3,888
Views
87
CrossRef citations to date
0
Altmetric
Review

STIM and ORAI proteins in the nervous system

Pages 245-252 | Received 17 Jun 2015, Accepted 07 Jul 2015, Published online: 01 Sep 2015

Abstract

Stromal interaction molecules (STIM) 1 and 2 are sensors of the calcium concentration in the endoplasmic reticulum. Depletion of endoplasmic reticulum calcium stores activates STIM proteins which, in turn, bind and open calcium channels in the plasma membrane formed by the proteins ORAI1, ORAI2, and ORAI3. The resulting store-operated calcium entry (SOCE), mostly controlled by the principal components STIM1 and ORAI1, has been particularly characterized in immune cells. In the nervous system, all STIM and ORAI homologs are expressed. This review summarizes current knowledge on distribution and function of STIM and ORAI proteins in central neurons and glial cells, i.e. astrocytes and microglia. STIM2 is required for SOCE in hippocampal synapses and cortical neurons, whereas STIM1 controls calcium store replenishment in cerebellar Purkinje neurons. In microglia, STIM1, STIM2, and ORAI1 regulate migration and phagocytosis. The isoforms ORAI2 and ORAI3 are candidates for SOCE channels in neurons and astrocytes, respectively. Due to the role of SOCE in neuronal and glial calcium homeostasis, dysfunction of STIM and ORAI proteins may have consequences for the development of neurodegenerative disorders, such as Alzheimer's disease.

STIM and ORAI Proteins Mediate Store-Operated Ca2+ Entry

Increases in the concentration of intracellular Ca2+ ([Ca2+]i) control diverse functions in virtually all cell types. These Ca2+ signals differ spatially, temporally and in magnitude and are mediated by numerous channels, transporters and Ca2+-sensing proteins. Opening of Ca2+ channels in either the plasma membrane or the membrane of intracellular Ca2+ stores directly elevates [Ca2+]i. Ca2+ entry from extracellular space and Ca2+ release from endoplasmic reticulum (ER) operate independently or together to regulate different Ca2+-dependent processes, such as exocytosis of neurotransmitters, contraction of skeletal muscle, activation of T lymphocytes or contraction of smooth muscle cells. Signaling via many cell surface receptors involves the activation of Ca2+ release from ER via inositol-1,4,5-triphosphate (IP3).Citation1 Because a second phase of IP3-mediated Ca2+ mobilization was frequently observed and identified as Ca2+ influx from extracellular space,Citation2 the hypothesis of “capacitative Ca2+ entry” was formulated by Putney in 1986.Citation3 He proposed a receptor-regulated pathway for Ca2+ that enters the depleted Ca2+ pool directly via plasma and ER membranes and is controlled by IP3 and the Ca2+ content of the ER.

The capacitative or store-operated Ca2+ entry (SOCE), has been recognized as a an essential route for Ca2+ uptake in a wide variety of cell types to replenish intracellular Ca2+ stores and to regulate secretion, gene transcription and cell cycle progression.Citation4 Several candidate molecules and ion channels mediating SOCE were discussed in recent years. In particular, some members of the transient receptor potential (TRP) cation channel family were found to generate receptor-activated Ca2+ entry.Citation5 However, electrophysiological characterization of SOCE in mast cells revealed a “Ca2+-release activated Ca2+ (CRAC) current”,Citation6 whose properties can be clearly distinguished from those of TRP channels.Citation5,7 In 2005 and 2006, different groups identified stromal interaction molecules (STIM) and ORAI proteins (named after the keepers of heaven's gate in greek mythology) as essential components of SOCE.Citation8-15

Stromal interaction molecule 1 (STIM1) was originally identified as a potential tumor suppressor gene coding for a transmembrane protein.Citation16,17 In vertebrates, one additional STIM1-related gene, STIM2, was described.Citation18 Both STIM homologues are ubiquitously expressed in different cell types with higher STIM1 levels in most tissues but a predominant expression of STIM2 in brain.Citation18,19 STIM1 and STIM2 each contain highly conserved domains, including a single transmembrane segment, an EF-hand Ca2+-binding domain, a sterile α-motif (SAM) domain and coiled-coil regions (for review see refs. Citation20 and 21). STIM1 and STIM2 are primarily located to the ER; STIM1 was also detected in the plasma membrane.Citation22 The EF-hand Ca2+-binding domains (together with the SAM domains) of STIMs are responsible for sensing [Ca2+] in the lumen of the ER.Citation8,10,23 A decrease in ER luminal Ca2+ concentration results in dissociation of Ca2+ from the EF-hand domain which, in turn, triggers oligomerization and activation of STIM1, a process that is reversed when luminal [Ca2+] returns to resting level (that is, 250–600 µM).Citation24 The luminal EF-hand domain of STIM1 binds Ca2+ with an apparent dissociation constant (Kd) of ∼250 µM, whereas the Kd of STIM2 for Ca2+ is ∼500 µM.Citation24,25 Hence, STIM2 is more sensitive to small changes in luminal [Ca2+] and partially active at resting ER Ca2+ levels.Citation26 STIM2 shows a slower oligomerization kinetics and is less effective than STIM1 in ORAI1 binding and activation, which might be critical for limiting excessive SOCE.Citation27,28

ORAI1 and its homologues ORAI2 and ORAI3 were discovered by genome wide RNAi screens for SOCE inhibition and by positional cloning in patients with immune deficiency and CRAC channel dysfunction.Citation11,13,15 ORAI1 proteins have 4 transmembrane domains and cytosolic N- and C-termini. Co-expression with STIM1 showed that ORAI1 is the pore-forming subunit of the CRAC channel.Citation12,13,29 ORAI1 channels (therefore, also named CRAC modulator 1; CRACM1) are activated upon Ca2+ store depletion, translocation of activated STIM1 to ER-plasma membrane junctions and interaction of cytosolic STIM1 domains with the C-terminal domains of ORAI1 tetramers.Citation30,31 Heterologously expressed ORAI2 and ORAI3 channels (together with STIM1) conduct CRAC-like currents but exhibit distinct inactivation and permeability properties.Citation32,33 Store-depletion signaling via STIM2 also activates all 3 ORAI channels.Citation34 Over-expression of ORAI1 or ORAI2 (but not ORAI3) alone reduced endogenous SOCE, suggesting a fine stoichiometry between STIM and ORAI proteins.Citation14 Although TRP channels do not account for CRAC currents, a contribution of some TRP family members to STIM1-induced and store-dependent Ca2+ entry has been shown.Citation35 In this context, a SOCE-signaling complex consisting of TRPC1, ORAI1, and STIM1 has been proposed.Citation36

The principal components STIM1 and ORAI1 essentially or nearly exclusively contribute to SOCE in different cell types. The function of the innate and adaptive immune system requires STIM1/ORAI1-dependent SOCE.Citation37,38 In mouse T lymphocytes, STIM2 partially contributes to cytokine production.Citation39 ORAI2 and/or ORAI3 appear to have redundant functions in mouse T cells, whereas human T cells lacking functional ORAI1 failed to proliferate in vitro and to produce cytokines.Citation38 STIM1 and ORAI1 regulate mast cell activation,Citation40,41 and STIM1 is essential for Fcγ receptor-dependent Ca2+ signaling and phagocytosis in macrophages and neutrophils.Citation42,43 Roles of both STIM1 and ORAI1 in other cell types include growth and contractility of skeletal muscle,Citation44,45 proliferation of vascular smooth muscle cells,Citation46,47 and aggregation of platelets.Citation48,49

SOCE in the nervous system has been reviewed with respect to Ca2+ signaling and homeostasis in neurons and neuroglia.Citation50-53 The present review is mostly based on studies regarding expression analyses or using gene silencing or knockout of STIMs and ORAIs and gives a current overview on the putative functions of these SOCE components in both neurons and glial cells, i.e., astrocytes and microglia.

Expression of STIMs and ORAIs in Brain

STIM and ORAI isoforms are broadly expressed in murine and human tissues. At the RNA level, STIM1 was consistently detected in skeletal muscle and brain,Citation18,54,55 whereas STIM2 was preferentially found in brain and heart from both species.Citation18,19 Both STIM proteins were detected in murine and human brain.Citation19,55,56 Within the murine brain, STIM1 and STIM2 are distributed in the cerebral cortex and can be assigned to hippocampal and cerebellar structures. Whereas STIM1 is most prominent in the cerebellum, STIM2 dominates in hippocampus and cortex.Citation57 In human brain, STIM1 protein expression is high in cerebellum, medium in cerebral cortex, and low in hippocampus. STIM2 protein levels are high in hippocampus and cerebral cortex, and medium in cerebellum,Citation58 indicating that the differential distribution of STIM1 and STIM2 in cerebrum and cerebellum is similar in human and murine brain.

Expression of STIM mRNAs at the cellular level was analyzed using cell isolation or separation by laser capture, cell soma harvest and cell culture. In primary hippocampal and cortical neurons, STIM2 is the predominant STIM isoform.Citation19,55,59 In hippocampal cultures, STIM2 was detected in both neuronal soma and dendrites, whereas STIM1 protein is restricted to the soma.Citation60 Furthermore, the expression of STIM1 and STIM2 increases during development of hippocampal neurons in vitro.Citation61,62

In Purkinje neurons, the principal neurons of the cerebellar cortex, STIM1 levels were higher than those of STIM2.Citation63 The second most important neuron within the cerebellum is the cerebellar granule cell, which is located in the nuclear layer and whose parallel fibers project to Purkinje neurons in the upper molecular layer. In cultured cerebellar granule cells, also a dominant STIM1 expression was found.Citation64

All three ORAI isoforms are detectable in murine and human brain. Interestingly, ORAI1 mRNA levels in both species appeared to be lower than those of ORAI3.Citation54,65,66 ORAI2 is prominently expressed in murine but not in human brain tissue.Citation54,57,58,65 A high abundance of ORAI2 was found in hippocampal neurons, cerebellar tissue and Purkinje neurons.Citation19,63 The origin of the strong ORAI3 expression, particularly in human brain, is presently unclear and might come from glial cells. In mouse cerebellum, ORAI3 overlaps with ORAI2 and was also detected in Purkinje neurons.Citation57,63 The role of ORAI2 in brain as well as in other tissues is still unclear.Citation67

Neurons

Store-operated Ca2+ entry has been implicated in neuronal Ca2+ signaling even before the discovery of STIM and ORAI. Ca2+ imaging experiments revealed SOCE in cultured and freshly dissociated cortical and hippocampal neurons.Citation68-70 A role for SOCE in spontaneous synaptic activity and in synaptic plasticity of hippocampal neurons was suggested from studies using the common SOCE inhibitors 2-aminoethoxydiphenyl borate (2-APB), SKF96365 and La3+.Citation71,72

First molecular evidence for neuronal SOCE provided a study using STIM and ORAI knockout mice. Berna-Erro et al. have shown that STIM2-deficient mice were protected from cerebral damage after ischemic stroke.Citation19 The reduced infarct size and the improved neurological outcome of STIM2−/− animals were independent of functional changes within the haematopoietic system. In cultured cortical neurons, SOCE was significantly decreased in the absence of STIM2 but not of STIM1 or ORAI1.Citation19 Hypoxia and hypoglycemia induced an increase in [Ca2+]i which was markedly reduced in STIM2−/− neurons.Citation19 Long lasting elevation of intracellular Ca2+ is critical for ischemic neuronal cell death.Citation73 Hypoxia/hypoglycemia inhibits ATP-dependent Ca2+ transport into the ER and might, therefore, trigger persistent STIM2 activity and SOCE-induced accumulation of cytosolic Ca2+ (). Glutamate-induced excitotoxicity is a process triggered by and contributing to neuronal Ca2+ accumulation during ischemia.Citation74 Sodero et al. reported that glutamate-mediated excitotoxicity leads to a loss of cholesterol in brain.Citation75 Cholesterol, a major component of the plasma membrane, plays a key role in regulating neuronal functions. Glutamate-induced cholesterol loss required high intracellular [Ca2+] and functional STIM2 in hippocampal neurons.Citation75 Apart from the role of STIM2 in ischemic stroke, behavioral tests revealed an impairment of hippocampus-dependent spatial learning in STIM2-deficient mice.Citation19

Figure 1. Roles of STIM1 and STIM2 in neuronal Ca2+ homeostasis and synaptic function. (A) Proposed model for the involvement of STIM2 in ischemic Ca2+ accumulation in hippocampal and cortical neurons. A disturbed refilling of intracellular Ca2+ stores during ischemia may be induced by inhibition of sarco-endoplasmic reticulum Ca2+ pumps. The reduced [Ca2+] in the endoplasmic reticulum (ER), [Ca2+]ER, leads to the activation of STIM2 and, possibly, of ORAI2 channels in the plasma membrane. Opening of ORAI channels results in SOCE, which contributes to deleterious Ca2+ accumulation in the cytosol. (B) Role of STIM2 in maintenance of postsynaptic mushroom spines in hippocampal neurons. Activation of STIM2 due to reduced [Ca2+]ER induces continuous SOCE via ORAI (supposedly, ORAI2) channels. Increased cytosolic [Ca2+] supports constant levels of Ca2+/calmodulin-dependent protein kinase II (CAMKII) and long-term stability of mushroom spines. (C) Role of STIM1 in cerebellar Purkinje neurons. Activation of metabotropic glutamate receptor type 1 (mGluR1) induces Ca2+ release from ER, a decrease in [Ca2+]ER, and the activation of STIM1. SOCE is probably mediated by opening of ORAI2 channels through STIM1. SOCE results in Ca2+ store refilling and supports Ca2+-dependent activation of the transient receptor potential channel TRPC3. TRPC3 mediates slow excitatory postsynaptic currents (EPSC) which are important for Purkinje neuron function and cerebellar motor behavior.

Figure 1. Roles of STIM1 and STIM2 in neuronal Ca2+ homeostasis and synaptic function. (A) Proposed model for the involvement of STIM2 in ischemic Ca2+ accumulation in hippocampal and cortical neurons. A disturbed refilling of intracellular Ca2+ stores during ischemia may be induced by inhibition of sarco-endoplasmic reticulum Ca2+ pumps. The reduced [Ca2+] in the endoplasmic reticulum (ER), [Ca2+]ER, leads to the activation of STIM2 and, possibly, of ORAI2 channels in the plasma membrane. Opening of ORAI channels results in SOCE, which contributes to deleterious Ca2+ accumulation in the cytosol. (B) Role of STIM2 in maintenance of postsynaptic mushroom spines in hippocampal neurons. Activation of STIM2 due to reduced [Ca2+]ER induces continuous SOCE via ORAI (supposedly, ORAI2) channels. Increased cytosolic [Ca2+] supports constant levels of Ca2+/calmodulin-dependent protein kinase II (CAMKII) and long-term stability of mushroom spines. (C) Role of STIM1 in cerebellar Purkinje neurons. Activation of metabotropic glutamate receptor type 1 (mGluR1) induces Ca2+ release from ER, a decrease in [Ca2+]ER, and the activation of STIM1. SOCE is probably mediated by opening of ORAI2 channels through STIM1. SOCE results in Ca2+ store refilling and supports Ca2+-dependent activation of the transient receptor potential channel TRPC3. TRPC3 mediates slow excitatory postsynaptic currents (EPSC) which are important for Purkinje neuron function and cerebellar motor behavior.

Whereas no obvious abnormalities were reported for brain structures of STIM2−/− mice,Citation19 Sun et al. showed that conditional knockout of the STIM2 gene in the hippocampus of 2–6 month old mice induced a massive neuronal loss in this brain region.Citation60 Deletion of STIM2 in hippocampal neurons caused a moderate reduction of somatic SOCE but a dramatic decrease of SOCE in dendritic spines.Citation60 The absence of STIM2 also reduced the number of spines and changed their morphology by reducing the fraction of mushroom spines which play an important role in the storage of memories.Citation60 A loss of mushroom spines, concurrent with the decrease in synaptic SOCE and the downregulation of STIM2 was observed in hippocampal neurons from the presenilin-1 M146V knockin mouse model of Alzheimer's disease.Citation60 Overexpression of STIM2 (but not of STIM1) rescued synaptic SOCE and increased the expression of phosphorylated Ca2+/calmodulin-dependent protein kinase II (CaMKII) suggesting that STIM2-dependent SOCE is required for CaMKII activity and stabilization of mushroom spines in healthy neurons ().Citation60 It is not clear why STIM2 but not STIM1 regulates synaptic SOCE in hippocampal neurons. STIM2 has a higher Kd for Ca2+ than STIM1 and induces SOCE near resting ER Ca2+ levels.Citation26 Thus, STIM2 stabilizes basal [Ca2+]i even at incomplete store depletion and, in turn, maintains steady-state CaMKII activity and integrity of mushroom spines.

Another recent study supports the role of STIM2 in regulating dendritic spine density and morphology in hippocampal neurons. Garcia-Alvarez et al. showed that STIM2 preferentially localizes to large dendritic spines and is enriched in the postsynaptic density.Citation61 STIM2 is required for regular synaptic activity and mediates cAMP-dependent phosphorylation and trafficking of the AMPA receptor subunit GluA1 to plasma membrane-ER junctions.Citation61 However, SOCE does not appear to be involved in STIM2-dependent phosphorylation of GluA1. Analogous to STIM interaction with ORAI1, the authors suggest that STIM2 binds via its cytosolic domain to GluA1 and couples GluA1 to cAMP-dependent protein kinase (PKA).Citation61

Signaling of STIM proteins through other pathways than SOCE has also been shown for the interaction of STIM1 with the voltage-gated Ca2+ channel (VGCC) subtype Cav1.2.Citation76,77 STIM1 inhibits VGCC activation by binding to Cav1.2 via the cytosolic STIM-ORAI activating domain.Citation76,77 It has been suggested that STIM1-mediated suppression of VGCCs is involved in the differentiation of neurons from embryonic stem cells.Citation78,79 Knockdown of STIM1 or STIM2 reduced SOCE and inhibited entry of mouse embryonic stem cells into neural lineage.Citation78 ORAI1 contributed to SOCE but was not required for neuronal differentiation. STIM1 knockdown induced an increase in voltage-dependent Ca2+ entry and treatment of cells with the VGCC blocker nifedipine facilitated neural differentiation.Citation78 Another study corroborates the role for STIM1 in neurogenesis. Somasundaram et al. found a markedly reduced SOCE after knockdown/knockout of STIM1 or ORAI1 in embryonic and neonatal neural precursor cells.Citation80 Suppression of STIM1 or ORAI1 diminished proliferation of neural precursors and inhibited activation of the transcription factor NFAT (nuclear factor of activated T cells).Citation80

Whereas STIM2 regulates SOCE in hippocampal and cortical neurons, STIM1 appears to be the primary STIM isoform in cerebellar granule cells and Purkinje neurons.Citation79 Lalonde et al. have shown that changes in the extracellular K+ concentration rapidly induce redistribution of overexpressed STIM1 together with overexpressed ORAI1 and ORAI2 in cerebellar granule neurons.Citation64 Because a switch of extracellular [K+] from a high (25–65 mM) to a low level (5 mM; near resting extracellular [K+]) induces plasma membrane hyperpolarization (repolarization), the authors suggest that changes in the membrane potential induce SOCE in granule neurons. Furthermore, ryanodine receptor-dependent Ca2+ stores were depleted by K+-induced hyperpolarization and a sustained SOCE signal was observed at resting extracellular [K+].Citation64 Knockdown of STIM1 in a neuroblastoma cell line and pharmacological inhibition of SOCE in granule neurons inhibited hyperpolarization-induced degradation of the neuron-specific transcription factor Sp4.Citation64 Sp4 has been implicated in dendrite patterning in cerebellar and hippocampal neurons as well as in memory and synaptic plasticity.Citation64,79 The activation of SOCE at resting (hyperpolarized) membrane potential suggests a homeostatic function of STIMs in neurons, i.e. the regulation of Ca2+-dependent gene transcription by controlling resting intracellular Ca2+ levels.

Hartmann et al. reported that STIM1 controls glutamate receptor-dependent synaptic transmission in Purkinje neurons and motor learning in mice.Citation63 The metabotropic glutamate receptor type 1 (mGluR1), that is highly expressed in Purkinje neurons, plays an important role in cerebellar functions like motor coordination.Citation81 Signal transduction downstream of mGluR1 involves the IP3-dependent Ca2+ release from ER and the activation of a slow excitatory postsynaptic current which is mediated by the TRP channel TRPC3.Citation82 In the absence of STIM1, the IP3-dependent Ca2+ release from dendritic ER Ca2+ stores and the TRPC3-mediated currents were abolished.Citation63 Interestingly, STIM1 was only required for Ca2+ store refilling and slow synaptic currents when Purkinje neurons were held at resting membrane potential. Depolarization-induced Ca2+ entry via VGCCs, however, induced a STIM1-independent recovery of ER Ca2+ release and TRPC3 activity.Citation63 The latter observation leads to the conclusion that activation of TRPC3 channels depends on intracellular [Ca2+] but not on interaction with STIM1 or the mGluR1-induced activation of phospholipase C (). The essential role of STIM1 in resting Purkinje neurons suggests that neuronal SOCE replenishes intracellular and ER Ca2+ levels when VGCC activity is low.

Astrocytes

Astrocytes are glial cells which are derived, like neurons and oligodendrocytes, from neuroepithelial progenitors. They perform a variety of homeostatic functions within the nervous system and communicate with neighboring glial cells and neurons by generating (intercellular) Ca2+ signals and by releasing and binding of transmitter molecules.Citation83 Cultured astrocytes and tumor cells of astroglial origin, i.e., glioblastoma cells, exhibit SOCE in response to metabotropic receptor agonists like ATP, histamine, and glutamate.Citation84-86

STIM1 and STIM2 proteins are expressed in cultured astrocytes while STIM1 appeared to be the dominant isoform.Citation55 STIM1 and ORAI1 have been implicated in Ca2+ signaling of astrocytes and glioblastoma cells. Moreno et al. reported that knockdown of STIM1 and ORAI1 or of ORAI1 alone diminishes thrombin-induced Ca2+ signals and SOCE in cultured rat astrocytes.Citation87 Motiani et al. showed a reduction of SOCE and of CRAC currents by silencing of STIM1 or ORAI1 in human glioblastoma cells.Citation88 Knockdown of STIM1 and ORAI1 slightly affected proliferation but clearly inhibited invasive glioblastoma cell migration.Citation88

Ronco et al. investigated the relative mRNA expression of ORAI homologs in cultured rat astrocytes.Citation89 Surprisingly, the authors were unable to detect ORAI1 but found a dominant expression of ORAI3 which was 6-fold more abundant than ORAI2.Citation89 Furthermore, previous studies suggest a role of TRP channels in astrocytic SOCE and, therefore, challenge a sole contribution of ORAI channels. Golovina reported that antisense oligonucleotides targeted to the TRP channel gene TRPC1 inhibited SOCE in murine astrocytes.Citation90 However, in a later study the same group showed a profound inhibition of astrocytic SOCE by knockdown of ORAI1.Citation91 Malarkey et al. reported that an antibody against TRPC1 reduces SOCE and abolishes ATP-mediated Ca2+ entry in cultured rat astrocytes.Citation92 Astrocytes also express further TRP channel subtypes, including TRPC4 and TRPC5,Citation90,92,93 which can form heteromeric channels with TRPC1.Citation94 A concordant regulation of TRPC1, TRPC4, ORAI3, and of receptor-induced Ca2+ entry has been observed in astrocytes treated with different pro-inflammatory agents and amyloid-β protein.Citation89 Thrombin treatment of astrocytes induced the up-regulation of a further TRP homolog, TRPC3, and an increase in SOCE.Citation95 The augmentation of SOCE was suppressed after knockdown of TRPC3.Citation95 In contrast to CRAC channels, which are highly selective for Ca2+, TRPC3 is permeable for some other divalent cations, including Sr2+.Citation4,96 Grimaldi et al. have shown that store depletion induces only a negligible Sr2+ entry in astrocytes suggesting a minor role of TRPC3 in astrocytic SOCE.Citation97

Microglia

Microglia are derived from myeloid precursors and constitute up to 20% of the total glia population. Microglia sense disturbances or loss of brain homeostasis and undergo morphological and functional changes in response to brain injury and infection.Citation98 This “microglial activation” includes shape changes toward an ameboid appearance, directed movement, proliferation, phagocytosis, and release of cytokines. Microglia are equipped with a variety of surface receptors for neurotransmitters, signaling molecules and pathogens. Extracellular nucleotides, i.e. ATP, ADP and UDP, accumulate during brain injury, activate purinergic P2Y receptors and stimulate migration and phagocytosis in microglia.Citation99-101

Receptor-induced Ca2+ signals play a central role in microglial function and activation.Citation98 SOCE and CRAC currents have been described in microglia.Citation102-104 Ohana et al. showed that CRAC/ORAI channels rather than TRP channels mediate SOCE in cultured rat microglia.Citation105 SOCE was inhibited by a high concentration of 2-APB (50 µM) and by exchange of extracellular Ca2+ by Sr2+ or Ba2+. From the high expression of ORAI1 and ORAI3 in microglia and the agonistic action of 50 µM 2-APB on ORAI3,Citation106 the authors suggested a mayor role of ORAI1 in microglial SOCE.Citation105 The same group detected high levels of ORAI1 and STIM1 in microglial podosomes, actin-rich structures involved in cell motility and invasion.Citation107 Treatment of cultured rat microglia with SOCE inhibitors strongly reduced migration, i.e., transmigration across filters with defined pores, and invasion, i.e. transmigration through pores coated with extracellular matrix molecules.Citation107

Heo et al. confirmed the expression of all ORAI isoforms and of STIM1 in purified cultured mouse microglia and showed that downregulation of ORAI1 and STIM1 reduces SOCE and UDP-induced Ca2+ signals.Citation108 Silencing of STIM1 inhibited lipopolysaccharide-induced activation of NFAT1 and production of interleukin 6 (IL-6) but did not affect activation of NF-κB. STIM1 knockdown slightly decreased release of TNF-α and reduced the UDP-stimulated phagocytosis of bacterial particles.Citation108 The P2Y6 receptor agonist UDP was previously shown to induce activation of NFAT1 and NFAT2 as well as expression of the chemokines CCL2 and CCL3 in microglia.Citation109

We investigated the role of STIM1, STIM2, and ORAI1 in microglia.Citation110 To estimate the relative contribution of STIM1 and STIM2 to microglial responses and to evaluate the role of ORAI1, cultured microglia were obtained from corresponding knockout mice.Citation19,48,49 Purity of microglial cultures was tested by staining with isolectin B4 and by using transgenic CX3CR1GFP mice,Citation111 where GFP is exclusively expressed in microglia. In our primary cultures, which showed a purity of ∼99.5%, we analyzed the mRNA levels of STIM and ORAI isoforms by quantitative RT-PCR. STIM1, STIM2, and ORAI1 were more abundant than ORAI2 and ORAI3.Citation110 Calcium imaging revealed a graded suppression of SOCE in the absence of STIM1, STIM2, or ORAI1. SOCE evoked by blockage of sarco-endoplasmic reticulum ATPase (SERCA) was nearly absent in STIM1−/− microglia (91% inhibition), whereas STIM2−/− cells showed a less pronounced but significant inhibition (by 30%). CRAC currents were suppressed in the absence of STIM1. SOCE was substantially decreased in ORAI1−/− microglia (by 70%). Purinergic stimulation with the P2Y6 receptor agonist UDP or the P2Y12 receptor agonist 2-methylthio-ADP (2-MeSADP) caused SOCE amplitudes which were significantly smaller than those evoked by SERCA inhibition. The P2Y12 receptor-mediated SOCE was blocked by 40% in STIM2−/− microglia and reduced by 77% in STIM1−/− cells, suggesting a decreased effect of STIM1 and an increased impact of STIM2 on SOCE of lower magnitude.Citation110

A role for STIM2 in modulating the threshold for Ca2+ entry has been reported previously. Kar et al. showed that STIM2 exclusively contributed to Ca2+ signals evoked by tyrosine kinase receptors but not by G-protein coupled receptors in leukemia cells.Citation112 Thiel et al. found a STIM2-dependent SOCE signal after mild store depletion which was completely replaced by a STIM1-dependent SOCE upon strong store depletion.Citation113 Ong et al. proposed that STIM2 triggers SOCE by recruiting STIM1 at low stimulus intensities when ER Ca2+ stores are mildly depleted.Citation114 Together, these data suggest that STIM2 increases the sensitivity of intracellular Ca2+ signals to extracellular agonists.

To evaluate the role of SOCE in microglial migration and phagocytosis we used STIM and ORAI knockout mice, the SOCE blockers 2-APB, La3+, and the TRP channel blocker N-(p-amylcinnamoyl)anthranilic acid (ACA).Citation115 We found that ACA effectively blocks SOCE in microglia with an IC50 of 0.4 µM.Citation110 Transmigration of cultured microglia stimulated with ATP was largely reduced by ACA and SOCE inhibitors. Uptake of zymosan particles by UDP-stimulated microglia was nearly abolished in the presence of 2-APB. P2Y receptor-induced migration (induced by 2-MeSADP, ATP, and UDP) was inhibited in STIM1−/−, STIM2−/−, and ORAI1−/− microglia. P2Y6 receptor-dependent phagocytosis was nearly abolished in STIM1−/− cells and largely decreased in STIM2−/− and ORAI1−/− microglia.Citation110 Basal migration and phagocytosis in the absence of purinergic stimuli were not affected by STIM or ORAI knockout.

From these data we concluded that STIM1 and ORAI1 are the mayor constituents of microglial SOCE and that STIM2 is an important component of this signaling pathway. Consequently, SOCE is essential for P2Y receptor-dependent Ca2+ entry, migration, and phagocytosis in microglia (). Possible downstream effectors of microglial SOCE involve protein kinase C (PKC) and the Ca2+/calmodulin-activated myosin light chain kinase (MLCK) which both activate phagocytosis in microglia.Citation116,117 The Ca2+-dependent phosphorylation of protein kinase B (Akt) is required for ADP-induced chemotaxis of microglia.Citation118

Figure 2. STIM1, STIM2, and ORAI1 regulate microglia functions. The G protein-coupled receptors P2Y6 and P2Y12 are activated by UDP and ADP/ATP, respectively. Activation of P2Y receptors induces Ca2+ release from endoplasmic reticulum (ER) and reduces [Ca2+]ER. In turn, STIM1 and STIM2 proteins activate ORAI1 channels. The resulting SOCE promotes activation of different Ca2+-dependent signaling molecules, including Ca2+/calmodulin-activated myosin light chain kinase (MLCK), protein kinase C (PKC), the serin/threonine specific kinase Akt, and the transcription factor NFAT. Remodeling of actin-myosin skeleton is probably involved in MLCK/PKC-dependent phagocytosis and in Akt-dependent cell migration. NFAT mediates UDP-induced expression of chemokines in microglia.

Figure 2. STIM1, STIM2, and ORAI1 regulate microglia functions. The G protein-coupled receptors P2Y6 and P2Y12 are activated by UDP and ADP/ATP, respectively. Activation of P2Y receptors induces Ca2+ release from endoplasmic reticulum (ER) and reduces [Ca2+]ER. In turn, STIM1 and STIM2 proteins activate ORAI1 channels. The resulting SOCE promotes activation of different Ca2+-dependent signaling molecules, including Ca2+/calmodulin-activated myosin light chain kinase (MLCK), protein kinase C (PKC), the serin/threonine specific kinase Akt, and the transcription factor NFAT. Remodeling of actin-myosin skeleton is probably involved in MLCK/PKC-dependent phagocytosis and in Akt-dependent cell migration. NFAT mediates UDP-induced expression of chemokines in microglia.

Conclusions and Implications for Neurological Disorders

STIM and ORAI proteins have been implicated in normal brain function but also in neurological disorders. STIM1, preferentially expressed in cerebellar neurons, and STIM2, more abundant in hippocampal and cortical neurons, trigger neuronal SOCE. The versatile role of STIMs and, consequently, of SOCE in neuronal signaling was rather unanticipated because synaptic activity was thought to provide a sufficient supply for intracellular Ca2+. However, recent studies suggest that SOCE is a mayor source for intracellular Ca2+ in resting neurons. STIM1 is required for slow glutamate receptor signals in Purkinje neurons and for Ca2+-dependent gene transcription in cerebellar granule neurons.Citation63,64 STIM2 controls steady-state activity of CAMKII and thereby stabilizes mushroom spines in hippocampal neurons.Citation60 Decreased STIM2 activity and loss of mushroom spines may be responsible for memory loss in aging neurons and in Alzheimer's disease.Citation60 STIM2 is also a potential target for the treatment of glutamate-induced excitotoxicity during epilepsy, brain trauma and cerebral infarction.Citation19,75 The role of ORAI proteins in neuronal Ca2+ signaling is still unresolved. In particular, consequences of the dominant ORAI2 expression should be clarified. In comparison to ORAI1, ORAI2-mediated currents show a smaller amplitude but undergo a decreased inactivation at higher [Ca2+]i.Citation32 In analogy to functional differences between STIM1 and STIM2, ORAI2 might, therefore, support a persistent and moderate “steady-state SOCE” in neurons.

Less is known about the role of STIMs and ORAIs in astrocytes. Here, a further ORAI isoform, ORAI3, might play a central role. ORAI3 is, in contrast to ORAI1 and ORAI2, inhibited by reactive oxygen species (ROS),Citation119 and is required for store-independent CRAC channels activated by arachidonic acid,Citation120,121 a phospholipase A2 (PLA2) metabolite. Both, PLA2 activity and ROS production were implicated in neurodegenerative diseases, such as cerebral ischemia and Alzheimer's disease,Citation122 while astrocytes play diverse supportive roles in maintaining neuronal health.Citation123 In a mouse model of Alzheimer's disease, a deregulation of astrocytic Ca2+ homeostasis and signaling, including increased basal [Ca2+]i and spontaneous network activity, was observed.Citation124 Since SOCE is upregulated in astrocytes treated with amyloid-β,Citation89 future work on the role of STIM and ORAI proteins might help to understand the pathophysiological regulation of astrocytic Ca2+ signaling and function.

In microglia, STIM1, STIM2, and ORAI1 represent the mayor SOCE components. While STIM1 is the primary ER Ca2+ sensor, STIM2 contributes to SOCE, particularly upon weak Ca2+ store depletion. Several microglial functions, i.e., release of cytokines, chemotaxis, and stimulated phagocytosis, are impaired in the absence or after downregulation of these SOCE components. Dysfunction and dystrophy of microglia has been recognized as a potential cause of neurodegeneration and impaired neuronal development.Citation125-128 For example, engulfment of dysfunctional synapses during development and phagocytosis of cellular debris (such as amyloid-β protein) are possibly regulated by intracellular Ca2+ release and SOCE in microglia.Citation102,129,130 Therefore, STIM1, STIM2, and Orai1 represent possible targets for the treatment of microglial dysfunction that may underlie neurodegenerative and neurodevelopmental disorders.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Funding

The author was supported by the Deutsche Forschungsgemeinschaft (DFG; KR3408/2-1).

References

  • Streb H, Irvine RF, Berridge MJ, Schulz I. Release of Ca2+ from a nonmitochondrial intracellular store in pancreatic acinar cells by inositol-1,4,5-trisphosphate. Nature 1983; 306:67-9; PMID:6605482
  • Putney JW, Jr. Stimulus-permeability coupling: role of calcium in the receptor regulation of membrane permeability. Pharmacol Rev 1978; 30:209-45; PMID:224401
  • Putney JW, Jr. A model for receptor-regulated calcium entry. Cell Calcium 1986; 7:1-12; PMID:2420465
  • Parekh AB, Penner R. Store depletion and calcium influx. Physiol Rev 1997; 77:901-30; PMID:9354808
  • Zitt C, Halaszovich CR, Lückhoff A. The TRP family of cation channels: probing and advancing the concepts on receptor-activated calcium entry. Prog Neurobiol 2002; 66:243-64; PMID:11960680
  • Hoth M, Penner R. Depletion of intracellular calcium stores activates a calcium current in mast cells. Nature 1992; 355:353-6; PMID:1309940
  • Gees M, Colsoul B, Nilius B. The role of transient receptor potential cation channels in Ca2+ signaling. Cold Spring Harb Perspect Biol 2010; 2:a003962; PMID:20861159; http://dx.doi.org/10.1101/cshperspect.a003962
  • Liou J, Kim ML, Heo WD, Jones JT, Myers JW, Ferrell JE, Jr, Meyer T. STIM is a Ca2+ sensor essential for Ca2+-store-depletion-triggered Ca2+ influx. Curr Biol 2005; 15:1235-41; PMID:16005298
  • Roos J, DiGregorio PJ, Yeromin AV, Ohlsen K, Lioudyno M, Zhang S, Safrina O, Kozak JA, Wagner SL, Cahalan MD, et al. STIM1, an essential and conserved component of store-operated Ca2+ channel function. J Cell Biol 2005; 169:435-45; PMID:15866891
  • Zhang SL, Yu Y, Roos J, Kozak JA, Deerinck TJ, Ellisman MH, Stauderman KA, Cahalan MD. STIM1 is a Ca2+ sensor that activates CRAC channels and migrates from the Ca2+ store to the plasma membrane. Nature 2005; 437:902-5; PMID:16208375
  • Feske S, Gwack Y, Prakriya M, Srikanth S, Puppel SH, Tanasa B, Hogan PG, Lewis RS, Daly M, Rao A. A mutation in Orai1 causes immune deficiency by abrogating CRAC channel function. Nature 2006; 441:179-85; PMID:16582901
  • Prakriya M, Feske S, Gwack Y, Srikanth S, Rao A, Hogan PG. Orai1 is an essential pore subunit of the CRAC channel. Nature 2006; 443:230-3; PMID:16921383
  • Yeromin AV, Zhang SL, Jiang W, Yu Y, Safrina O, Cahalan MD. Molecular identification of the CRAC channel by altered ion selectivity in a mutant of Orai. Nature 2006; 443:226-9; PMID:16921385
  • Mercer JC, Dehaven WI, Smyth JT, Wedel B, Boyles RR, Bird GS, Putney JW, Jr. Large store-operated calcium selective currents due to co-expression of Orai1 or Orai2 with the intracellular calcium sensor, Stim1. J Biol Chem 2006; 281:24979-90; PMID:16807233
  • Vig M, Peinelt C, Beck A, Koomoa DL, Rabah D, Koblan-Huberson M, Kraft S, Turner H, Fleig A, Penner R, Kinet JP. CRACM1 is a plasma membrane protein essential for store-operated Ca2+ entry. Science 2006; 312:1220-3; PMID:16645049
  • Parker NJ, Begley CG, Smith PJ, Fox RM. Molecular cloning of a novel human gene (D11S4896E) at chromosomal region 11p15.5. Genomics 1996; 37:253-6; PMID:8921403
  • Sabbioni S, Barbanti-Brodano G, Croce CM, Negrini M. GOK: a gene at 11p15 involved in rhabdomyosarcoma and rhabdoid tumor development. Cancer Res 1997; 57:4493-7; PMID:9377559
  • Williams RT, Manji SS, Parker NJ, Hancock MS, Van Stekelenburg L, Eid JP, Senior PV, Kazenwadel JS, Shandala T, Saint R, et al. Identification and characterization of the STIM (stromal interaction molecule) gene family: coding for a novel class of transmembrane proteins. Biochem J 2001; 357:673-85; PMID:11463338
  • Berna-Erro A, Braun A, Kraft R, Kleinschnitz C, Schuhmann MK, Stegner D, Wultsch T, Eilers J, Meuth SG, Stoll G, Nieswandt B. STIM2 regulates capacitive Ca2+ entry in neurons and plays a key role in hypoxic neuronal cell death. Sci Signal 2009; 2:ra67; PMID:19843959; http://dx.doi.org/10.1126/scisignal.2000522
  • Soboloff J, Rothberg BS, Madesh M, Gill DL. STIM proteins: dynamic calcium signal transducers. Nat Rev Mol Cell Biol 2012; 13:549-65; PMID:22914293; http://dx.doi.org/10.1038/nrm3414
  • Stathopulos PB, Ikura M. Structural aspects of calcium-release activated calcium channel function. Channels (Austin) 2013; 7:344-53; PMID:24213636; http://dx.doi.org/10.4161/chan.26734
  • Spassova MA, Soboloff J, He LP, Xu W, Dziadek MA, Gill DL. STIM1 has a plasma membrane role in the activation of store-operated Ca(2+) channels. Proc Natl Acad Sci U S A 2006; 103:4040-5; PMID:16537481
  • Soboloff J, Spassova MA, Hewavitharana T, He LP, Xu W, Johnstone LS, Dziadek MA, Gill DL. STIM2 is an inhibitor of STIM1-mediated store-operated Ca2+ entry. Curr Biol 2006; 16:1465-70; PMID:16860747
  • Stathopulos PB, Zheng L, Li GY, Plevin MJ, Ikura M. Structural and mechanistic insights into STIM1-mediated initiation of store-operated calcium entry. Cell 2008; 135:110-22; PMID:18854159; http://dx.doi.org/10.1016/j.cell.2008.08.006
  • Zheng L, Stathopulos PB, Li GY, Ikura M. Biophysical characterization of the EF-hand and SAM domain containing Ca2+ sensory region of STIM1 and STIM2. Biochem Biophys Res Commun 2008; 369:240-6; PMID:18166150; http://dx.doi.org/10.1016/j.bbrc.2007.12.129
  • Brandman O, Liou J, Park WS, Meyer T. STIM2 is a feedback regulator that stabilizes basal cytosolic and endoplasmic reticulum Ca2+ levels. Cell 2007; 131:1327-39; PMID:18160041
  • Stathopulos PB, Zheng L, Ikura M. Stromal interaction molecule (STIM) 1 and STIM2 calcium sensing regions exhibit distinct unfolding and oligomerization kinetics. J Biol Chem 2009; 284:728-32; PMID:19019825; http://dx.doi.org/10.1074/jbc.C800178200
  • Wang X, Wang Y, Zhou Y, Hendron E, Mancarella S, Andrake MD, Rothberg BS, Soboloff J, Gill DL. Distinct Orai-coupling domains in STIM1 and STIM2 define the Orai-activating site. Nat Commun 2014; 5:3183; PMID:24492416; http://dx.doi.org/10.1038/ncomms4183
  • Vig M, Beck A, Billingsley JM, Lis A, Parvez S, Peinelt C, Koomoa DL, Soboloff J, Gill DL, Fleig A, et al. CRACM1 multimers form the ion-selective pore of the CRAC channel. Curr Biol 2006; 16:2073-9; PMID:16978865
  • Schindl R, Muik M, Fahrner M, Derler I, Fritsch R, Bergsmann J, Romanin C. Recent progress on STIM1 domains controlling Orai activation. Cell Calcium 2009; 46:227-32; PMID:19733393; http://dx.doi.org/10.1016/j.ceca.2009.08.003
  • Shim AH, Tirado-Lee L, Prakriya M. Structural and Functional Mechanisms of CRAC Channel Regulation. J Mol Biol 2015; 427:77-93; PMID:25284754; http://dx.doi.org/10.1016/j.jmb.2014.09.021
  • Lis A, Peinelt C, Beck A, Parvez S, Monteilh-Zoller M, Fleig A, Penner R. CRACM1, CRACM2, and CRACM3 are store-operated Ca2+ channels with distinct functional properties. Curr Biol 2007; 17:794-800; PMID:17442569
  • DeHaven WI, Smyth JT, Boyles RR, Putney JW, Jr. Calcium inhibition and calcium potentiation of Orai1, Orai2, and Orai3 calcium release-activated calcium channels. J Biol Chem 2007; 282:17548-56; PMID:17452328
  • Parvez S, Beck A, Peinelt C, Soboloff J, Lis A, Monteilh-Zoller M, Gill DL, Fleig A, Penner R. STIM2 protein mediates distinct store-dependent and store-independent modes of CRAC channel activation. FASEB J 2008; 22:752-61; PMID:17905723
  • Yuan JP, Zeng W, Huang GN, Worley PF, Muallem S. STIM1 heteromultimerizes TRPC channels to determine their function as store-operated channels. Nat Cell Biol 2007; 9:636-45; PMID:17486119
  • Ong HL, Ambudkar IS. The dynamic complexity of the TRPC1 channelosome. Channels (Austin) 2011; 5:424-31; PMID:21747233; http://dx.doi.org/10.4161/chan.5.5.16471
  • Feske S. ORAI1 and STIM1 deficiency in human and mice: roles of store-operated Ca2+ entry in the immune system and beyond. Immunol Rev 2009; 231:189-209; PMID:19754898; http://dx.doi.org/10.1111/j.1600-065X.2009.00818.x
  • Bergmeier W, Weidinger C, Zee I, Feske S. Emerging roles of store-operated Ca2+ entry through STIM and ORAI proteins in immunity, hemostasis and cancer. Channels (Austin) 2013; 7:379-91; PMID:23511024; http://dx.doi.org/10.4161/chan.24302
  • Oh-Hora M, Yamashita M, Hogan PG, Sharma S, Lamperti E, Chung W, Prakriya M, Feske S, Rao A. Dual functions for the endoplasmic reticulum calcium sensors STIM1 and STIM2 in T cell activation and tolerance. Nat Immunol 2008; 9:432-43; PMID:18327260; http://dx.doi.org/10.1038/ni1574
  • Baba Y, Nishida K, Fujii Y, Hirano T, Hikida M, Kurosaki T. Essential function for the calcium sensor STIM1 in mast cell activation and anaphylactic responses. Nat Immunol 2008; 9:81-8; PMID:18059272
  • Vig M, DeHaven WI, Bird GS, Billingsley JM, Wang H, Rao PE, Hutchings AB, Jouvin MH, Putney JW, Kinet JP. Defective mast cell effector functions in mice lacking the CRACM1 pore subunit of store-operated calcium release-activated calcium channels. Nat Immunol 2008; 9:89-96; PMID:18059270
  • Braun A, Gessner JE, Varga-Szabo D, Syed SN, Konrad S, Stegner D, Vögtle T, Schmidt RE, Nieswandt B. STIM1 is essential for Fcgamma receptor activation and autoimmune inflammation. Blood 2009; 113:1097-104; PMID:18941110; http://dx.doi.org/10.1182/blood-2008-05-158477
  • Zhang H, Clemens RA, Liu F, Hu Y, Baba Y, Theodore P, Kurosaki T, Lowell CA. STIM1 calcium sensor is required for activation of the phagocyte oxidase during inflammation and host defense. Blood 2014; 123:2238-49; PMID:24493668; http://dx.doi.org/10.1182/blood-2012-08-450403
  • Stiber J, Hawkins A, Zhang ZS, Wang S, Burch J, Graham V, Ward CC, Seth M, Finch E, Malouf N, et al. STIM1 signalling controls store-operated calcium entry required for development and contractile function in skeletal muscle. Nat Cell Biol 2008; 10:688-97; PMID:18488020; http://dx.doi.org/10.1038/ncb1731
  • Wei-Lapierre L, Carrell EM, Boncompagni S, Protasi F, Dirksen RT. Orai1-dependent calcium entry promotes skeletal muscle growth and limits fatigue. Nat Commun 2013; 4:2805; PMID:24241282; http://dx.doi.org/10.1038/ncomms3805
  • Zhang W, Halligan KE, Zhang X, Bisaillon JM, Gonzalez-Cobos JC, Motiani RK, Hu G, Vincent PA, Zhou J, Barroso M, et al. Orai1-mediated I (CRAC) is essential for neointima formation after vascular injury. Circ Res 2011; 109:534-42; PMID:21737791; http://dx.doi.org/10.1161/CIRCRESAHA.111.246777
  • Mancarella S, Potireddy S, Wang Y, Gao H, Gandhirajan RK, Autieri M, Scalia R, Cheng Z, Wang H, Madesh M, et al. Targeted STIM deletion impairs calcium homeostasis, NFAT activation, and growth of smooth muscle. FASEB J 2013; 27:893-906; PMID:23159931; http://dx.doi.org/10.1096/fj.12-215293
  • Varga-Szabo D, Braun A, Kleinschnitz C, Bender M, Pleines I, Pham M, Renné T, Stoll G, Nieswandt B. The calcium sensor STIM1 is an essential mediator of arterial thrombosis and ischemic brain infarction. J Exp Med 2008; 205:1583-91; PMID:18559454; http://dx.doi.org/10.1084/jem.20080302
  • Braun A, Varga-Szabo D, Kleinschnitz C, Pleines I, Bender M, Austinat M, Bösl M, Stoll G, Nieswandt B. Orai1 (CRACM1) is the platelet SOC channel and essential for pathological thrombus formation. Blood 2009; 113:2056-63; PMID:18832659; http://dx.doi.org/10.1182/blood-2008-07-171611
  • Putney JW, Jr. Capacitative calcium entry in the nervous system. Cell Calcium 2003; 34:339-44; PMID:12909080
  • Verkhratsky A, Parpura V. Store-operated calcium entry in neuroglia. Neurosci Bull 2014; 30:125-33; PMID:23677809; http://dx.doi.org/10.1007/s12264-013-1343-x
  • Majewski L, Kuznicki J. SOCE in neurons: Signaling or just refilling? Biochim Biophys Acta 2015; 1853:1940-52; PMID:25646572; http://dx.doi.org/10.1016/j.bbamcr.2015.01.019
  • Moccia F, Zuccolo E, Soda T, Tanzi F, Guerra G, Mapelli L, Lodola F, D'Angelo E. Stim and Orai proteins in neuronal Ca(2+) signaling and excitability. Front Cell Neurosci 2015; 9:153; PMID:25964739; http://dx.doi.org/10.3389/fncel.2015.00153
  • Gross SA, Wissenbach U, Philipp SE, Freichel M, Cavalié A, Flockerzi V. Murine ORAI2 splice variants form functional Ca2+ release-activated Ca2+ (CRAC) channels. J Biol Chem 2007; 282:19375-84; PMID:17463004
  • Steinbeck JA, Henke N, Opatz J, Gruszczynska-Biegala J, Schneider L, Theiss S, Hamacher N, Steinfarz B, Golz S, Brüstle O, et al. Store-operated calcium entry modulates neuronal network activity in a model of chronic epilepsy. Exp Neurol 2011; 232:185-94; PMID:21906591; http://dx.doi.org/10.1016/j.expneurol.2011.08.022
  • Skibinska-Kijek A, Wisniewska MB, Gruszczynska-Biegala J, Methner A, Kuznicki J. Immunolocalization of STIM1 in the mouse brain. Acta Neurobiol Exp (Wars) 2009; 69:413-28; PMID:20048759
  • Lein ES, Hawrylycz MJ, Ao N, Ayres M, Bensinger A, Bernard A, Boe AF, Boguski MS, Brockway KS, Byrnes EJ, et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature 2007; 445:168-76; PMID:17151600
  • Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, Sivertsson Å, Kampf C, Sjöstedt E, Asplund A, et al. Proteomics. Tissue-based map of the human proteome. Science 2015; 347:1260419; PMID:25613900; http://dx.doi.org/10.1126/science.1260419
  • Gruszczynska-Biegala J, Pomorski P, Wisniewska MB, Kuznicki J. Differential roles for STIM1 and STIM2 in store-operated calcium entry in rat neurons. PLoS One 2011; 6:e19285; PMID:21541286; http://dx.doi.org/10.1371/journal.pone.0019285
  • Sun S, Zhang H, Liu J, Popugaeva E, Xu NJ, Feske S, White CL, 3rd, Bezprozvanny I. Reduced synaptic STIM2 expression and impaired store-operated calcium entry cause destabilization of mature spines in mutant presenilin mice. Neuron 2014; 82:79-93; PMID:24698269; http://dx.doi.org/10.1016/j.neuron.2014.02.019
  • Keil JM, Shen Z, Briggs SP, Patrick GN. Regulation of STIM1 and SOCE by the ubiquitin-proteasome system (UPS). PLoS One 2010; 5:e13465; PMID:20976103; http://dx.doi.org/10.1371/journal.pone.0013465
  • Garcia-Alvarez G, Lu B, Yap KA, Wong LC, Thevathasan JV, Lim L, Ji F, Tan KW, Mancuso JJ, Tang W, et al. STIM2 regulates PKA-dependent phosphorylation and trafficking of AMPARs. Mol Biol Cell 2015; 26:1141-59; PMID:25609091; http://dx.doi.org/10.1091/mbc.E14-07-1222
  • Hartmann J, Karl RM, Alexander RP, Adelsberger H, Brill MS, Rühlmann C, Ansel A, Sakimura K, Baba Y, Kurosaki T, et al. STIM1 controls neuronal Ca2+ signaling, mGluR1-dependent synaptic transmission, and cerebellar motor behavior. Neuron 2014; 82:635-44; PMID:24811382; http://dx.doi.org/10.1016/j.neuron.2014.03.027
  • Lalonde J, Saia G, Gill G. Store-operated calcium entry promotes the degradation of the transcription factor Sp4 in resting neurons. Sci Signal 2014; 7:ra51; PMID:24894994; http://dx.doi.org/10.1126/scisignal.2005242
  • Gwack Y, Srikanth S, Feske S, Cruz-Guilloty F, Oh-hora M, Neems DS, Hogan PG, Rao A. Biochemical and functional characterization of Orai proteins. J Biol Chem 2007; 282:16232-43; PMID:17293345
  • Takahashi Y, Murakami M, Watanabe H, Hasegawa H, Ohba T, Munehisa Y, Nobori K, Ono K, Iijima T, Ito H. Essential role of the N-terminus of murine Orai1 in store-operated Ca2+ entry. Biochem Biophys Res Commun 2007; 356:45-52; PMID:17343823
  • Hoth M, Niemeyer BA. The neglected CRAC proteins: Orai2, Orai3, and STIM2. Curr Top Membr 2013; 71:237-71; PMID:23890118; http://dx.doi.org/10.1016/B978-0-12-407870-3.00010-X
  • Bouron A. Activation of a capacitative Ca(2+) entry pathway by store depletion in cultured hippocampal neurones. FEBS Lett 2000; 470:269-72; PMID:10745080
  • Bouron A, Mbebi C, Loeffler JP, De Waard M. The beta-amyloid precursor protein controls a store-operated Ca2+ entry in cortical neurons. Eur J Neurosci 2004; 20:2071-8; PMID:15450086
  • Bouron A, Altafaj X, Boisseau S, De Waard M. A store-operated Ca2+ influx activated in response to the depletion of thapsigargin-sensitive Ca2+ stores is developmentally regulated in embryonic cortical neurons from mice. Brain Res Dev Brain Res 2005; 159:64-71; PMID:16099516
  • Emptage NJ, Reid CA, Fine A. Calcium stores in hippocampal synaptic boutons mediate short-term plasticity, store-operated Ca2+ entry, and spontaneous transmitter release. Neuron 2001; 29:197-208; PMID:11182091
  • Baba A, Yasui T, Fujisawa S, Yamada RX, Yamada MK, Nishiyama N, Matsuki N, Ikegaya Y. Activity-evoked capacitative Ca2+ entry: implications in synaptic plasticity. J Neurosci 2003; 23:7737-41; PMID:12944501
  • Lipton P. Ischemic cell death in brain neurons. Physiol Rev 1999; 79:1431-568; PMID:10508238
  • Lau A, Tymianski M. Glutamate receptors, neurotoxicity and neurodegeneration. Pflugers Arch 2010; 460:525-42; PMID:20229265; http://dx.doi.org/10.1007/s00424-010-0809-1
  • Sodero AO, Vriens J, Ghosh D, Stegner D, Brachet A, Pallotto M, Sassoè-Pognetto M, Brouwers JF, Helms JB, Nieswandt B, et al. Cholesterol loss during glutamate-mediated excitotoxicity. EMBO J 2012; 31:1764-73; PMID:22343944; http://dx.doi.org/10.1038/emboj.2012.31
  • Park CY, Shcheglovitov A, Dolmetsch R. The CRAC channel activator STIM1 binds and inhibits L-type voltage-gated calcium channels. Science 2010; 330:101-5; PMID:20929812; http://dx.doi.org/10.1126/science.1191027
  • Wang Y, Deng X, Mancarella S, Hendron E, Eguchi S, Soboloff J, Tang XD, Gill DL. The calcium store sensor, STIM1, reciprocally controls Orai and CaV1.2 channels. Science 2010; 330:105-9; PMID:20929813; http://dx.doi.org/10.1126/science.1191086
  • Hao B, Lu Y, Wang Q, Guo W, Cheung KH, Yue J. Role of STIM1 in survival and neural differentiation of mouse embryonic stem cells independent of Orai1-mediated Ca2+ entry. Stem Cell Res 2014; 12:452-66; PMID:24424349; http://dx.doi.org/10.1016/j.scr.2013.12.005
  • Hooper R, Rothberg BS, Soboloff J. Neuronal STIMulation at rest. Sci Signal 2014; 7:pe18; PMID:25056876; http://dx.doi.org/10.1126/scisignal.2005556
  • Somasundaram A, Shum AK, McBride HJ, Kessler JA, Feske S, Miller RJ, Prakriya M. Store-operated CRAC channels regulate gene expression and proliferation in neural progenitor cells. J Neurosci 2014; 34:9107-23; PMID:24990931; http://dx.doi.org/10.1523/JNEUROSCI.0263-14.2014
  • Ichise T, Kano M, Hashimoto K, Yanagihara D, Nakao K, Shigemoto R, Katsuki M, Aiba A. mGluR1 in cerebellar Purkinje cells essential for long-term depression, synapse elimination, and motor coordination. Science 2000; 288:1832-5; PMID:10846166
  • Hartmann J, Dragicevic E, Adelsberger H, Henning HA, Sumser M, Abramowitz J, Blum R, Dietrich A, Freichel M, Flockerzi V, et al. TRPC3 channels are required for synaptic transmission and motor coordination. Neuron 2008; 59:392-8; PMID:18701065; http://dx.doi.org/10.1016/j.neuron.2008.06.009
  • Verkhratsky A, Kettenmann H. Calcium signalling in glial cells. Trends Neurosci 1996; 19:346-52; PMID:8843604
  • Hartmann J, Verkhratsky A. Relations between intracellular Ca2+ stores and store-operated Ca2+ entry in primary cultured human glioblastoma cells. J Physiol 1998; 513:411-24; PMID:9806992
  • Golovina VA, Blaustein MP. Unloading and refilling of two classes of spatially resolved endoplasmic reticulum Ca(2+) stores in astrocytes. Glia 2000; 31:15-28; PMID:10816603
  • Jung S, Pfeiffer F, Deitmer JW. Histamine-induced calcium entry in rat cerebellar astrocytes: evidence for capacitative and non-capacitative mechanisms. J Physiol 2000; 527:549-61; PMID:10990540
  • Moreno C, Sampieri A, Vivas O, Peña-Segura C, Vaca L. STIM1 and Orai1 mediate thrombin-induced Ca(2+) influx in rat cortical astrocytes. Cell Calcium 2012; 52:457-67; PMID:22944608; http://dx.doi.org/10.1016/j.ceca.2012.08.004
  • Motiani RK, Hyzinski-García MC, Zhang X, Henkel MM, Abdullaev IF, Kuo YH, Matrougui K, Mongin AA, Trebak M. STIM1 and Orai1 mediate CRAC channel activity and are essential for human glioblastoma invasion. Pflugers Arch 2013; 465:1249-60; PMID:23515871; http://dx.doi.org/10.1007/s00424-013-1254-8
  • Ronco V, Grolla AA, Glasnov TN, Canonico PL, Verkhratsky A, Genazzani AA, Lim D. Differential deregulation of astrocytic calcium signalling by amyloid-β, TNFα, IL-1β and LPS. Cell Calcium 2014; 55:219-29; PMID:24656753; http://dx.doi.org/10.1016/j.ceca.2014.02.016
  • Golovina VA. Visualization of localized store-operated calcium entry in mouse astrocytes. Close proximity to the endoplasmic reticulum. J Physiol 2005; 564:737-49; PMID:15731184
  • Linde CI, Baryshnikov SG, Mazzocco-Spezzia A, Golovina VA. Dysregulation of Ca2+ signaling in astrocytes from mice lacking amyloid precursor protein. Am J Physiol Cell Physiol 2011; 300:C1502-12; PMID:21368296; http://dx.doi.org/10.1152/ajpcell.00379.2010
  • Malarkey EB, Ni Y, Parpura V. Ca2+ entry through TRPC1 channels contributes to intracellular Ca2+ dynamics and consequent glutamate release from rat astrocytes. Glia 2008; 56:821-35; PMID:18338793; http://dx.doi.org/10.1002/glia.20656
  • Song X, Zhao Y, Narcisse L, Duffy H, Kress Y, Lee S, Brosnan CF. Canonical transient receptor potential channel 4 (TRPC4) co-localizes with the scaffolding protein ZO-1 in human fetal astrocytes in culture. Glia 2005; 49:418-29; PMID:15540229
  • Strübing C, Krapivinsky G, Krapivinsky L, Clapham DE. Formation of novel TRPC channels by complex subunit interactions in embryonic brain. J Biol Chem 2003; 278:39014-9; PMID:12857742
  • Shirakawa H, Sakimoto S, Nakao K, Sugishita A, Konno M, Iida S, Kusano A, Hashimoto E, Nakagawa T, Kaneko S. Transient receptor potential canonical 3 (TRPC3) mediates thrombin-induced astrocyte activation and upregulates its own expression in cortical astrocytes. J Neurosci 2010; 30:13116-29; PMID:20881130; http://dx.doi.org/10.1523/JNEUROSCI.1890-10.2010
  • Ma HT, Patterson RL, van Rossum DB, Birnbaumer L, Mikoshiba K, Gill DL. Requirement of the inositol trisphosphate receptor for activation of store-operated Ca2+ channels. Science 2000; 287:1647-51; PMID:10698739
  • Grimaldi M, Maratos M, Verma A. Transient receptor potential channel activation causes a novel form of [Ca2+]i oscillations and is not involved in capacitative Ca2+ entry in glial cells. J Neurosci 2003; 23:4737-45; PMID:12805313
  • Kettenmann H, Hanisch UK, Noda M, Verkhratsky A. Physiology of microglia. Physiol Rev 2011; 91:461-553; PMID:21527731; http://dx.doi.org/10.1152/physrev.00011.2010
  • Honda S, Sasaki Y, Ohsawa K, Imai Y, Nakamura Y, Inoue K, Kohsaka S. Extracellular ATP or ADP induce chemotaxis of cultured microglia through Gi/o-coupled P2Y receptors. J Neurosci 2001; 21:1975-82; PMID:11245682
  • Haynes SE, Hollopeter G, Yang G, Kurpius D, Dailey ME, Gan WB, Julius D. The P2Y12 receptor regulates microglial activation by extracellular nucleotides. Nat Neurosci 2006; 9:1512-9; PMID:17115040
  • Koizumi S, Shigemoto-Mogami Y, Nasu-Tada K, Shinozaki Y, Ohsawa K, Tsuda M, Joshi BV, Jacobson KA, Kohsaka S, Inoue K. UDP acting at P2Y6 receptors is a mediator of microglial phagocytosis. Nature 2007; 446:1091-5; PMID:17410128
  • Möller T, Nolte C, Burger R, Verkhratsky A, Kettenmann H. Mechanisms of C5a and C3a complement fragment-induced [Ca2+]i signaling in mouse microglia. J Neurosci 1997; 17:615-24; PMID:8987784
  • Toescu EC, Möller T, Kettenmann H, Verkhratsky A. Long-term activation of capacitative Ca2+ entry in mouse microglial cells. Neuroscience 1998; 86:925-35; PMID:9692728
  • Beck A, Penner R, Fleig A. Lipopolysaccharide-induced down-regulation of Ca2+ release-activated Ca2+ currents (I CRAC) but not Ca2+-activated TRPM4-like currents (I CAN) in cultured mouse microglial cells. J Physiol 2008; 586:427-39; PMID:17991695
  • Ohana L, Newell EW, Stanley EF, Schlichter LC. The Ca2+ release-activated Ca2+ current (I(CRAC)) mediates store-operated Ca2+ entry in rat microglia. Channels (Austin) 2009; 3:129-39; PMID:19411837
  • Peinelt C, Lis A, Beck A, Fleig A, Penner R. 2-Aminoethoxydiphenyl borate directly facilitates and indirectly inhibits STIM1-dependent gating of CRAC channels. J Physiol 2008; 586:3061-73; PMID:18403424; http://dx.doi.org/10.1113/jphysiol.2008.151365
  • Siddiqui TA, Lively S, Vincent C, Schlichter LC. Regulation of podosome formation, microglial migration and invasion by Ca(2+)-signaling molecules expressed in podosomes. J Neuroinflammation 2012; 9:250; PMID:23158496; http://dx.doi.org/10.1186/1742-2094-9-250
  • Heo DK, Lim HM, Nam JH, Lee MG, Kim JY. Regulation of phagocytosis and cytokine secretion by store-operated calcium entry in primary isolated murine microglia. Cell Signal 2015; 27:177-86; PMID:25451082; http://dx.doi.org/10.1016/j.cellsig.2014.11.003
  • Kim B, Jeong HK, Kim JH, Lee SY, Jou I, Joe EH. Uridine 5′-diphosphate induces chemokine expression in microglia and astrocytes through activation of the P2Y6 receptor. J Immunol 2011; 186:3701-9; PMID:21317391; http://dx.doi.org/10.4049/jimmunol.1000212
  • Michaelis M, Nieswandt B, Stegner D, Eilers J, Kraft R. STIM1, STIM2, and Orai1 regulate store-operated calcium entry and purinergic activation of microglia. Glia 2015; 63:652-63; PMID:25471906; http://dx.doi.org/10.1002/glia.22775
  • Jung S, Aliberti J, Graemmel P, Sunshine MJ, Kreutzberg GW, Sher A, Littman DR. Analysis of fractalkine receptor CX(3)CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol Cell Biol 2000; 20:4106-14; PMID:10805752
  • Kar P, Bakowski D, Di Capite J, Nelson C, Parekh AB. Different agonists recruit different stromal interaction molecule proteins to support cytoplasmic Ca2+ oscillations and gene expression. Proc Natl Acad Sci U S A 2012; 109:6969-74; PMID:22509043; http://dx.doi.org/10.1073/pnas.1201204109
  • Thiel M, Lis A, Penner R. STIM2 drives Ca2+ oscillations through store-operated Ca2+ entry caused by mild store depletion. J Physiol 2013; 591:1433-45; PMID:23359669; http://dx.doi.org/10.1113/jphysiol.2012.245399
  • Ong HL, de Souza LB, Zheng C, Cheng KT, Liu X, Goldsmith CM, Feske S, Ambudkar IS. STIM2 enhances receptor-stimulated Ca2+ signaling by promoting recruitment of STIM1 to the endoplasmic reticulum-plasma membrane junctions. Sci Signal 2015; 8:ra3; PMID:25587190; http://dx.doi.org/10.1126/scisignal.2005748
  • Kraft R, Grimm C, Frenzel H, Harteneck C. Inhibition of TRPM2 cation channels by N-(p-amylcinnamoyl)anthranilic acid. Br J Pharmacol 2006; 148:264-73; PMID:16604090
  • Gitik M, Reichert F, Rotshenker S. Cytoskeleton plays a dual role of activation and inhibition in myelin and zymosan phagocytosis by microglia. FASEB J 2010; 24:2211-21; PMID:20179145; http://dx.doi.org/10.1096/fj.09-146118
  • Uesugi A, Kataoka A, Tozaki-Saitoh H, Koga Y, Tsuda M, Robaye B, Boeynaems JM, Inoue K. Involvement of protein kinase D in uridine diphosphate-induced microglial macropinocytosis and phagocytosis. Glia 2012; 60:1094-105; PMID:22488958; http://dx.doi.org/10.1002/glia.22337
  • Irino Y, Nakamura Y, Inoue K, Kohsaka S, Ohsawa K. Akt activation is involved in P2Y12 receptor-mediated chemotaxis of microglia. J Neurosci Res 2008; 86:1511-9; PMID:18183622; http://dx.doi.org/10.1002/jnr.21610
  • Bogeski I, Kummerow C, Al-Ansary D, Schwarz EC, Koehler R, Kozai D, Takahashi N, Peinelt C, Griesemer D, Bozem M, et al. Differential redox regulation of ORAI ion channels: a mechanism to tune cellular calcium signaling. Sci Signal 2010; 3:ra24; PMID:20354224; http://dx.doi.org/10.1126/scisignal.2000672
  • Mignen O, Thompson JL, Shuttleworth TJ. Both Orai1 and Orai3 are essential components of the arachidonate-regulated Ca2+-selective (ARC) channels. J Physiol 2008; 586:185-95; PMID:17991693
  • Thompson J, Mignen O, Shuttleworth TJ. The N-terminal domain of Orai3 determines selectivity for activation of the store-independent ARC channel by arachidonic acid. Channels (Austin) 2010; 4:398-410; PMID:20818184; http://dx.doi.org/10.4161/chan.4.5.13226
  • Sun GY, Horrocks LA, Farooqui AA. The roles of NADPH oxidase and phospholipases A2 in oxidative and inflammatory responses in neurodegenerative diseases. J Neurochem 2007; 103:1-16; PMID:17561938
  • Kimelberg HK, Nedergaard M. Functions of astrocytes and their potential as therapeutic targets. Neurotherapeutics 2010; 7:338-53; PMID:20880499; http://dx.doi.org/10.1016/j.nurt.2010.07.006
  • Kuchibhotla KV, Lattarulo CR, Hyman BT, Bacskai BJ. Synchronous hyperactivity and intercellular calcium waves in astrocytes in Alzheimer mice. Science 2009; 323:1211-5; PMID:19251629; http://dx.doi.org/10.1126/science.1169096
  • Kettenmann H, Kirchhoff F, Verkhratsky A. Microglia: new roles for the synaptic stripper. Neuron 2013; 77:10-8; PMID:23312512; http://dx.doi.org/10.1016/j.neuron.2012.12.023
  • Aguzzi A, Barres BA, Bennett ML. Microglia: scapegoat, saboteur, or something else? Science 2013; 339:156-61; PMID:23307732; http://dx.doi.org/10.1126/science.1227901
  • Mosher KI, Wyss-Coray T. Microglial dysfunction in brain aging and Alzheimer's disease. Biochem Pharmacol 2014; 88:594-604; PMID:24445162; http://dx.doi.org/10.1016/j.bcp.2014.01.008
  • Streit WJ, Xue QS, Tischer J, Bechmann I. Microglial pathology. Acta Neuropathol Commun 2014; 2:142; PMID:25257319; http://dx.doi.org/10.1186/s40478-014-0142-6
  • Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, Ransohoff RM, Greenberg ME, Barres BA, Stevens B. Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner. Neuron 2012; 74:691-705; PMID:22632727; http://dx.doi.org/10.1016/j.neuron.2012.03.026
  • Brawek B, Schwendele B, Riester K, Kohsaka S, Lerdkrai C, Liang Y, Garaschuk O. Impairment of in vivo calcium signaling in amyloid plaque-associated microglia. Acta Neuropathol 2014; 127:495-505; PMID:24407428; http://dx.doi.org/10.1007/s00401-013-1242-2