospholipid PI (4.5)P2 at the inner leaflet of the plasma membrane via the lysine (K)-rich domain (5b). This stabilises pre-existing ER-plasma membrane junctions and forms new ones. Exposed CAD/SOAR binds to the C-terminus of diffusing Orai1 channels, trapping them at the junctions and gating them open (6). SERCA pump activity, together with a fall in InsP3-dependent Ca2+ release, enables store refilling of Ca2+ (7). Ca2+ then reassociates with the EF hand of STIM1, leading to disaggregation of the STIM1-Orai1 complex and termination of Ca2+ influx (1). Adapted and modified from [12].

Figure 2:
Molecular basis of the Ca2+ release-activated Ca2+ (CRAC) channel.

In resting cells with endoplasmic reticulum (ER) replete with Ca2+ (~400 µM), STIM1 dimers and Orai1 channels are in inactive states in the ER and plasma membrane, respectively (1). Stimulation of cell-surface receptors increases InsP3 (2), and this leads to Ca2+ store depletion (3), where ER Ca2+ content falls significantly. The drop in lumen Ca2+ results in Ca2+ disassociation from the EF hand of STIM1 (4a). Unbinding of Ca2+ activates STIM1 dimers by promoting aggregation and interaction of EF-SAM domains within the ER lumen (4a), extending the coiled coil 1 (CC1) domain and exposing the locked STIM1 cytosolic channel-activating domain/STIM Orai-activating region (CAD/SOAR) region and the PI(4,5)P2 binding lysine (K)-rich domain (4b). STIM1 forms oligomers, which bind the phospholipid PI (4.5)P2 at the inner leaflet of the plasma membrane via the lysine (K)-rich domain (5b). This stabilises pre-existing ER-plasma membrane junctions and forms new ones. Exposed CAD/SOAR binds to the C-terminus of diffusing Orai1 channels, trapping them at the junctions and gating them open (6). SERCA pump activity, together with a fall in InsP3-dependent Ca2+ release, enables store refilling of Ca2+ (7). Ca2+ then reassociates with the EF hand of STIM1, leading to disaggregation of the STIM1-Orai1 complex and termination of Ca2+ influx (1). Adapted and modified from [12].

Close modal

X-ray crystallographic studies on Drosophila Orai revealed that the channel pore is composed of a hexameric ring of Orai proteins, each with four transmembrane (TM) helices, where six TM1 helices, one from each subunit, form the pore [17]. Numerous studies have investigated the gating rearrangements in Orai1 that enable the channel pore to transition between closed and open states following STIM1-mediated binding, and various residues and segments on both Orai1 and STIM1 have been identified as critical for channel activation and inactivation [18,19]. A wide range of mutations, including disease-causing gain-of-function (GoF) mutations, have been reported in Orai1, enabling constitutive Ca2+ entry independent of store depletion and STIM1 activation. While the majority of GoF mutations studied compromise the selectivity of the Orai channel for Ca2+ ions, mutations at only two loci—one in TM2 (H134) and another in the TM4 cytosolic extension (LVSHK)—have been suggested to mimic the STIM1-gated state of the channel. The TM4 region of human Orai1 features a kink at the conserved proline residue (P245) and a cytosolic ‘nexus’ sequence (LVSHK) crucial for channel activation. Early studies on Drosophila melanogaster Orai described the TM4 extension as being locked in a ‘latched’ state, preventing pore opening, but the physiological relevance of this state remains contested. Mutations in various Orai regions, including pore-lining residues such as F99 and V102, cause constitutive channel opening by altering gating mechanisms, while mutations in TM2 (e.g. H134) and TM4 can trigger conformational changes leading to channel activation [18,20]. Future structural studies exploring both the closed and open states of the Orai channel in complex with STIM1 are expected to address these unresolved questions on the gating mechanisms of CRAC channels in health and disease.

Orai1 participates in a reversible signalosome with AKAP79. AKAP79 is a scaffolding protein embedded in the inner leaflet of the plasma membrane and acts as a signalling hub through binding protein kinases A and C, calmodulin, as well as the Ca2+-activated protein phosphatase calcineurin [21]. Activated calcineurin dephosphorylates four members of the transcription factor nuclear factor of activated T cells (NFAT1-4), exposing a nuclear localisation sequence, which enables the transcription factor to translocate to the nucleus, where it regulates the expression of various cytokines and chemokines that help shape an inflammatory response. After store depletion, AKAP79 associates with the N-terminus of Orai1, thereby placing the Ca2+-activated calcineurin within the realm of the local Ca2+ signal adjacent to the open channels [22]. This robustly activates the phosphatase and leads to dephosphorylation and subsequent nuclear translocation of NFAT. Intimate functional association between Orai1 and AKAP79 enables CRAC channels to have privileged access to the nucleus. CRAC channels thus not only engage local Ca2+ signalling to drive downstream Ca2+-dependent processes but also sustain Ca2+ signalling by enabling Ca2+ store refilling via the action of SERCA pumps.

Important insight into the function of STIM and Orai1 proteins has been gleaned from studies on patients suffering from immunodeficiencies with loss-of-function (LoF) mutations in these genes. Although very rare, all the mutations produce a similar set of phenotypes, leading to what Stefan Feske has called the CRAC channelopathy [23]. The channelopathy encompasses immunodeficiency, exemplified by recurrent severe infections to a broad range of pathogens, including viruses, bacteria, mycobacteria and fungi. These infections can lead to death shortly after birth unless haematopoietic stem cell transplantation is provided. The immunodeficiency was traced to an inability of CD4+ and CD8+ T cells to proliferate and produce cytokines [24]. In the absence of store-operated Ca2+ entry through Orai1, NFAT activation is prevented, and this contributes to impaired cytokine production. The patients also present with myopathy, exhibiting global muscle hypotonia with reduced muscle strength, and anhidrotic ectodermal dysplasia manifested as reduced sweat production and loss of dental enamel [25]. Most of the patients with LoF mutations in STIM1 also show thrombocytopenia and autoimmune haemolytic anaemia. Interestingly, thrombocytopenia was less common in patients with LoF mutations in Orai1 [25].

GI tract

Acute pancreatitis

Acute pancreatitis is caused by inflammation of the pancreas, typically triggered by excessive alcohol consumption, bile duct obstruction (e.g. due to gallstones), certain medications (including thiazides, tetracycline, sulfonamides and the immunosuppressant azathioprine), pancreatic cancer, elevated blood triglycerides or pancreatic trauma resulting from injury or surgery [26]. The condition arises from inappropriate activation of trypsinogen and other proteolytic proenzymes that are stored within zymogen granules in the apical pole of pancreatic acinar cells (Figure 3). These proteases then autodigest the pancreas, resulting in necrosis [27]. Agents that trigger acute pancreatitis, such as alcohol and fatty acid ethyl esters, induce a sustained elevation of cytosolic Ca2+ due to Orai1-based CRAC channel opening, which leads to the activation of trypsin within endocytic vacuoles and mitochondrial Ca2+ overload, disrupting ATP production (Figure 3) [28,29]. Pharmacological block of the CRAC channel with GSK-7975A suppressed Ca2+ entry and significantly decreased both intracellular protease activation and necrosis induced by palmitoleic acid ethyl ester. In mouse models of acute pancreatitis, the Orai1 channel inhibitors GSK-7975A and CM_128/CM4620 independently suppressed local and systemic pathophysiology [30] and prevented recurrent acute pancreatitis and early stages of chronic pancreatitis [31]. A phase 2 study on 21 patients with acute pancreatitis, systemic inflammatory response syndrome and hypoxemia showed a favourable safety profile and outcomes in patients treated with a CRAC channel blocker [32].

Orai1 channels play a central role in the development of acute pancreatitis.
Figure 3:
Orai1 channels play a central role in the development of acute pancreatitis.

Sustained Ca2+ entry through Orai1 channels in PACs following stimulation with bile acids or alcohol leads to premature activation and secretion of trypsin, which autodigests the pancreas. Damaged PACs also release other signals, including kallikrein and adenine nucleotides. Kallikrein cleaves kininogens to generate bradykinin, which acts on bradykinin type 2 receptors in PSCs to open Orai1 channels. This generates nitric oxide, which diffuses to PACs and promotes necrosis. ATP/ADP released from PACs activates Orai1 channels in immune cells, which leads to a local inflammatory response through the release of cytokines. Blue circles at apical pole denote trypsin-containing granules, which are normally released into the pancreatic duct.

Figure 3:
Orai1 channels play a central role in the development of acute pancreatitis.

Sustained Ca2+ entry through Orai1 channels in PACs following stimulation with bile acids or alcohol leads to premature activation and secretion of trypsin, which autodigests the pancreas. Damaged PACs also release other signals, including kallikrein and adenine nucleotides. Kallikrein cleaves kininogens to generate bradykinin, which acts on bradykinin type 2 receptors in PSCs to open Orai1 channels. This generates nitric oxide, which diffuses to PACs and promotes necrosis. ATP/ADP released from PACs activates Orai1 channels in immune cells, which leads to a local inflammatory response through the release of cytokines. Blue circles at apical pole denote trypsin-containing granules, which are normally released into the pancreatic duct.

Close modal

Although effective, CRAC channel blockers do not fully prevent acute pancreatitis even at high doses. A recent study reported that the combination of CM4620 and galactose, an energy supplement that restores ATP production, was more effective than CM4620 alone [33]. Moreover, the combination allowed for a significantly lower dose of CM4620 to be used, reducing potential off-targets arising that could be induced by a higher dose of drug [33].

CM4620 reduced the severity of acute pancreatitis in a rat model but did so not only by targeting the pancreatic acinar and stellate cells in the pancreas but also by reducing the neutrophil oxidative burst and the release of inflammatory mediators from invading immune cells [34], although Orai1 in macrophages might not be involved [35]. The pancreatic duct transports enzymes secreted from acinar cells to the small intestine, and ductal cells secrete bicarbonate-rich fluid to neutralise acidity in the gut from H+ secretion by parietal cells in the stomach. Ca2+ overload in ductal cells would compromise function and reduce the removal of digestive enzymes from the duct, exacerbating pancreatic autodigestion. CM5620, another CRAC channel blocker, reduced Ca2+ influx into pancreatic ductal cells in response to bile acids or ethanol challenge, protecting secretory ductal function. CRAC channel block also partially maintained pancreatic secretion in a mouse model of cerulein-induced pancreatitis [36]. Therefore, a CRAC channel blocker targets multiple cell types associated with acute pancreatitis at the same time (Figure 3).

Another potential approach could target CRAC channels indirectly. Store-operated calcium entry-associated regulatory factor (SARAF) is an ER-resident protein that negatively regulates CRAC channel activity. Expression of SARAF was found to fall in mice exposed to triggers of pancreatitis and in patients with pancreatitis [37]. Mice in which the SARAF gene had been knocked out developed more pronounced pancreatitis, whereas overexpression of SARAF suppressed inflammation and dampened pancreatitis [37].

Inflammatory bowel disease (IBD)

Inflammatory bowel disease (IBD) is characterised by chronic inflammation of the digestive tract, with the two main manifestations being ulcerative colitis and Crohn’s disease. Although idiopathic in nature, IBD is thought to arise from a combination of genetic and environmental factors, with the latter involving the gut microbiome, surgery, smoking or exposure to a range of early life factors, including GI infections and antibiotics. Prolonged IBD is associated with the development of colorectal cancer. A hallmark of IBD is a dysregulated intestinal immune response, particularly within the lamina propria. T cells isolated from human inflamed mucosa showed substantially larger store-operated Ca2+ entry and proliferation, when compared with those extracted from non-inflamed tissue [38]. This can be explained by the ~fivefold increase in STIM1 protein expression in human inflamed intestinal mucosa with significant staining in CD45+ cells within the lamina propria [39]. The CRAC channel inhibitor Synta66 reduced the levels of inflammatory cytokines IFN-γ, IL-2 and IL-17 in biopsy specimens from patients with IBD, pointing to an important role for CRAC channels in immune cell activation [40]. The immunosuppressant FK506, which would inhibit all CRAC channel-dependent gene expression through NFAT activation, also reduced IFN-γ, IL-2 and IL-17 levels to a similar extent to Synta66. However, FK506 also suppressed IL-8 expression, whereas Synta66 was ineffective [41]. The reason for this difference remains unclear. Genetic evidence for a role for CRAC channels was provided in a study that generated Orai1 knock-in mice expressing inactive R93W (human R91W) channels. These channels are expressed normally but cannot be gated by STIM proteins and, therefore, do not conduct Ca2+. T cells expressing the R93W mutant channels failed to induce colitis in an adoptive transfer model [41]. A detailed study utilising mass cytometry found an increased number of naïve T cells, CD4+ T cells, IFN-γ-producing CD8+ T cells, T-regs and innate lymphoid cells in the lamina propria of inflamed intestinal tissue from IBD patients [42]. The CRAC channel blocker BTP2 inhibited store-operated Ca2+ entry and reduced the production of multiple pro-inflammatory cytokines. Importantly, BTP2 had no effect on intestinal epithelia cell differentiation or on barrier function, demonstrating a beneficial effect on immune cells without compromising epithelial cell activity or network integrity. Another CRAC channel blocker, CM4620, was found to be more potent than BTP2 and was effective in reducing intestinal inflammation and subsequent damage in a mouse model of IBD [42].

Lung

Asthma

Asthma is a chronic inflammation of the lung that results in prolonged bouts of coughing, tightness of the chest, wheezing and shortness of breath. Asthma is a heterogeneous disease that can be driven by both genetic and environmental factors [43]. Sources of environmental aero-allergen include those released by the house dust mite, cockroach, fungi, animal dander and pollen. The house dust mite is the major source of aero-allergens in humans, and sensitisation to mite-derived allergens is a common cause of asthma [44,45]. The main types of house dust mite are Dermatophagoides pteronyssinus and Dermatophagoides farina, with allergens derived from them referred to as Der p and Der f proteins. Many mite-derived allergens are enzymes, particularly cysteine and serine proteases [44].

Inhaled allergens stimulate airway epithelia to release alarmins from their basolateral side, which then activate various lung-resident immune cells, including innate lymphoid type 2 (ILC2) cells, dendritic cells and mast cells (Figure 4) [44-47]. Released cytokines, chemokines, enzymes and paracrine signals recruit other immune cells, including Th2 lymphocytes and IgE-producing B cells, contract airway smooth muscle and drive remodelling of the airways (Figure 4). Meanwhile, allergens weaken the integrity of the epithelial barrier, damaging tight junctions and enabling their passage through the paracellular pathway. STIM1 is up-regulated in airway smooth muscle of asthmatic mice and is required for the metabolic and transcriptional reprogramming that is necessary for remodelling [48]. Whether store-operated Ca2+ entry contributes to epithelial barrier function is currently not known.

CRAC channels and asthma.
Figure 4:
CRAC channels and asthma.

Interplay between airway epithelia, smooth muscle and immune cells leads to the development of chronic inflammation in lungs following exposure to allergens. Immature dendritic cells, after being exposed to allergens and alarmins, travel to draining lymph nodes where they activate naïve CD4+ T cells, which then migrate to the lung. TLR denotes toll-like receptor; TSLP is thymic stromal lymphoprotein; DC is dendritic cell; Eos is eosinophil; and ILC-2 is innate lymphoid type 2 cell. See text for further details. Not all immune cells are displayed for simplicity.

Figure 4:
CRAC channels and asthma.

Interplay between airway epithelia, smooth muscle and immune cells leads to the development of chronic inflammation in lungs following exposure to allergens. Immature dendritic cells, after being exposed to allergens and alarmins, travel to draining lymph nodes where they activate naïve CD4+ T cells, which then migrate to the lung. TLR denotes toll-like receptor; TSLP is thymic stromal lymphoprotein; DC is dendritic cell; Eos is eosinophil; and ILC-2 is innate lymphoid type 2 cell. See text for further details. Not all immune cells are displayed for simplicity.

Close modal

Exposure of airway epithelial cells, mast cells or Jurkat T lymphocytes to mite extract resulted in Ca2+ entry through CRAC channels, which led to increased expression of pro-inflammatory genes and cell migration [49,50]. These responses were accomplished through activation of protease-activated receptors (PARs) type 4 and 2 (PAR4 and PAR2). PARS are Gq-coupled receptors that stimulate phospholipase C to generate InsP3. InsP3 releases Ca2+ from the ER into the cytosol, lowering store Ca2+ content and leading to subsequent gating of Orai1 by STIM1 through the canonical pathway. CRAC channel blockers or knockdown of STIM1 or Orai1 inhibited the sustained Ca2+ signal evoked by house dust mite, as well as gene expression and cell migration [49,51]. In airway epithelia, CRAC channel activation leads to NFAT signalling and subsequent expression of pro-inflammatory cytokines IL-6 and IL-8 [49], as well as c-fos activation [51]. CRAC channels are also required for epithelial cell migration [51].

House dust mite extract contains a plethora of allergens along with adjuvants, lipids and other signalling molecules. Combining cytosolic Ca2+ measurements with extract fractionation and mass spectrometry, Lin et al. identified Der p3 as a critical component in the mite extract that activated CRAC channels [51]. Der p3 is a serine protease and stimulates PAR4. Like other serine proteases, inactive pro-Der p3 is cleaved by the cysteine protease Der p1 to release active Der p3. The combination of recombinant Der p1 and pro-Der p3 mimicked the ability of dust mite extract to open CRAC channels. Targeting both CRAC channels and PAR4 pharmacologically at the same time, with sub-maximal concentrations of each inhibitor, suppressed house dust mite-induced activation of mast cells [51]. These in vitro studies suggest that targeting Der p3, PARs and CRAC channels should open up new targets for intervention, alone or in combination.

Several studies have demonstrated the efficacy of CRAC channel blockers in dampening the severity of allergic asthma in various models. BTP2 inhibited cytokine release and eosinophil infiltration in rat and guinea pig models of asthma [52]. Synta66 substantially decreased cytokine release from lymphocytes that had been isolated from bronchioalveolar lavage of healthy and asthmatic donors [53]. The pyrazole RP3128 reduced eosinophil and mast cell infiltration following antigen challenge of sensitised guinea pigs [54]. In a mouse model of house dust mite-induced asthma, oral application of the CRAC channel blocker CM4620 significantly reduced the severity of the asthmatic response [55]. Peribronchial inflammation, mucus production, levels of pro-inflammatory cytokines (IL-4, IL-5, IL-13 and IL-16) were all reduced following CRAC channel inhibition, as were the number of B cells, eosinophils and CD4+ T cells found in bronchoalveolar lavage fluid [55].

Group 2 ILC2s are activated by various alarmins from airway epithelia to release type-2 cytokines that help drive lung inflammation (Figure 4). Orai channels in ILC2 cells are important for the development of airway hypersensitivity and inflammation in the lung [56]. Challenge with the fungus Alternaria alternata increased airway resistance, as well as the number of eosinophils in bronchoalveolar lavage fluid, and these responses were less pronounced in the presence of a CRAC channel blocker [56]. Importantly, adoptive transfer of Orai1-/- or Orai1/2-/- ILC2 cells reduced airway hypersensitivity and eosinophilia compared with control ILC2 cells [56].

Collectively, these experiments provide strong evidence for the role of CRAC channels in asthma in an in vivo model and reinforce the view that targeting the channels should be an effective treatment for managing the condition.

Teeth

Enamel

One of the hallmarks of patients with a CRAC channelopathy is ectodermal dysplasia, which results in severely dysplastic dental enamel [23,41,57]. Enamel is produced by ameloblasts, which secrete proteins, such as enamelin and amelogenin, that mineralise and form hard enamel. Specific deletion of STIM1/2 genes in murine ameloblasts resulted in diminished store-operated Ca2+ entry and a hypomineralised enamel with a lower Ca2+ content, which was thinner and mechanically weaker than normal enamel [58]. The loss of STIM proteins led to an increase in reactive oxygen species, reduced mitochondrial function, altered mitochondrial shape and a redistribution of the organelle within the cytosol. Detailed investigation into a patient homozygous for a frameshift mutation and subsequent premature stop codon in TM3 of Orai1 (V181SfsX8) confirmed loss of store-operated Ca2+ entry [59]. The patient presented with pitted enamel hypoplasia of the primary central incisors and hypomineralisation of the primary maxillary incisors [59]. Ca2+ entry into ameloblasts through Orai1, therefore, plays a central role in enamel production. Potential effects of Orai1 inhibitors on enamel production will need to be carefully monitored, particularly in children when such compounds are used in the clinic.

Sweat glands

Anhidrosis

Eccrine sweat glands are found dispersed throughout human skin, where they play an important role in the regulation of sweating and thermoregulation. Patients with LoF mutations in STIM1 or Orai1 exhibit marked anhidrosis [23]. In normal sweat glands, Ca2+ entry through Orai1 channels activates TMEM16A, a Ca2+-activated Cl- channel in the plasma membrane (Figure 5). Cl- efflux from the cell provides the driving force for water flow through aquaporins.

Salt secretion in eccrine sweat glands requires Orai1.
Figure 5:
Salt secretion in eccrine sweat glands requires Orai1.

Stimulation of muscarinic receptors on the basolateral membrane increases InsP3, leading to Ca2+ release from the ER. Store depletion opens Orai1 channels in the apical membrane. Ca2+ entry through the channels stimulates Ca2+-activated Cl- channels (TMEM16A), which results in Cl- secretion across the apical membrane. Cl- levels in the cytosol are maintained by electroneutral Na+- and K+-coupled Cl- co-transport (NKCC1) in the basolateral membrane. Na+ is expelled by the Na+-K+ ATPase pump, and the increase in cytosolic K+ then exits through basolateral K+ channels. Na+ is secreted transcellularly through the paracellular pathway, driven by the small negative potential in the lumen of the gland (apical side). Water follows through aquaporin channels.

Figure 5:
Salt secretion in eccrine sweat glands requires Orai1.

Stimulation of muscarinic receptors on the basolateral membrane increases InsP3, leading to Ca2+ release from the ER. Store depletion opens Orai1 channels in the apical membrane. Ca2+ entry through the channels stimulates Ca2+-activated Cl- channels (TMEM16A), which results in Cl- secretion across the apical membrane. Cl- levels in the cytosol are maintained by electroneutral Na+- and K+-coupled Cl- co-transport (NKCC1) in the basolateral membrane. Na+ is expelled by the Na+-K+ ATPase pump, and the increase in cytosolic K+ then exits through basolateral K+ channels. Na+ is secreted transcellularly through the paracellular pathway, driven by the small negative potential in the lumen of the gland (apical side). Water follows through aquaporin channels.

Close modal

In mice, eccrine sweat glands are found only on the foot pads. Cholinergic stimulation of the hind paws resulted in an increase in the number of open sweat pores in wildtype mice, but very few opened in animals in which Orai1 had been deleted in tissues of ectodermal origin, including sweat glands [60]. Interestingly, sweat glands had normal morphology in the Orai1 knockout mice, but each lumen was narrower. The finding that patients with LoF mutations in Orai1 present with anhidrosis suggests that an intimate association between Orai1 and TMEM16A exists, because other sources of Ca2+ cannot substitute for Orai1.

Skin

Psoriasis

Psoriasis vulgaris is a chronic inflammatory skin disorder that affects ~2–4% of the global population and manifests as marked plaque-like skin lesions that significantly reduce quality of life. The lesions arise from epidermal hyperplasia and immune cell infiltration, but the mechanisms involved are only partly understood. Psoriasis is driven, in part, by a Th-17 specific immune response [61]. Dendritic cells and keratinocytes overproduce IL-23, which then stimulates various cell types within the dermis, including Th-17 cells, mast cells and macrophages. These cells then release pro-inflammatory cytokines, including IL-17A and TNF-α, which trigger keratinocyte hyperproliferation, as well as secretion of chemokines that recruit neutrophils. Keratinocytes and skin immune cells express STIM and Orai proteins, and Orai1 is required for keratinocyte proliferation and polarised motility [62]. The CRAC channel blocker BTP2 inhibited contact hypersensitivity to the dermatitis-inducing allergen TNCB, as well as delayed type hypersensitivity to sheep red blood cells [63], which is often used as a model of dermatological autoimmune disease. Two CRAC channel blockers were reported to reduce serum cytokine levels and psoriatic plaques in a mouse model of psoriasis [64].

In another study, pathogenic Th17 cells were generated in mice following T cell-specific expression of a hyperactive form of the transcription factor STAT3C [65]. This led to psoriasis-like skin inflammation, hyperkeratosis and psoriatic arthritis. Importantly, these phenotypes were prevented by STIM1 deletion in T cells, resulting in a loss of store-operated Ca2+ entry [65]. Therefore, CRAC channels are required for Th17-dependent psoriatic lesions.

Autoimmune disease

Primary Sjögren’s disease

Primary Sjögren’s disease is an autoimmune disease that affects 0.5–1% of the population and disproportionately affects females [66]. Hallmarks include exocrinopathy, leading to dryness of the mouth and eyes due to dysfunction of salivary and lacrimal glands, general fatigue and joint pain. In primary Sjögren’s syndrome, the salivary and lacrimal glands exhibit prominent inflammation, lymphocyte infiltration and ultimately are damaged to the extent that function is heavily impaired [66].

A mouse with conditional knockout of STIM1 and STIM2 genes in T cells showed spontaneous and progressive submandibular gland inflammation, typically within 3 months, marked salivary gland damage, loss of stimulated fluid secretion, lymphocyte invasion of the glands and elevated primary Sjögren’s disease-specific autoantibodies [67]. Peripheral blood mononuclear cells from patients with primary Sjögren’s disease had reduced STIM protein expression and substantially reduced store-operated Ca2+ entry compared with healthy volunteers [67]. It was suggested that dysregulation of STIM protein expression with subsequent aberrations in store-operated Ca2+ entry in T lymphocytes underlies the pathogenesis of primary Sjögren’s syndrome [67]. A recent study used a K14Cre driver to specifically delete STIM1 and STIM2 in the ectoderm and its derivatives, which includes salivary glands [68]. Store-operated Ca2+ entry was decreased in parotid acini from STIM1/2-deficient mice. In the acini, store-operated Ca2+ entry activates ANO1, a Ca2+-activated Cl- channel, resulting in Cl- secretion into the lumen of the gland. This generates a negative electrical potential, which draws Na+ into the lumen through the paracellular pathway and water through aquaporin 5 channels. The reduction in store-operated Ca2+ entry in the STIM-deficient animals, therefore, resulted in less Cl- and Na+ secretion, and consequently less saliva production compared with controls. It is important to note that in this study [68], STIM had been deleted in secretory epithelial cells and not T cells. Therefore, the work is more relevant to non-autoimmune Sicca’s syndrome but not Sjögren’s disease. A recent rigorous study has demonstrated that regulatory T cells (Tregs) are particularly important in this context [69]. Deletion of STIM1/2 specifically from Foxp3+ Tregs in mice fully recapitulated all the features of primary Sjögren’s disease [69]. The animals showed salivary and lacrimal gland inflammation, marked lymphocyte infiltration and increased autoantibodies in serum, and met all classification criteria for human disease. The disease was caused by interferon gamma-producing CD4+ T cells. Inhibition of interferon signalling through application of a JAK1/2 inhibitor reduced CD4+ T cell-induced Sjögren’s disease [69].

Skeletal muscle

Tubular aggregate myopathy and Stormorken’s syndrome

CRAC channels play a crucial, albeit indirect, role in supporting skeletal muscle contraction through preventing fatigue, regulating metabolism and supporting muscle development, growth and regeneration. Several dominant GoF mutations in Orai1 and STIM1 have been identified in patients with tubular aggregate myopathy (TAM) and Stormorken syndrome (STRMK) [70]. TAM is primarily characterised by progressive muscle weakness, exercise intolerance and tubular aggregates (TAs). The latter are formed by densely packed tubules derived from the sarcoplasmic reticulum (SR) in skeletal muscle. In contrast, STRMK presents a broader multisystemic disorder, including thrombocytopenia, miosis, hypersplenism, ichthyosis, short stature and dyslexia, alongside muscle weakness and TAs. Six mutations in the transmembrane domains of Orai1 (S97C, G98S, V107M, L138F, T184M and P245L) and 22 in different STIM1 domains [71] (H72Q, N80T, G81D, D84E/G, S88G, L92V, L96V, Y98C, K104N, F108I/L, H109N/R/Y, I115F, V138I, L303P, R304Q/W, K365N and I484RfsX21) have been found in TAM/STRMK patients (Figure 6). These STIM1/Orai1 GoF mutations result in constitutive Ca²+ entry and enhanced store-operated Ca2+ influx, even in the absence of SR Ca²+ store depletion. The most prevalent STIM1 mutation, R304W, has been observed in STRMK patients from 12 unrelated families, and corresponding knock-in mouse models suggest a central role for dysregulated Ca2+ entry in disease aetiology [72,73]. While most known STIM1 TAM/STRMK-linked mutations affect STIM1 activation by releasing the conformational lock and resulting in Orai1-mediated Ca2+ entry even under resting conditions, one exception involves a STIM1 frameshift mutation (I484RfsX21) that lifts the inactivation brake of the CRAC channel [74]. The TAM-associated STIM1I484R mutation increases ORAI1 channel function due to a reduced STIM1 inactivation break and an absence of microtubule trapping. None of the STIM1 TAM/STRMK GoF mouse models (Stim1R304W/+, Stim1I115F/+ and Stim1D84G/+) exhibit TAs, despite displaying other clinical hallmarks such as muscle weakness [71]. However, the very recently developed Orai1 TAM mice models (Orai1G100S/+ and Orai1V109M/+), equivalent to human G98S and V107M, showed the presence of TAs along with muscle weakness [75,76]. Constitutive Ca²+ entry was observed in muscle cells during early development of Orai1G100S/+ mice but was abolished in adulthood; store-operated Ca2+ entry (SOCE) was significantly reduced, likely due to decreased Orai1 expression, and mitochondrial dysfunction was also observed, possibly linking Orai1 activity to mitochondrial damage.

Human Orai1 and STIM1 GoF mutations in TAM are shown.
Figure 6:
Human Orai1 and STIM1 GoF mutations in TAM are shown.

Human Orai1 model was modified from [20]. Only two Orai1 subunits are shown for simplicity. GoF, gain-of-function; TAM, tubular aggregate myopathy. PM plasma membrane, Ext. extracellular, Int intracellular.

Figure 6:
Human Orai1 and STIM1 GoF mutations in TAM are shown.

Human Orai1 model was modified from [20]. Only two Orai1 subunits are shown for simplicity. GoF, gain-of-function; TAM, tubular aggregate myopathy. PM plasma membrane, Ext. extracellular, Int intracellular.

Close modal

While the precise mechanism of TAs formation remains unclear, these structures may play a protective role in TAM/STRMK muscle by trapping misfolded proteins and excess Ca2+, thereby reducing cellular stress and preventing myofibre damage [70]. However, caution is needed when analysing muscle phenotypes in transgenic mice, as TAs, identical with those in human diseases, can be naturally found in muscle fibres of male inbred mice and are influenced by age and sex [77]. Mutations in CASQ1 and RYR1 have also been linked to TAM [71].

Orai1 has garnered significant attention as a major therapeutic target for TAM/STRMK. Decreasing the expression of Orai1 in Stim1R304W/+ mice by crossing it with Orai1+/- mice improved the multi-systemic TAM/STRMK phenotype [78]. Another recent study showed that crossing the STIM1R304W/+ mouse model with the partly obstructed ORAI1R93W/+ variant improved muscle function and alleviated multisystemic abnormalities [79]. In cellulo, the constitutive Ca2+ influx through most of the human Orai1 TAM/STRMK mutants (except the G98S Orai1) could be blocked by sub-micromolar concentrations of the CRAC channel inhibitor GSK-7975A [80]. Newly discovered SOCE modulators, CIC-37 and CIC-39, were able to inhibit Ca2+ influx through various STIM1/Orai1 GoF TAM/STRMK mutants including Orai1 G98S [81]. Stim1 I115F GoF mice treated with CIC-39 restored platelet count and prevented abnormal bleeding, offering a potential treatment for thrombocytopenia in the TAM patients [82].

STIM and Orai1 proteins are closely linked to a variety of human diseases affecting multiple organ systems. While some conditions, such as severe combined immunodeficiency (SCID), TAM and ectodermal dysplasia, arise from LoF or GoF mutations, others, including acute pancreatitis and allergic asthma, are characterised by excessive Ca2+ influx through otherwise unmutated channels. In many of these diseases, the pathology is not driven by a single homogeneous cell type; rather, multiple cell types, particularly immune cells, play significant roles.

In this context, the widespread distribution of CRAC channels, initially seen as a potential source of severe off-target effects for CRAC channel-focused therapies, could offer advantages. Given that CRAC channel-driven cross-talk between various cell types accelerates disease progression, evident in the positive feedback loops between airway epithelia, diverse lung immune cells and smooth muscle, partial inhibition of CRAC channels across all cell types may exert a supra-linear suppressive effect. This concept has already been demonstrated in vitro [83]. By partially inhibiting CRAC channels, the risk of off-target effects is reduced, as is the likelihood of globally impairing CRAC channel activity.

Perspectives

  • Store-operated CRAC channels serve as a key pathway for Ca²⁺ entry in most cell types and have been implicated in a growing number of human diseases. Understanding how CRAC channels contribute to organ dysfunction across different physiological systems could unveil commonalities across various pathological conditions.

  • A recurring theme emerging from studies of seemingly unrelated disorders is that CRAC channel activity is altered in multiple cell types within an organ, with tissue-resident immune cells playing a particularly prominent role.

  • As a result, targeting CRAC channels represents an appealing therapeutic approach, as it could simultaneously affect various cell types, potentially mitigating the severity of the disease.

The author declares no competing interest.

This work was supported by an Intramural Research Program of the NIH, National Institute of Environmental Health Sciences.

This article has been published open access under our Subscribe to Open programme, made possible through the support of our subscribing institutions, learn more here: https://portlandpress.com/pages/open_access_options_and_prices#conditional

RB: Writing - Original Draft, Writing - Review & Editing, Visualisation; ABP Conceptualisation, Resources, Supervision, Visualisation, Writing - Original Draft, Writing - Review & Editing

CAD

channel-activating domain

CRAC

Ca2+ release-activated Ca2+

ER

endoplasmic reticulum

GoF

gain-of-function

ILC2

innate lymphoid type 2

LoF

loss-of-function

MCU

mitochondrial Ca²+ uniporter

NFAT

nuclear factor of activated T cell

PACs

pancreatic acinar cells

PARs

protease-activated receptors

SARAF

store-operated calcium entry-associated regulatory factor

SCID

severe combined immunodeficiency

SERCA

Sarco-endoplasmic reticulum Ca2+ ATPase

SOAR

STIM Orai-activating region

SR

sarcoplasmic reticulum

STIM

Stromal Interaction Molecule

STIM

Stromal Interaction Molecule

STRMK

Stormorken syndrome

TAM

tubular aggregate myopathy

TM

transmembrane

1
Clapham
,
D.E
. (
2007
)
Calcium signaling
.
Cell
131
,
1047
1058
https://doi.org/10.1016/j.cell.2007.11.028
2
Parekh
,
A.B
. (
2011
)
Decoding cytosolic Ca2+ oscillations
.
Trends Biochem. Sci.
36
,
78
87
https://doi.org/10.1016/j.tibs.2010.07.013
3
Berridge
,
M.J
. (
2016
)
The inositol trisphosphate/calcium signaling pathway in health and disease
.
Physiol. Rev.
96
,
1261
1296
https://doi.org/10.1152/physrev.00006.2016
4
De Stefani
,
D.
,
Rizzuto
,
R.
and
Pozzan
,
T
. (
2016
)
Enjoy the trip: calcium in mitochondria back and forth
.
Annu. Rev. Biochem.
85
,
161
192
https://doi.org/10.1146/annurev-biochem-060614-034216
5
Bakowski
,
D.
,
Murray
,
F.
and
Parekh
,
A.B
. (
2021
)
Store-operated Ca2+ channels: mechanism, function, pharmacology, and therapeutic targets
.
Annu. Rev. Pharmacol. Toxicol.
61
,
629
654
https://doi.org/10.1146/annurev-pharmtox-031620-105135
6
Prakriya
,
M.
and
Lewis
,
R.S
. (
2015
)
Store-operated calcium channels
.
Physiol. Rev.
95
,
1383
1436
https://doi.org/10.1152/physrev.00020.2014
7
Hoth
,
M.
and
Penner
,
R
. (
1992
)
Depletion of intracellular calcium stores activates a calcium current in mast cells
.
Nature
355
,
353
356
https://doi.org/10.1038/355353a0
8
Feske
,
S.
,
Gwack
,
Y.
,
Prakriya
,
M.
,
Srikanth
,
S.
,
Puppel
,
S.H.
,
Tanasa
,
B.
et al.
(
2006
)
A mutation in Orai1 causes immune deficiency by abrogating CRAC channel function
.
Nature
441
,
179
185
https://doi.org/10.1038/nature04702
9
Prakriya
,
M.
,
Feske
,
S.
,
Gwack
,
Y.
,
Srikanth
,
S.
,
Rao
,
A.
and
Hogan
,
P.G
. (
2006
)
Orai1 is an essential pore subunit of the CRAC channel
.
Nature
443
,
230
233
https://doi.org/10.1038/nature05122
10
Yeromin
,
A.V.
,
Zhang
,
S.L.
,
Jiang
,
W.
,
Yu
,
Y.
,
Safrina
,
O.
and
Cahalan
,
M.D
. (
2006
)
Molecular identification of the CRAC channel by altered ion selectivity in a mutant of Orai
.
Nature
443
,
226
229
https://doi.org/10.1038/nature05108
11
Vig
,
M.
,
Beck
,
A.
,
Billingsley
,
J.M.
,
Lis
,
A.
,
Parvez
,
S.
,
Peinelt
,
C.
et al.
(
2006
)
CRACM1 multimers form the ion-selective pore of the CRAC channel
.
Curr. Biol.
16
,
2073
2079
https://doi.org/10.1016/j.cub.2006.08.085
12
Bhardwaj
,
R.
,
Hediger
,
M.A.
and
Demaurex
,
N
. (
2016
)
Redox modulation of STIM-ORAI signaling
.
Cell Calcium
60
,
142
152
https://doi.org/10.1016/j.ceca.2016.03.006
13
Roos
,
J.
,
DiGregorio
,
P.J.
,
Yeromin
,
A.V.
,
Ohlsen
,
K.
,
Lioudyno
,
M.
,
Zhang
,
S.
et al.
(
2005
)
STIM1, an essential and conserved component of store-operated Ca2+ channel function
.
J. Cell Biol.
169
,
435
445
https://doi.org/10.1083/jcb.200502019
14
Liou
,
J.
,
Kim
,
M.L.
,
Heo
,
W.D.
,
Jones
,
J.T.
,
Myers
,
J.W.
,
Ferrell
,
J.E.
et al.
(
2005
)
STIM is a Ca2+ sensor essential for Ca2+-store-depletion-triggered Ca2+ influx
.
Curr. Biol.
15
,
1235
1241
https://doi.org/10.1016/j.cub.2005.05.055
15
Fröhlich
,
M.
,
Söllner
,
J.
and
Derler
,
I
. (
2024
)
Insights into the dynamics of the Ca2+ release-activated Ca2+ channel pore-forming complex Orai1
.
Biochem. Soc. Trans.
52
,
747
760
https://doi.org/10.1042/BST20230815
16
Yeung
,
P.S.W.
,
Yamashita
,
M.
and
Prakriya
,
M
. (
2020
)
Molecular basis of allosteric Orai1 channel activation by STIM1
.
J. Physiol.
598
,
1707
1723
https://doi.org/10.1113/JP276550
17
Hou
,
X.
,
Pedi
,
L.
,
Diver
,
M.M.
and
Long
,
S.B
. (
2012
)
Crystal structure of the calcium release-activated calcium channel Orai
.
Science
338
,
1308
1313
https://doi.org/10.1126/science.1228757
18
Tiffner
,
A.
,
Maltan
,
L.
,
Weiß
,
S.
and
Derler
,
I
. (
2021
)
The Orai pore opening mechanism
.
Int. J. Mol. Sci.
22
, 533 https://doi.org/10.3390/ijms22020533
19
Bhardwaj
,
R.
,
Augustynek
,
B.S.
,
Ercan-Herbst
,
E.
,
Kandasamy
,
P.
,
Seedorf
,
M.
and
Peinelt
,
C
. (
2020
)
Ca2+/calmodulin binding to STIM1 hydrophobic residues facilitates slow Ca2+-dependent inactivation of the Orai1 channel
.
Cell. Physiol. Biochem.
54
,
252
270
https://doi.org/10.33594/000000218
20
Augustynek
,
B.
,
Gyimesi
,
G.
,
Dernič
,
J.
,
Sallinger
,
M.
,
Albano
,
G.
,
Klesse
,
G.J.
et al.
(
2022
)
Discovery of novel gating checkpoints in the Orai1 calcium channel by systematic analysis of constitutively active mutants of its paralogs and orthologs
.
Cell Calcium
105
, e102616 https://doi.org/10.1016/j.ceca.2022.102616
21
Gold
,
M.G.
,
Stengel
,
F.
,
Nygren
,
P.J.
,
Weisbrod
,
C.R.
,
Bruce
,
J.E.
,
Robinson
,
C.V.
et al.
(
2011
)
Architecture and dynamics of an A-kinase anchoring protein 79 (AKAP79) signaling complex
.
Proc. Natl. Acad. Sci. U.S.A.
108
,
6426
6431
https://doi.org/10.1073/pnas.1014400108
22
Kar
,
P.
,
Lin
,
Y.P.
,
Bhardwaj
,
R.
,
Tucker
,
C.J.
,
Bird
,
G.S.
,
Hediger
,
M.A.
et al.
(
2021
)
The N terminus of Orai1 couples to the AKAP79 signaling complex to drive NFAT1 activation by local Ca2+ entry
.
Proc. Natl. Acad. Sci. U.S.A.
118
, e2012908118 https://doi.org/10.1073/pnas.2012908118
23
Feske
,
S
. (
2010
)
CRAC channelopathies
.
Pflugers Arch.
460
,
417
435
https://doi.org/10.1007/s00424-009-0777-5
24
Lian
,
J.
,
Cuk
,
M.
,
Kahlfuss
,
S.
,
Kozhaya
,
L.
,
Vaeth
,
M.
,
Rieux-Laucat
,
F
, et al.
(
2018
)
ORAI1 mutations abolishing store-operated Ca2+ entry cause anhidrotic ectodermal dysplasia with immunodeficiency
.
J. Allergy Clin. Immunol.
142
,
1297
1310
https://doi.org/10.1016/j.jaci.2017.10.031
25
Feske
,
S
. (
2019
)
CRAC channels and disease - from human CRAC channelopathies and animal models to novel drugs
.
Cell Calcium
80
,
112
116
https://doi.org/10.1016/j.ceca.2019.03.004
26
Mallory
,
A.
and
Kern
,
F.
(
1980
)
Drug-induced pancreatitis: a critical review
.
Gastroenterology
78
,
813
820
https://doi.org/10.1016/0016-5085(80)90689-7
27
Gerasimenko
,
J.V.
,
Gerasimenko
,
O.V.
and
Petersen
,
O.H
. (
2014
)
The role of Ca2+ in the pathophysiology of pancreatitis
.
J. Physiol.
592
,
269
280
https://doi.org/10.1113/jphysiol.2013.261784
28
Raraty
,
M.
,
Ward
,
J.
,
Erdemli
,
G.
,
Vaillant
,
C.
,
Neoptolemos
,
J.P.
,
Sutton
,
R.
et al.
(
2000
)
Calcium-dependent enzyme activation and vacuole formation in the apical granular region of pancreatic acinar cells
.
Proc. Natl. Acad. Sci. U.S.A.
97
,
13126
13131
https://doi.org/10.1073/pnas.97.24.13126
29
Gerasimenko
,
J.V.
,
Gryshchenko
,
O.
,
Ferdek
,
P.E.
,
Stapleton
,
E.
,
Hébert
,
T.O.G.
,
Bychkova
,
S.
et al.
(
2013
)
Ca2+ release-activated Ca2+ channel blockade as a potential tool in antipancreatitis therapy
.
Proc. Natl. Acad. Sci. U.S.A.
110
,
13186
13191
https://doi.org/10.1073/pnas.1300910110
30
Wen
,
L.
,
Voronina
,
S.
,
Javed
,
M.A.
,
Awais
,
M.
,
Szatmary
,
P.
,
Latawiec
,
D
, et al.
(
2015
)
Inhibitors of ORAI1 prevent cytosolic calcium-associated injury of human pancreatic acinar cells and acute pancreatitis in 3 mouse models
.
Gastroenterology
149
,
481
492
https://doi.org/10.1053/j.gastro.2015.04.015
31
Szabó
,
V.
,
Csákány-Papp
,
N.
,
Görög
,
M.
,
Madácsy
,
T.
,
Varga
,
Á.
,
Kiss
,
A.
et al.
(
2023
)
Orai1 calcium channel inhibition prevents progression of chronic pancreatitis
.
JCI Insight
8
, e167645 https://doi.org/10.1172/jci.insight.167645
32
Bruen
,
C.
,
Miller
,
J.
,
Wilburn
,
J.
,
Mackey
,
C.
,
Bollen
,
T.L.
,
Stauderman
,
K.
et al.
(
2021
)
Auxora for the treatment of patients with acute pancreatitis and accompanying systemic inflammatory response syndrome: clinical development of a calcium release-activated calcium channel inhibitor
.
Pancreas
50
,
537
543
https://doi.org/10.1097/MPA.0000000000001793
33
Lewis
,
S.
,
Evans
,
D.L.
,
Tsugorka
,
T.T.
,
Peng
,
S.
,
Stauderman
,
K.
,
Gerasimenko
,
O.
et al.
(
2024
)
Combination of the CRAC channel inhibitor CM4620 and galactose as a potential therapy for acute pancreatitis
.
Function (Oxf)
5
, zqae017 https://doi.org/10.1093/function/zqae017
34
Waldron
,
R.T.
,
Chen
,
Y.
,
Pham
,
H.
,
Go
,
A.
,
Su
,
H.Y.
,
Hu
,
C.
et al.
(
2019
)
The Orai Ca2+ channel inhibitor CM4620 targets both parenchymal and immune cells to reduce inflammation in experimental acute pancreatitis
.
J. Physiol.
597
,
3085
3105
https://doi.org/10.1113/JP277856
35
Mei
,
W.
,
Zhang
,
X.
,
Niu
,
M.
,
Li
,
L.
,
Guo
,
X.
,
Wang
,
G
, et al.
(
2024
)
Deletion of myeloid-specific Orai1 calcium channel does not affect pancreatic tissue damage in experimental acute pancreatitis
.
Pancreatology
24
,
528
537
https://doi.org/10.1016/j.pan.2024.04.001
36
Pallagi
,
P.
,
Görög
,
M.
,
Papp
,
N.
,
Madácsy
,
T.
,
Varga
,
Á.
,
Crul
,
T.
et al.
(
2022
)
Bile acid‐ and ethanol‐mediated activation of Orai1 damages pancreatic ductal secretion in acute pancreatitis
.
J. Physiol.
600
,
1631
1650
https://doi.org/10.1113/JP282203
37
Son
,
A.
,
Ahuja
,
M.
,
Schwartz
,
D.M.
,
Varga
,
A.
,
Swaim
,
W.
,
Kang
,
N
, et al.
(
2019
)
Ca2+ influx channel inhibitor SARAF protects mice from acute pancreatitis
.
Gastroenterology
157
,
1660
1672
https://doi.org/10.1053/j.gastro.2019.08.042
38
Schwarz
,
A.
,
Tutsch
,
E.
,
Ludwig
,
B.
,
Schwarz
,
E.C.
,
Stallmach
,
A.
and
Hoth
,
M
. (
2004
)
Ca2+ signaling in identified T-lymphocytes from human intestinal mucosa. Relation to hyporeactivity, proliferation, and inflammatory bowel disease
.
J. Biol. Chem.
279
,
5641
5647
https://doi.org/10.1074/jbc.M309317200
39
Liang
,
X.
,
Xie
,
J.
,
Liu
,
H.
,
Zhao
,
R.
,
Zhang
,
W.
,
Wang
,
H
, et al.
(
2022
)
STIM1 deficiency in intestinal epithelium attenuates colonic inflammation and tumorigenesis by reducing ER stress of goblet cells
.
Cell. Mol. Gastroenterol. Hepatol.
14
,
193
217
https://doi.org/10.1016/j.jcmgh.2022.03.007
40
Di Sabatino
,
A.
,
Rovedatti
,
L.
,
Kaur
,
R.
,
Spencer
,
J.P.
,
Brown
,
J.T.
,
Morisset
,
V.D.
et al.
(
2009
)
Targeting gut T cell Ca2+ release-activated Ca2+ channels inhibits T cell cytokine production and T-box transcription factor T-bet in inflammatory bowel disease
.
J. Immunol.
183
,
3454
3462
https://doi.org/10.4049/jimmunol.0802887
41
McCarl
,
C.A.
,
Khalil
,
S.
,
Ma
,
J.
,
Oh-hora
,
M.
,
Yamashita
,
M.
,
Roether
,
J.
et al.
(
2010
)
Store-operated Ca2+ entry through ORAI1 is critical for T cell-mediated autoimmunity and allograft rejection
.
J. Immunol.
185
,
5845
5858
https://doi.org/10.4049/jimmunol.1001796
42
Letizia
,
M.
,
Wang
,
Y.H.
,
Kaufmann
,
U.
,
Gerbeth
,
L.
,
Sand
,
A.
,
Brunkhorst
,
M.
et al.
(
2022
)
Store-operated calcium entry controls innate and adaptive immune cell function in inflammatory bowel disease
.
EMBO Mol. Med.
14
, e15687 https://doi.org/10.15252/emmm.202215687
43
Holgate
,
S.T.
,
Wenzel
,
S.
,
Postma
,
D.S.
,
Weiss
,
S.T.
,
Renz
,
H.
and
Sly
,
P.D
. (
2015
)
Asthma
.
Nat. Rev. Dis. Primers
1
, 15025 https://doi.org/10.1038/nrdp.2015.25
44
Jacquet
,
A
. (
2023
)
The HDM allergen orchestra and its cysteine protease maestro: stimulators of kaleidoscopic innate immune responses
.
Mol. Immunol.
156
,
48
60
https://doi.org/10.1016/j.molimm.2023.03.002
45
Holgate
,
S.T.
and
Polosa
,
R
. (
2008
)
Treatment strategies for allergy and asthma
.
Nat. Rev. Immunol.
8
,
218
230
https://doi.org/10.1038/nri2262
46
Gregory
,
L.G.
and
Lloyd
,
C.M
. (
2011
)
Orchestrating house dust mite-associated allergy in the lung
.
Trends Immunol.
32
,
402
411
https://doi.org/10.1016/j.it.2011.06.006
47
Parekh
,
A.B
. (
2024
)
House dust mite allergens, store-operated Ca(2+) channels and asthma
.
J. Physiol
602
,
6021
6038
https://doi.org/10.1113/JP284931
48
Johnson
,
M.T.
,
Xin
,
P.
,
Benson
,
J.C.
,
Pathak
,
T.
,
Walter
,
V.
,
Emrich
,
S.M.
et al.
(
2022
)
STIM1 is a core trigger of airway smooth muscle remodeling and hyperresponsiveness in asthma
.
Proc. Natl. Acad. Sci. U.S.A.
119
, e2114557118 https://doi.org/10.1073/pnas.2114557118
49
Jairaman
,
A.
,
Maguire
,
C.H.
,
Schleimer
,
R.P.
and
Prakriya
,
M
. (
2016
)
Allergens stimulate store-operated calcium entry and cytokine production in airway epithelial cells
.
Sci. Rep.
6
, 32311 https://doi.org/10.1038/srep32311
50
Lin
,
Y.P.
,
Nelson
,
C.
,
Kramer
,
H.
and
Parekh
,
A.B
. (
2018
)
The allergen der p3 from house dust mite stimulates store-operated Ca2+ channels and mast cell migration through PAR4 receptors
.
Mol. Cell
70
,
228
241
https://doi.org/10.1016/j.molcel.2018.03.025
51
Samanta
,
K.
,
Bakowski
,
D.
and
Parekh
,
A.B
. (
2014
)
Key role for store-operated Ca2+ channels in activating gene expression in human airway bronchial epithelial cells
.
PLoS ONE
9
, e105586 https://doi.org/10.1371/journal.pone.0105586
52
Yoshino
,
T.
,
Ishikawa
,
J.
,
Ohga
,
K.
,
Morokata
,
T.
,
Takezawa
,
R.
,
Morio
,
H.
et al.
(
2007
)
YM-58483, a selective CRAC channel inhibitor, prevents antigen-induced airway eosinophilia and late phase asthmatic responses via Th2 cytokine inhibition in animal models
.
Eur. J. Pharmacol.
560
,
225
233
https://doi.org/10.1016/j.ejphar.2007.01.012
53
Kaur
,
M.
,
Birrell
,
M.A.
,
Dekkak
,
B.
,
Reynolds
,
S.
,
Wong
,
S.
,
De Alba
,
J
, et al.
(
2015
)
The role of CRAC channel in asthma
.
Pulm. Pharmacol. Ther.
35
,
67
74
https://doi.org/10.1016/j.pupt.2015.09.002
54
Sutovska
,
M.
,
Kocmalova
,
M.
,
Franova
,
S.
,
Vakkalanka
,
S.
and
Viswanadha
,
S
. (
2016
)
Pharmacodynamic evaluation of RP3128, a novel and potent CRAC channel inhibitor in guinea pig models of allergic asthma
.
Eur. J. Pharmacol.
772
,
62
70
https://doi.org/10.1016/j.ejphar.2015.12.047
55
Wang
,
Y.H.
,
Noyer
,
L.
,
Kahlfuss
,
S.
,
Raphael
,
D.
,
Tao
,
A.Y.
,
Kaufmann
,
U.
et al.
(
2022
)
Distinct roles of ORAI1 in T cell-mediated allergic airway inflammation and immunity to influenza A virus infection
.
Sci. Adv.
8
, eabn6552 https://doi.org/10.1126/sciadv.abn6552
56
Howard
,
E.
,
Hurrell
,
B.P.
,
Helou
,
D.G.
,
Shafiei-Jahani
,
P.
,
Hasiakos
,
S.
,
Painter
,
J.
et al.
(
2023
)
Orai inhibition modulates pulmonary ILC2 metabolism and alleviates airway hyperreactivity in murine and humanized models
.
Nat. Commun.
14
, 5989 https://doi.org/10.1038/s41467-023-41065-4
57
Lacruz
,
R.S.
and
Feske
,
S
. (
2015
)
Diseases caused by mutations in ORAI1 and STIM1
.
Ann. N. Y. Acad. Sci.
1356
,
45
79
https://doi.org/10.1111/nyas.12938
58
Eckstein
,
M.
,
Vaeth
,
M.
,
Fornai
,
C.
,
Vinu
,
M.
,
Bromage
,
T.G.
,
Nurbaeva
,
M.K.
et al.
(
2017
)
Store-operated Ca2+ entry controls ameloblast cell function and enamel development
.
JCI Insight
2
, e91166 https://doi.org/10.1172/jci.insight.91166
59
Eckstein
,
M.
,
Vaeth
,
M.
,
Aulestia
,
F.J.
,
Costiniti
,
V.
,
Kassam
,
S.N.
,
Bromage
,
T.G.
et al.
(
2019
)
Differential regulation of Ca2+ influx by ORAI channels mediates enamel mineralization
.
Sci. Signal.
12
, eaav4663 https://doi.org/10.1126/scisignal.aav4663
60
Concepcion
,
A.R.
,
Vaeth
,
M.
,
Wagner
,
L.E.
,
Eckstein
,
M.
,
Hecht
,
L.
,
Yang
,
J
, et al.
(
2016
)
Store-operated Ca2+ entry regulates Ca2+-activated chloride channels and eccrine sweat gland function
.
J. Clin. Invest.
126
,
4303
4318
https://doi.org/10.1172/JCI89056
61
Bugaut
,
H.
and
Aractingi
,
S
. (
2021
)
Major role of the IL17/23 axis in psoriasis supports the development of new targeted therapies
.
Front. Immunol.
12
, 621956 https://doi.org/10.3389/fimmu.2021.621956
62
Vandenberghe
,
M.
,
Raphaël
,
M.
,
Lehen’kyi
,
V.
,
Gordienko
,
D.
,
Hastie
,
R.
,
Oddos
,
T.
et al.
(
2013
)
ORAI1 calcium channel orchestrates skin homeostasis
.
Proc. Natl. Acad. Sci. U.S.A.
110
,
E4839
E4848
https://doi.org/10.1073/pnas.1310394110
63
Ohga
,
K.
,
Takezawa
,
R.
,
Arakida
,
Y.
,
Shimizu
,
Y.
and
Ishikawa
,
J
. (
2008
)
Characterization of YM-58483/BTP2, a novel store-operated Ca2+ entry blocker, on T cell-mediated immune responses in vivo
.
Int. Immunopharmacol.
8
,
1787
1792
https://doi.org/10.1016/j.intimp.2008.08.016
64
Kong
,
X.
,
Wang
,
F.
,
Chen
,
Y.
,
Liang
,
X.
,
Yin
,
Y.
,
Liu
,
H
, et al.
(
2023
)
Molecular action mechanisms of two novel and selective calcium release-activated calcium channel antagonists
.
Int. J. Biol. Macromol.
253
,
126937
https://doi.org/10.1016/j.ijbiomac.2023.126937
65
Kaufmann
,
U.
,
Kahlfuss
,
S.
,
Yang
,
J.
,
Ivanova
,
E.
,
Koralov
,
S.B.
and
Feske
,
S
. (
2019
)
Calcium signaling controls pathogenic Th17 cell-mediated inflammation by regulating mitochondrial function
.
Cell Metab.
29
,
1104
1118
https://doi.org/10.1016/j.cmet.2019.01.019
66
Mariette
,
X.
and
Criswell
,
L.A
. (
2018
)
Primary Sjögren’s syndrome
.
N. Engl. J. Med.
378
,
931
939
https://doi.org/10.1056/NEJMcp1702514
67
Cheng
,
K.T.
,
Alevizos
,
I.
,
Liu
,
X.
,
Swaim
,
W.D.
,
Yin
,
H.
,
Feske
,
S.
et al.
(
2012
)
STIM1 and STIM2 protein deficiency in T lymphocytes underlies development of the exocrine gland autoimmune disease, Sjogren’s syndrome
.
Proc. Natl. Acad. Sci. U.S.A.
109
,
14544
14549
https://doi.org/10.1073/pnas.1207354109
68
Son
,
G.Y.
,
Zou
,
A.
,
Wahl
,
A.
,
Huang
,
K.T.
,
Zorgit
,
S.
,
Vinu
,
M.
et al.
(
2025
)
Loss of STIM1 and STIM2 in salivary glands disrupts ANO1 function but does not induce Sjogren’s disease
.
Function (Oxf)
6
, zqae047 https://doi.org/10.1093/function/zqae047
69
Wang
,
Y.H.
,
Li
,
W.
,
McDermott
,
M.
,
Son
,
G.Y.
,
Maiti
,
G.
,
Zhou
,
F.
et al.
(
2024
)
IFN-γ-producing TH1 cells and dysfunctional regulatory T cells contribute to the pathogenesis of Sjögren’s disease
.
Sci. Transl. Med.
16
, eado4856 https://doi.org/10.1126/scitranslmed.ado4856
70
Silva-Rojas
,
R.
,
Laporte
,
J.
and
Böhm
,
J
. (
2020
)
STIM1/ORAI1 loss-of-function and gain-of-function mutations inversely impact on SOCE and calcium homeostasis and cause multi-systemic mirror diseases
.
Front. Physiol.
11
, 604941 https://doi.org/10.3389/fphys.2020.604941
71
Protasi
,
F.
,
Girolami
,
B.
,
Roccabianca
,
S.
and
Rossi
,
D
. (
2023
)
Store-operated calcium entry: from physiology to tubular aggregate myopathy
.
Curr. Opin. Pharmacol.
68
,
102347
https://doi.org/10.1016/j.coph.2022.102347
72
Silva-Rojas
,
R.
,
Treves
,
S.
,
Jacobs
,
H.
,
Kessler
,
P.
,
Messaddeq
,
N.
,
Laporte
,
J.
et al.
(
2019
)
STIM1 over-activation generates a multi-systemic phenotype affecting the skeletal muscle, spleen, eye, skin, bones and immune system in mice
.
Hum. Mol. Genet.
28
,
1579
1593
https://doi.org/10.1093/hmg/ddy446
73
Gamage
,
T.H.
,
Lengle
,
E.
,
Gunnes
,
G.
,
Pullisaar
,
H.
,
Holmgren
,
A.
,
Reseland
,
J.E.
et al.
(
2020
)
STIM1 R304W in mice causes subgingival hair growth and an increased fraction of trabecular bone
.
Cell Calcium
85
, e102110 https://doi.org/10.1016/j.ceca.2019.102110
74
Kim
,
J.H.
,
Carreras-Sureda
,
A.
,
Didier
,
M.
,
Henry
,
C.
,
Frieden
,
M.
and
Demaurex
,
N
. (
2022
)
The TAM-associated STIM1I484R mutation increases ORAI1 channel function due to a reduced STIM1 inactivation break and an absence of microtubule trapping
.
Cell Calcium
105
, e102615 https://doi.org/10.1016/j.ceca.2022.102615
75
Zhao
,
N.
,
Michelucci
,
A.
,
Pietrangelo
,
L.
,
Malik
,
S.
,
Groom
,
L.
,
Leigh
,
J.
et al.
(
2024
)
An Orai1 gain-of-function tubular aggregate myopathy mouse model phenocopies key features of the human disease
.
EMBO J.
43
,
5941
5971
https://doi.org/10.1038/s44318-024-00273-4
76
Pérez-Guàrdia
,
L.
,
Lafabrie
,
E.
,
Diedhiou
,
N.
,
Spiegelhalter
,
C.
,
Laporte
,
J.
and
Böhm
,
J
. (
2024
)
A gain-of-function mutation in the Ca2+ channel ORAI1 causes Stormorken syndrome with tubular aggregates in mice
.
Cells
13
, 1829 https://doi.org/10.3390/cells13221829
77
Boncompagni
,
S.
,
Protasi
,
F.
and
Franzini-Armstrong
,
C
. (
2012
)
Sequential stages in the age-dependent gradual formation and accumulation of tubular aggregates in fast twitch muscle fibers: SERCA and calsequestrin involvement
.
Age (Dordr).
34
,
27
41
https://doi.org/10.1007/s11357-011-9211-y
78
Silva-Rojas
,
R.
,
Pérez-Guàrdia
,
L.
,
Lafabrie
,
E.
,
Moulaert
,
D.
,
Laporte
,
J.
and
Böhm
,
J
. (
2022
)
Silencing of the Ca2+ channel ORAI1 improves the multi-systemic phenotype of tubular aggregate myopathy (TAM) and Stormorken syndrome (STRMK) in mice
.
Int. J. Mol. Sci.
23
, 6968 https://doi.org/10.3390/ijms23136968
79
Silva-Rojas
,
R.
,
Pérez-Guàrdia
,
L.
,
Simon
,
A.
,
Djeddi
,
S.
,
Treves
,
S.
,
Ribes
,
A.
et al.
(
2024
)
ORAI1 inhibition as an efficient preclinical therapy for tubular aggregate myopathy and Stormorken syndrome
.
JCI Insight
9
, e174866 https://doi.org/10.1172/jci.insight.174866
80
Bulla
,
M.
,
Gyimesi
,
G.
,
Kim
,
J.H.
,
Bhardwaj
,
R.
,
Hediger
,
M.A.
,
Frieden
,
M.
et al.
(
2019
)
ORAI1 channel gating and selectivity is differentially altered by natural mutations in the first or third transmembrane domain
.
J. Physiol. (Lond.)
597
,
561
582
https://doi.org/10.1113/JP277079
81
Riva
,
B.
,
Pessolano
,
E.
,
Quaglia
,
E.
,
Cordero-Sanchez
,
C.
,
Bhela
,
I.P.
,
Topf
,
A.
et al.
(
2022
)
STIM1 and ORAI1 mutations leading to tubular aggregate myopathies are sensitive to the store-operated Ca2+-entry modulators CIC-37 and CIC-39
.
Cell Calcium
105
, e102605 https://doi.org/10.1016/j.ceca.2022.102605
82
Cordero-Sanchez
,
C.
,
Pessolano
,
E.
,
Riva
,
B.
,
Vismara
,
M.
,
Trivigno
,
S.M.G.
,
Clemente
,
N.
et al.
(
2022
)
CIC-39Na reverses the thrombocytopenia that characterizes tubular aggregate myopathy
.
Blood Adv.
6
,
4471
4484
https://doi.org/10.1182/bloodadvances.2021006378
83
Di Capite
,
J.
,
Nelson
,
C.
,
Bates
,
G.
and
Parekh
,
A.B
. (
2009
)
Targeting Ca2+ release-activated Ca2+ channel channels and leukotriene receptors provides a novel combination strategy for treating nasal polyposis
.
J. Allergy Clin. Immunol.
124
,
1014
21
. https://doi.org/10.1016/j.jaci.2009.08.030
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).