You are on page 1of 8

[Channels 2:4, 1-8; July/August 2008]; ©2008 Landes Bioscience

Review

The STIM/Orai coupling machinery


Irene Frischauf, Rainer Schindl, Isabella Derler, Judith Bergsmann, Marc Fahrner and Christoph Romanin*

Institute for Biophysics; University of Linz; Linz, Austria

Key words: CRAC, STIM, Orai


This manuscript has been published online, prior to printing.Once the issue is complete and page numbers have been assigned, the citation will change accordingly.

E .
Calcium (Ca2+) entry into non-excitable cells is mainly carried Ca2+ entry’ hypothesis in which the depletion of ER Ca2+ stores

UT
by store-operated channels (SOCs), which serve essential func- itself activates Ca2+ channels in the plasma-membrane to refill
tions ranging from regulation of transcription to cell growth. The internal stores (later renamed store-operated calcium entry or

RIB
best-characterized store-operated current, ICRAC, is the calcium SOCE).4 Putney’s hypothesis has received strong support from Ca2+-
release-activated calcium (CRAC) current initially discovered imaging experiments in various cell types where inhibitors of the
in T-lymphocytes and mast cells. The search for the molecular ER-Ca2+ uptake like thapsigargin have been shown to activate Ca2+

IST
components of the CRAC channel lasted over 20 years. Recently entry from the extracellular milieu without the involvement of cell
STIM1 has been identified as the Ca2+ sensor in the endoplasmic surface receptors or IP3.5 Subsequently the use of the patch-clamp

D
reticulum (ER) that accumulates into punctae close to the plasma- technique has enabled to identify and characterize a store-operated
membrane following store-depletion. The identification of STIM1 Ca2+ current, the so called Ca2+ release—activated Ca2+ (CRAC)

OT
has been closely followed by the discovery of Orai1 as the CRAC current, or ICRAC, in mast cells6 as well as in Jurkat T-cells.7,8 The
channel pore in human T-cells. Upon punctae formation STIM1 distinctive biophysical characteristics of the CRAC channel comprise
activates Ca2+ influx via Orai1 channels. This review covers func-
tional details concerning the activation cascade of the STIM1/
ON a high selectivity for Ca2+, a pronounced inwardly rectifying current/
voltage relationship and a very low single channel conductance.9
Orai1 complex from ER Ca2+ sensing to Ca2+ influx through
.D
Many studies have clearly demonstrated that the CRAC channel
Orai1. Furthermore, functional domains within STIM1 and Orai1 is required for the response of T-cells to antigens,10,11 for prolif-
in comparison to their structural homologs STIM2 as well as Orai2 eration and cytokine secretion.12 However, despite intense studies
CE

and Orai3, respectively, are displayed together with recent findings over 20 years, the decoding of the molecular choreography of the
IEN

on the pore architecture and selectivity filter of Orai channels. A CRAC channel has turned out to be very challenging. For example,
broad tissue expression of STIM and Orai proteins together with proteins of the canonical transient receptor potential (TRP) family
substantial effects in STIM1/Orai1 knock-out mice suggests an have been for a long time in the focus as components of CRAC
SC

essential physiological role in store-operated Ca2+ signalling in or SOC channels.9 A variety of CRAC blockers have so far been
human health and disease. identified,13 however, due to their non-specificity in the context
BIO

with other channels and signalling pathways, these compounds


Discovery of STIM and Orai Proteins have been less valuable for the identification of CRAC channels.14
Cytosolic calcium signals are adjusted by intracellular sources Emphasized studies on the elucidation of the CRAC components
ES

such as the endo-/sarcoplasmic reticulum, Ca2+ binding proteins and and their signalling machinery have remained inefficient until high-
via plasma-membrane Ca2+ permeable channels. The intracellular throughput RNAi screens (searching for molecules involved in SOC
ND

free calcium, Ca2+i, is essential for various cellular processes ranging influx of Drosophila S2 and HeLa cells) have led to the identifica-
from contraction and secretion to transcription, cell growth and cell tion of a protein that is required for store-operated Ca2+ entry: the
LA

division.1,2 The endoplasmic reticulum (ER) functions as a store for stromal interaction molecule 1 (STIM1).15,16 STIM1 functions as an
the intracellular pool of Ca2+ which is released when ER-localized ER calcium-sensor10,15 that translocates to defined punctae close to
08

inositol-1,4,5-triphosphate (IP3) receptors are activated, a process the plasma-membrane following store-depletion thereby activating
triggerd by the phospholipase C product IP3, allowing for Ca2+ CRAC currents.10 The elucidation of STIM1 as a Ca2+ sensor has
20

efflux into the cytosol.3 Due to the limited capacity of the ER to been closely followed by the identification of the CRAC channel’s
store Ca2+, it needs to be refilled following depletion. More than pore-forming subunit, named Orai1/CRACM1.17-19 Up to now,
©

two decades ago, in 1986, James Putney has proposed a ‘­capacitative three human homologs, termed Orai1-3 have been identified.
These proteins contain four transmembrane segments with both
*Correspondence to: Christoph Romanin; Institute for Biophysics; University of N- and C-termini located intracellularly.18 Unveiling Orai1 as a
Linz; Altenbergerstr. 69; Linz 4040 Austria; Tel.: 43.732.2468.9272; Fax: CRAC component has been additionally carried out by a modified
43.732.2468.9280; Email: christoph.romanin@jku.at
linkage analysis, where a naturally occurring single point mutation
Submitted: 07/20/08; Accepted: 07/21/08 at position 91 (R91W) results in impaired T-cell signalling, which
Previously published online as a Channels E-publication: leads to severe combined immunodefiency (SCID) syndrome. This
http://www.landesbioscience.com/journals/channels/article/6705 single point mutation disrupts ICRAC in human T-cells of SCID

1 Channels 2008; Vol. 2 Issue 4


The STIM/Orai coupling machinery

patients,11,17-19 however, the mechanism leading to complete loss of transmitting ER store depletion to STIM1/Orai1 clusters thereby
function is still unresolved. activating store-operated Ca2+ entry.50 TIRF microscopy in combi-
nation with electrophysiology has revealed that STIM1 translocation
Molecular Structure of STIM1 and Orai Proteins proceeds CRAC activation by 6–10 seconds compatible with an
STIM1 domains. STIM1, initially characterised as a phospho- upstream step in CRAC channel activation.45 In addition, fluorescence
protein,20 includes a single transmembrane domain located in the ER quenching and microscopic analysis visualise that STIM1 clusters
in resting cells (Fig. 1A). The N-terminus of STIM1 stretches into colocalise with areas of elevated Ca2+ concentrations from endog-
the ER lumen, while the C-terminus extends into the cytosol. Within enous CRAC or coexpressed Orai1 channels47 and are located within
the former, a single EF-hand Ca2+-binding motif acts as luminal Ca2+ a distance of 10–25 nm from the plasma-membrane.45 Moreover,
sensor.10,21 A sterile-alpha motif (SAM), including two N-linked upon store-depletion Orai1 proteins require STIM1 co-expression to
glycosylation sites, is followed by the transmembrane domain and a form a clustered localisation in the plasma-membrane at sites in close
cytosolic C-terminus with two coiled-coil regions overlapping with proximity to STIM1 punctae.47,51 The process of store-operated
an ezrin-radixin-moesin (ERM)-like domain. Subsequent gluta- STIM1 aggregation has been demonstrated by FRET microscopy to
mate-, serine/proline-, serine/threonine- and lysine-rich regions are be fully reversible following store repletion.49,51 A STIM1 EF-hand
additionally located at the C-terminus (Fig. 1A).10,22-24 STIM2, mutant that is no longer able to sense ER Ca2+ concentration,
the second protein within the STIM family, is structurally 61% already exhibits a punctuate pattern. Furthermore, expression of the
homologous to STIM1. Both proteins diverge significantly in their STIM1 EF-hand mutant results in constitutive CRAC currents even
C-terminal region after the ERM/coiled-coil domain.25 when stores are full.10,16,19,34 It is under debate if store-depletion
Orai domains. All three Orai proteins, containing four predicted mediated translocation of STIM1 results in insertion into or accu-
transmembrane segments (Fig. 1B), form Ca2+ selective plasma- mulation underneath the plasma-membrane. A partial integration of
membrane channels. Within the cytosolic strands, the N-terminus STIM1 into the plasma-membrane has been reported by a number
of only Orai1 includes a proline/arginine-rich region.26 In their of laboratories employing biotinylation experiments.16,20,21,24,52,53
C-terminus all three Orai proteins contain a putative coiled-coil Gill and coworkers have proposed an interaction of endoplasmic
domain, a common protein interaction motif (Fig. 1B), with varying and plasma-membrane localised STIM1, due to a constant STIM1
predicted probabilities which are 5–6 fold higher for Orai2 and Orai3 plasma-membrane expression independent of store-depletion.16 In
than for Orai1 [http://www.isrec.isb-sib.ch/webmarcoil/webmar- contrast, an increase in plasma-membrane expression of STIM1
coilC1.html; http://www.ch.embnet.org/software/COILS_form. upon store-depletion has been observed with immunofluorescence,
html]. The extracellular loop of Orai1 between the third and fourth immuno-electron microscopy and biotinylation experiments.21,53
transmembrane segment comprises an N-glycosylation site.26,27 Moreover, a small hexahistidine-Zn2+-tag fused to the N-terminus
However, it has been shown not to be essential for Orai1 function.28 of STIM1 has allowed demonstrating its plasma membrane inser-
Based on coimmunoprecipitation experiments, Orai proteins are able tion following store-depletion. It is of note that large N-terminal
to assemble into homo- as well as heteromers.29,30 Co-expression of fusion proteins in contrast to C-terminal ones interfere with STIM1
STIM1 with Orai1 is required to fully reconstitute CRAC currents plasma-membrane insertion.54 Functional evidence for plasma-
while their expression alone results in no significant increase in or membrane STIM1 is presented by antibodies against an N-terminal
even reduced store-operated Ca2+ entry.31,32 Additionally co-expres- sequence of STIM1 that results in partial suppression of SOCs,16,55
sion of STIM1 with Orai2 as well as Orai3 resulted in Ca2+ current but could not be confirmed in another study.52 Reports that observe
activation with biophysical characteristics similar to CRAC currents, STIM1 distribution underneath but not in the plasma-membrane
however, with slightly different selectivity profiles and differences have been widely based on TIRF microscopy using YFP-tagged
in feedback to intracellular Ca2+.33-35 The selectivity filter of Orai STIM1 as well as other, even larger tags. In line, antibodies against
pores are formed by acidic residues in transmembrane domain one these fluorophores fail to detect any insertion of YFP-STIM1 into
and three and the first loop domain.27,32,36,37 Also STIM2 is able to the plasma-membrane.10,34 As CFP or YFP N-terminally tagged
activate all three Orai proteins.38 STIM1 is apparently able to activate Orai1 currents to a similar
extent as C-terminally tagged,51 a major role of plasma-membrane
Store-Operated Activation of STIM/Orai
inserted STIM1 for the activation of Orai1 currents seems unlikely.
Store-depletion dependent STIM1 redistribution. STIM1 Functional STIM1 domains. STIM1 displays multiple roles
mediated robust Orai1 currents are initially stimulated by Ca2+ in the CRAC signalling cascade, first sensing the decrease of ER
store-depletion sensed by the STIM1 luminal EF-hand. In resting Ca2+ concentration via its EF-hand, followed by its aggregation
cells, STIM1 is uniformly distributed within the ER, displays and punctae formation that culminates in activation of CRAC
tubular structures and binds to the microtubule-plus-end-tracking channels.49 These diverse actions are accomplished by distinct
protein EB1 at those sites where microtubule ends come in contact structures within both N- and C-termini of the STIM1 protein.
with the ER.39 Moreover, STIM1 co-localises with endogenous An N-terminal fragment consisting of both EF-hand and SAM
α-tubulin.23,40 The STIM1 EF-hand is adequate to sense a decrease domain exists in vitro as a monomer when Ca2+ is bound and as an
in the ER Ca2+ level that is approximately 300–500 μM at rest.41-43 oligomer when Ca2+ is absent.56 Hence, this correlates with in vivo
Subsequently, STIM1 forms oligomers before it redistributes with observations, where STIM1 is evenly distributed when Ca2+ stores
an EC50 of 210 μM Ca2+,44 into punctuate clusters close to the are filled, and following ER depletion it forms oligomers.45,49,51
plasma-membrane.10,21,23,34,45-49 It has recently been shown that In line, co-immunoprecipitation experiments suggest an essential
oligomerisation of about four STIM1 proteins is the critical process role of the N-terminal part, including the SAM domain in STIM1-

www.landesbioscience.com Channels 2
The STIM/Orai coupling machinery

Figure 1. (A and B) Predicted structures of the Orai and STIM protein families. (A) Structure of STIM1 and STIM2. STIM proteins are single-transmembrane
spanning proteins located in the ER, with their N-termini extending into the ER lumen. The N-termini contain a Ca2+-sensing EF-hand and an SAM domain. The
single transmembrane domain is followed by the C-terminus containing two coiled-coil motifs and an overlapping ERM domain with a subsequent glutamate-,
serine/proline- and a lysine-rich region. Only STIM1 additionally comprises a glutamine- and a serine/threonine-rich region. (B) Structure of Orai1, Orai2
and Orai3, with only Orai1 containing two proline- and one arginine- rich region in its N-terminal strand. All three Orai proteins consist of four transmem-
brane regions and a putative coiled-coil domain in their C-terminus which is predicted with an about 5-6-fold higher probability for Orai2 and Orai3 than
for Orai1. Further, Orai3 displays a much longer second extracellular loop than Orai1 and Orai2. (C) Model for STIM1/Orai1 interaction. Following ER
store depletion and punctae formation, STIM1 directly or indirectly interacts with Orai1 triggering CRAC channel activation. On the one hand it is proposed
that the Orai1/STIM1 coupling is mediated by intermediate steps probably involving CIF.61 Nevertheless, this might be not required for the STIM1 C-terminal
fragment that activates Orai1 currents independent of store-depletion. On the other hand, the C-terminus of STIM1 may directly interact with the C-terminus
of Orai1 as shown by in vitro pulldown experiments, probably via coiled-coil interactions.51 Further auxiliary proteins including calmodulin (CaM) may also
regulate/modulate STIM1-Orai1 coupling.

3 Channels 2008; Vol. 2 Issue 4


The STIM/Orai coupling machinery

STIM1 homomeric interaction.57 Deletion of the SAM domain (aa have failed to detect an interaction.28 The distance between ER
122–199)23 prevents punctae formation and CRAC activation. The and PM estimated in a range between 10–25 nm45 is in principle
C-terminus of STIM1 seems particularly important for Orai/CRAC small enough to allow direct STIM1/Orai1 interaction. Balla and
activation, as the cytosolic C-terminal strand alone is sufficient to coworkers have utilized chemically inducible bridges of different
stimulate both endogenous CRAC24 as well as Orai1 currents.30,51 length to define the distance between plasma- and ER-membrane
This fragment shows predominantly targeting to the plasma- in an attempt to identify whether STIM1, Orai1 and STIM1/Orai1
membrane when co-expressed with Orai151 in HEK293 cells, but assemblies fit into distinct ER plasma-membrane space.58 Before
it is incapable of punctuate clustering yet closely co-localises with store-depletion STIM1 localises in regions of a 4–6 nm ER-PM
Orai1.16,51 A STIM1 mutant lacking the C-terminus fails to co-loca- distance, while Orai1 requires distances in the range of about 11–14
lise with co-expressed Orai1 upon store depletion and is diffusely nm. Upon store-depletion STIM1 moves into the larger regions
distributed throughout the ER membrane.29 Deletion of the coiled- and colocalizses with Orai1 suggesting that Orai1 channels are part
coil domain results in much larger tubularvesicular arrangement of a larger macromolecular complex. An extended conformation of
and this mutant lacks homo-oligomerisation and punctae formation STIM1 probably might span the distance to one of the Orai1 strands.
upon store-depletion as well as CRAC activation.23,24 Additionally We and others demonstrated by FRET microscopy a distance closer
disruption of the C-terminus after the ERM domain similarly than 10 nm between CFP/YFP-labels linked to STIM1 and Orai1
impairs STIM1 redistribution and fails to activate both CRAC and in store-depleted cells.51,59 Moreover a cytosolic STIM1 C-terminal
Orai1.29 However, deletion of a glutamate-rich domain that overlaps fragment couples to Orai1 in in vitro as well as in vivo studies and
with the first coiled-coil in the ERM domain, as well as the serine/ accordingly activates constitutive Orai1 currents.51 Thus, at least
proline-rich regions (aa 600–629) has been reported to be less essen- in the case of STIM1 C-terminus we propose a rather direct than
tial for CRAC activation.24 Deletion of the lysine-rich domain at the indirect interaction of these two proteins via their C-termini. STIM1
very end of the C-terminus has been proposed to eliminate punctae coupling to Orai1 as well as STIM1 homomerization is reversed
formation despite preserved STIM1-STIM1 interaction. Thus the when Ca2+ stores are refilled.10,51
lysine-rich domain allows the separation of STIM1 homomeric Alternative to store-dependent direct coupling that may involve
interaction from punctae formation.49 Moreover, this mutant results the putative coiled-coil domains of STIM1 and Orai1 as depicted
in delayed stimulation when co-expressed with Orai1.29 Therefore in the model (Fig. 1C), a yet unidentified Ca2+ influx factor (CIF)
both coiled-coil domain and lysine-rich region of STIM1 are essen- might additionally be involved. STIM1 has been proposed to
tial for Orai1/CRAC activation. trigger the production of CIF,60 which could contribute to signal
The role of STIM2. Worms and flies only possess one STIM transmission from STIM1 to Orai1.61 CIF has been suggested
protein, while mammals include both STIM1 and STIM2.10,15 Both to displace calmodulin (CaM) from phospholipase A2 (iPLA2),
proteins are approximately 61% homologous and show ubiquitous stimulating lysophospholipid generation that in turn activates
tissue expression. Besides homomeric STIM1-STIM1 interaction, CRAC.62 However, bromenolactone (BEL), an inhibitor of iPLA2
STIM1 can form heteromers with STIM2.46,55 Co-expression of fails to prevent activation of heterologously co-expressed STIM1/
both STIM proteins favors translocation of STIM2 into punctae Orai1 currents14 in contrast to its blocking effect onto endogenous
upon store depletion.46 Functionally Soboloff et al.,46 have also CRAC in RBL cells and SOC in smooth muscle cells.63 Additionally
revealed that STIM2 is a powerful SOC inhibitor when expressed in Ca2+ dependent CaM binding has been detected within a peptide
HEK293, PC12, A7r5 and Jurkat T cells. including residues 667–685 in STIM1 C-terminus.64 A CaM
In the absence of Ca2+, STIM2 does not similarly aggregate as binding domain search [http://calcium.uhnres.utoronto.ca/ctdb/
STIM1.25 This functional difference is due to their distinct EF-hand ctdb/sequence.html] has revealed additional putative CaM binding
Ca2+ affinity. While the EF-hand of STIM1 binds Ca2+ with low sites both in the C-terminus of STIM1 as well as in the N-terminus
affinity in a range ideally adjusted to sense substantial changes in of Orai1 (Fig. 1C). Since CIF has been proposed to displace CaM
ER Ca2+ concentrations, the EF-hand of STIM2 is more sensitive to from PLA2, in the context with the STIM1/Orai1 system, it might
mild reduction of ER Ca2+.44 STIM2 already forms punctae with an be interesting to see whether CIF would also be able to displace CaM
EC50 at 406 μM ER Ca2+ that represents resting levels. Accordingly, either from STIM1 or Orai1 as an additional way of modulation
STIM2 knock-down in Jurkat T-cells selectively lowers basal cyto- within the STIM1/Orai1 signaling cascade.
solic and ER Ca2+ concentrations. Therefore STIM2 is partially In recent studies, Orai1 domains that are involved in the coupling
active at resting ER Ca2+ levels,44 covering both store-dependent process to STIM1 and Ca2+ current activation have been identified.
or -independent modes of Orai1 activation.38 A plausible model While the C-terminus of Orai1 is required for STIM1 coupling and
suggests that STIM2 is part of a feedback module that keeps basal subsequent current activation, Orai1 N-terminus is suggested to be
cytosolic and ER Ca2+ concentrations within tight limits. STIM2 essential for Orai1 gating.29,51 A highly conserved 26 amino acid
deficiency, however, imposes a smaller effect on Ca2+ influx than long N-terminal region immediately before the first transmembrane
STIM1 knock-out. domain is essential for channel activation.29 Moreover, the proline/
STIM1 and Orai1 coupling. Following store-depletion labeled arginine-rich domain in Orai1 N-terminus has a regulatory role
STIM1 proteins form clusters in close proximity to Orai1 leading as Orai2 chimeras containing the N-terminus of Orai1 have been
to CRAC channel activation.29,47,48 However, it remains unre- reported to show enhanced Ca2+ entry.65 Orai1 C-terminal dele-
solved if the two proteins interact directly or if the signal cascade tion mutants fail to colocalize with STIM1 clusters upon Ca2+
involves a third, intermediate component. While some laboratories store-depletion29,51 and accordingly lack ­store-operated ­activation.51
report co-immunoprecipitation of STIM1 and Orai1,32,36 others Pull-down experiments provide evidence for an interaction of STIM1

www.landesbioscience.com Channels 4
The STIM/Orai coupling machinery

C-terminus with the C-terminus rather than the N-terminus of Orai1 selective channels.28,37,71 The more conservative Orai1-E190D27 as
further underscoring the role of Orai1 C-terminus for coupling to well as Orai3-E165D70 mutations show similar pore characteristics
STIM1. Especially a predicted putative coiled-coil domain within like wild-type Orai channels in both a Ca2+ and a monovalent bath
Orai1 C-terminus has been suggested as functional coupling domain, solution. The aspartates D110/112/114 in the first extracellular
as a destabilizing single point mutation (L273S) eliminates coupling loop additionally contribute to the increased Ca2+ selectivity of
to STIM1 and Ca2+ current activation.51 Therefore, it is tempting Orai1 as their mutation to alanine (D110/112/114A) enhanced
to speculate that potential direct interaction of STIM1 and Orai1 monovalent cation permeation.36,68 Structurally these mutations of
is mainly mediated by their C-terminal coiled-coil domains— the pore-relevant amino acids lower Ca2+ selectivity and result in a
possibly together with additional auxiliary components (see Fig. 1C). striking increase in Cs+ permeation, which suggests an enlargement
Accordingly, a putative coiled-coil domain is also predicted within of the unusually narrow pore of the CRAC channel, relieving steric
the C-terminus of Orai2 and Orai3, even with a 5–6 fold higher hindrance for Cs+ permeation. In addition, the mutations diminish
probability than that of Orai1. This may allow for a tighter or at least Ca2+-mediated fast inactivation, a key mode of CRAC channel
distinct coupling to STIM1 that has still to be evaluated. regulation.68 Pore mutants that modify Orai1 channel voltage sensi-
tivity include point mutation V102I as well as V105I close to the
Channel Architecture of Orai Channels selectivity filter.72 In conclusion these studies reveal that structural
Oligomerisation of Orai1 proteins. Many ion channel fami- elements contributing to ion permeation are located close to those
lies have been shown to require a multimeric assembly of various involved in the voltage gating of Orai1 channels.
numbers of individual subunits.66 Coimmunoprecipitation28,36 Comparison of Orai1, Orai2 and Orai3. The three highly homol-
studies have shown for Drosophila Orai as well as mammalian ogous Orai1, Orai2 and Orai3 proteins are widely expressed at the
Orai1 that they assemble to dimers or higher order multimers. mRNA level and display similar plasma-membrane expression.28,73
Moreover all combinations of heteromeric Orai channel assemblies All three channels activate in a store-dependent manner when
are possible.28,30,33 A recent study67 has demonstrated that the func- co-expressed with STIM1, while the extent of their current ampli-
tional CRAC channel pore is formed by a tetrameric assembly of tudes is 2–3 fold smaller for Orai2 and Orai3 compared to Orai1.
Orai1 subunits. This evidence is based on maximal CRAC currents These reduced currents may reflect differences in their expression
developed after expression of preassembled tandem Orai1 multimers levels, efficiency in coupling to STIM1 and distinct single channel
comprising different numbers of subunits into cells stably overex- properties.33 Moreover the three Orai homologs exhibit distinct
pressing STIM1. Assembly of Orai1 channels is mainly mediated properties in terms of selectivity for Ca2+ and Na+ and strikingly
by their transmembrane domains29,51 as deletion of one of their different feedback regulation by intracellular Ca2+.33 Orai3 chan-
cytosolic strands has not affected interaction of Orai1 channels. nels undergo a lower degree of depotentiation than Orai1 or Orai2.
Additionally the N-terminus of Orai1 functions dominant negative Nonetheless the Na+ currents through Orai1, Orai2 and Orai3 as well
on co-expressed Orai1 channels.65 However, discrete domains that as the endogenous store-operated Na+ currents in HEK293 cells, are
are involved in the multimerisation remain so far elusive. all inhibited by extracellular Ca2+ with a half-maximal concentration
Orai pore architecture. Further evidence that Orai channels form of approximately 20 μM.35 Further, the three Orai proteins exhibit
the pore of CRAC channels is provided by point mutations in the differential pharmacological effects in response to 2-aminoethoxydi-
putative selectivity filter of Orai1. The narrowest region of native phenyl borate (2-APB). While STIM1-mediated Orai1 currents are
CRAC, Orai1 or Orai3 pore is only ~3.9 Å wide.37,68,69 Amino stimulated by low concentrations of 2-APB (5–10 μM), high concen-
acids that form part of the selectivity filter in Orai1 include E106 trations (50 μM) completely inhibit Orai1 and partially suppress
in the first transmembrane domain, E190 in the third transmem- Orai2 currents.30,33,35,37,71 In contrast, Orai3 has been demonstrated
brane domain and D110, D112 as well as D114 in the first loop to be exclusively activated by 2-APB independent of store-depletion
domain.27,32,36 While the glutamates within the transmembrane and STIM1, leading to an increased conductance for monovalent
domains are fully conserved in Orai2 and Orai3, the three aspar- ions.30,33,37,71 Substitution of the 2nd and 3rd transmembrane domain
tates (D110, D112, D114) in the loop domain of Orai1 are either of Orai1 with those of Orai3 conferred 2-APB stimulation onto
glutamic acids or glutamines in Orai2 (E84, Q86, Q88) and Orai3 Orai1.30 Moreover single point mutations within the selectivity filter
(E85, D87, E89). The glutamates and aspartates play an essential of Orai3 have been reported to affect 2-APB sensitivity.37 Hence, the
role for the Orai1 channel’s high Ca2+ selectivity, its Ca2+ block, positively charged 2-APB74 might exert part of its stimulatory action
regulation by Ca2+ and determine its pore geometry.36 Mutation via interaction with the Orai3 selectivity filter together with further
of the respective glutamates in Orai1-E106 and -E190 in the first structural requirements located within transmembrane 2 and 3 that
and third transmembrane region as well as corresponding E81 and finally result in stimulated currents with altered permeation proper-
E165 in Orai3, alters ion selectivity, which underlines that Orai1 is a ties. For a more detailed view on other drugs that affect STIM as well
pore subunit of the CRAC channel. Orai1-E106D36 and an analog as Orai proteins, we refer to Derler et al.14 Besides these three Orai
Orai3-E81D37 show reduced Ca2+ selectivity in comparison to wild- channels, further Orai2 splice variants have been identified on two
type. In contrast Orai1-E106Q acts in a dominant negative manner chromosomal loci,73 one containing an intronless gene and a second
on all three STIM1 mediated Orai Ca2+ currents33,36 as well as locus that gives rise to the splice variants Orai2 long (Orai2L) and
endogenous CRAC currents in T-cells.27,28 Yet this E106Q mutant Orai2 short (Orai2S). When co-expressed with STIM1 both Orai2S
is able to couple to STIM1.70 The mutation E190Q or E190A in and Orai2L represent Ca2+ selective channels.73
the third transmembrane region of Orai1, as well as Orai3-E165Q Physiological role of STIM and Orai channels. CRAC channels
leads to a leftward shift in the reversal potential resulting in less Ca2+ are key mediators of sustained Ca2+ signaling in T-cells mediating

5 Channels 2008; Vol. 2 Issue 4


The STIM/Orai coupling machinery

stable formation of immunological synapses. When a T-cell gets in EF-hand mutant of STIM1 revealed macrothrombocytopenia and a
contact with an antigen-presenting dendritic cell, both STIM1 and bleeding disorder. A preactivation state of platelets has been observed
Orai1 redistribute to the immunological synapse, leading to Ca2+ as a result of an increase in basal intracellular calcium levels leading
influx.70 These elevated calcium levels induced by CRAC channels to platelet consumption.87 Hence, STIM1 displays an essential role
prolong dephosphorylation of NFAT (nuclear factor of activated in the regulation and function of platelets.
T-cells) and accordingly its accumulation into the nucleus where it acts In hepatocytes bile acids lead to punctae formation of STIM1 and
as a transcription factor in the early immune response of T-cells.75,76 activation of Ca2+-selective SOC currents. Further knock-down of
Moreover long-term function of CRAC channel activation includes STIM1 has caused inhibited calcium entry activated by bile acids.88
lymphocyte proliferation, effector functions as well as differentiation Human T-cells that contain a point mutation in Orai1 R91W,
of naïve T-cells.77 The importance of Ca2+ influx through CRAC which has recently been identified in SCID patients result in
channels in T-cells is further highlighted by the existence of at least defective T-cell signalling.11,17,89 It causes a complete loss of store-
three families of patients with SCID resulting in severe defects in operated Ca2+ entry into T-cells, however, the detailed mechanism
function of store-operated Ca2+ entry, impaired cytokine expression for this impairment of Ca2+ flux across Orai1 R91W is so far elusive.
and lymphocyte function.17,77,78 In addition CRAC channels play In contrast to human T-cells that require Orai1 for CRAC channel
an important role in mast cell activation, degranulation as well as formation, mice may utilize Orai2, as their thymocytes and T-cells
secretion of proinflammatory lipid mediators.79,80 express much more Orai2 than Orai1 or Orai3. Orai1 plays a major
Knock-down of either STIM or Orai proteins has been shown to role in mice mast cells, as mice deficient in Orai1 exhibit defective
impair SOCE/CRAC entry in a variety of cell types.10,15-18,21,31,46,77,79‑82 mast cell degranulation and cytokine secretion, lack allergic reactions
STIM1 as well as STIM2 are detectable in various primary and are smaller in size.78,80
lymphocytes such as Th, TC and B-cells.83 Furthermore both are In contrast, store-operated calcium influx of naïve T cells, their
expressed at a low, uniform level in heart, brain, kidney, thymus, development and proliferation are unaffected in Orai1-deficient
lung, spleen, skeletal muscle, small intestine as well as in primary mice.78,80 However, differentiated T-cells have been identified to
aortic endothelial cells.83,84 High expression levels of STIM1 are express higher amounts of Orai1 than of Orai2 and Orai3 consistent
found in certain cell types and cellular regions such as the central and with a severe but incomplete loss of store-operated Ca2+ entry. B-cells
the peripheral nervous systems as well as platelets.55 Nothern blot express besides Orai1 and Orai2, Orai3 transcripts at highest amounts
analysis of heart, brain, kidney, thymus, lung, spleen, skeletal muscle, among all lymphocytes. Orai1 deficient splenic B-cells exhibit also a
small intestine, placenta, primary aortic endothelial and mast cells decrease in store-operated Ca2+ entry. Moreover, Orai1-/- mice suffer
suggests an ubiquitous expression of Orai1 and Orai3 proteins, while from eyelid irritation as well as sporadic hair loss and display much
Orai2 is predominantly expressed in brain and to a lower extent in thinner skin with fewer, more elongated keratinocytes.78
the lung, spleen and small intestine.28,73,84 Moreover all three Orai Conclusions and perspectives. It has taken 20 years of exhaus-
proteins are expressed in primary lymphocytes. With the exception of tive studies on store-operated calcium entry before Orai1 and
a lower expression of Orai3 in skeletal muscle and placenta, it seems STIM1 could be identified as the major components of ICRAC in
to be expressed in the same tissues as Orai1. human T-cells. SOCs have been intensively examined over these past
In mice the regulatory role of STIM as well as Orai proteins years, revealing an involvement in a wide range of cellular processes
has been recently examined in particular by knock-out and knock- together with their pharmacological, physiological and pathophysi-
down studies. Mouse T-cells and fibroblasts lacking STIM1 (either ological roles.9 In the focus of autoimmune and inflammatory
by knock-out or siRNA) exhibit impaired SOCE, while it was only immune disorders an involvement of dysregulated Ca2+ signaling is
moderately diminished in STIM2-deficient T-cells.77 STIM1, as well out of question. The recent identification of STIM1 and Orai1-3
as STIM2-knock-out T-cells, show less cytokine production, nuclear and their role in calcium signaling through SOC/CRAC channels
translocation of NFAT and additionally the number of regulatory opens the repertoire for targeting autoimmune diseases, e.g., rheuma-
T-cells is decreased. Thus, these STIM proteins are required as a toid arthritis, inflammatory disorders or allograft rejection.14 Orai1
prerequisite for T-cell development.77,79 seems to be of primary interest as in SCID patients the point muta-
Moreover STIM1 has been identified as critical to mast cell func- tion Orai1 R91W abolishes CRAC currents only in T-cells, whereas
tion, since STIM1-deficient mast cells of mice display much less currents from B-cells and fibroblasts are only reduced.17 Yet there are
degranulation and cytokine production after FcεRI stimulation. no specific inhibitors of the STIM1/Orai1 pathway available to test
Hence, STIM1 acts as a key for mast cell activation and anaphy- the effect of blocking this pathway in vivo.14 With the discovery of
laxis.79 Besides the requirement of STIM1 in non-excitable cells, STIM1 and Orai1 as the key components of CRAC channels, a solid
there is further evidence that STIM1 has a regulatory role in excit- basis is reached to study their molecular composition, stoichiometry
able cells, such as skeletal muscle and myotubes. STIM1 has been and activation mechanism. STIM1 as well as Orai1 have been shown
demonstrated to be expressed in both tissues. Mice lacking STIM1 to interact with members of the canonical transient receptor potential
die perinatally from a skeletal myopathy and their myotubes fail channels, a family of phospholipase C regulated channels.24,53,90-95
to show SOC and fatigue rapidly.85 Further STIM1 is abundantly Further studies are required to reveal the nature of SOCE channels
expressed in platelets. STIM1-deficient platelets display a marked in various tissues, possibly manifested by a combination of various
defect in agonist-induced Ca2+ response, impaired activation and Orai and TRP channels. This will clarify the contribution of STIM1
thrombus formation under flow suggesting STIM1 as an impor- and Orai1 to SOCE in different tissues and will provide a more
tant mediator of ischemic cardio- and cerebrovascular events.86 A widespread view of store-operated channels and their physiological
mouse cell line expressing a constitutively CRAC current activating roles in health and disease.

www.landesbioscience.com Channels 6
The STIM/Orai coupling machinery

Acknowledgements 34. Mercer JC, Dehaven WI, Smyth JT, et al. Large store-operated calcium selective currents
due to co-expression of Orai1 or Orai2 with the intracellular calcium sensor, Stim1. J Biol
This work was supported by Austrian Science Fund project Chem 2006; 281:24979-90.
P18169 to Christoph Romanin. 35. DeHaven WI, Smyth JT, Boyles RR, et al. Calcium inhibition and calcium potentiation
of Orai1, Orai2 and Orai3 calcium release-activated calcium channels. J Biol Chem 2007;
References 282:17548-56.
1. Berridge MJ, Bootman MD, Roderick HL. Calcium signalling: dynamics, homeostasis and 36. Vig M, Beck A, Billingsley JM, et al. CRACM1 multimers form the ion-selective pore of
remodelling. Nat Rev Mol Cell Biol 2003; 4:517-29. the CRAC channel. Curr Biol 2006; 16:2073-9.
2. Berridge MJ, Lipp P, Bootman MD. The versatility and universality of calcium signalling. 37. Schindl R, Bergsmann J, Frischauf I, et al. 2-aminoethoxydiphenyl borate alters selectivity
Nat Rev Mol Cell Biol 2000; 1:11-21. of Orai3 channels by increasing their pore size. J Biol Chem 2008; 283:20261-7.
3. Streb H, Irvine RF, Berridge MJ, et al. Release of Ca2+ from a nonmitochondrial intracellu- 38. Parvez S, Beck A, Peinelt C, et al. STIM2 protein mediates distinct store-dependent and
lar store in pancreatic acinar cells by inositol-1,4,5-trisphosphate. Nature 1983; 306:67-9. store-independent modes of CRAC channel activation. Faseb J 2008; 22:752-61.
4. Putney JW Jr. A model for receptor-regulated calcium entry. Cell Calcium 1986; 7:1-12. 39. Grigoriev I, Gouveia SM, van der Vaart B, et al. STIM1 is a MT-plus-end-tracking protein
5. Putney JW Jr, Bird GS. The inositol phosphate-calcium signaling system in nonexcitable involved in remodeling of the ER. Curr Biol 2008; 18:177-82.
cells. Endocr Rev 1993; 14:610-31. 40. Smyth JT, DeHaven WI, Bird GS, et al. Role of the microtubule cytoskeleton in the func-
6. Hoth M, Penner R. Depletion of intracellular calcium stores activates a calcium current in tion of the store-operated Ca2+ channel activator STIM1. J Cell Sci 2007; 120:3762-71.
mast cells. Nature 1992; 355:353-6. 41. Brini M, Pinton P, Pozzan T, et al. Targeted recombinant aequorins: tools for monitoring
7. Lewis RS, Cahalan MD. Mitogen-induced oscillations of cytosolic Ca2+ and transmem- Ca2+ in the various compartments of a living cell. Microsc Res Tech 1999; 46:380-9.
brane Ca2+ current in human leukemic T cells. Cell Regul 1989; 1:99-112. 42. Yu R, Hinkle PM. Rapid turnover of calcium in the endoplasmic reticulum during signal-
8. Zweifach A, Lewis RS. Mitogen-regulated Ca2+ current of T lymphocytes is activated by ing. Studies with cameleon calcium indicators. J Biol Chem 2000; 275:23648-53.
depletion of intracellular Ca2+ stores. Proc Natl Acad Sci USA 1993; 90:6295-9. 43. Montero M, Alonso MT, Albillos A, et al. Mitochondrial Ca2+-induced Ca2+ release medi-
9. Parekh AB, Putney JW Jr. Store-operated calcium channels. Physiol Rev 2005; ated by the Ca2+ uniporter. Mol Biol Cell 2001; 12:63-71.
85:757-810. 44. Brandman O, Liou J, Park WS, et al. STIM2 is a feedback regulator that stabilizes basal
10. Liou J, Kim ML, Heo WD, et al. STIM is a Ca2+ sensor essential for Ca2+-store-depletion- cytosolic and endoplasmic reticulum Ca2+ levels. Cell 2007; 131:1327-39.
triggered Ca2+ influx. Curr Biol 2005; 15:1235-41. 45. Wu MM, Buchanan J, Luik RM, et al. Ca2+ store depletion causes STIM1 to accumulate in
11. Feske S, Prakriya M, Rao A, et al. A severe defect in CRAC Ca2+ channel activation and ER regions closely associated with the plasma membrane. J Cell Biol 2006; 174:803-13.
altered K+ channel gating in T cells from immunodeficient patients. J Exp Med 2005; 46. Soboloff J, Spassova MA, Hewavitharana T, et al. STIM2 Is an Inhibitor of STIM1-
202:651-62. Mediated Store-Operated Ca2+ Entry. Curr Biol 2006; 16:1465-70.
12. Lewis RS. Calcium signaling mechanisms in T lymphocytes. Annu Rev Immunol 2001; 47. Luik RM, Wu MM, Buchanan J, et al. The elementary unit of store-operated Ca2+ entry:
19:497-521. local activation of CRAC channels by STIM1 at ER-plasma membrane junctions. J Cell
13. Franzius D, Hoth M, Penner R. Non-specific effects of calcium entry antagonists in mast Biol 2006; 174:815-25.
cells. Pflugers Arch 1994; 428:433-8. 48. Xu P, Lu J, Li Z, et al. Aggregation of STIM1 underneath the plasma membrane induces
14. Isabella Derler, Reinhard Fritsch, Rainer Schindl, et al. CRAC inhibitors: Identification and clustering of Orai1. Biochem Biophys Res Commun 2006; 350:969-76.
potential. Expert Op Drug Discovery 2008; 3:787-800. 49. Liou J, Fivaz M, Inoue T, et al. Live-cell imaging reveals sequential oligomerization and local
15. Roos J, DiGregorio PJ, Yeromin AV, et al. STIM1, an essential and conserved component plasma membrane targeting of stromal interaction molecule 1 after Ca2+ store depletion.
of store-operated Ca2+ channel function. J Cell Biol 2005; 169:435-45. Proc Natl Acad Sci USA 2007; 104:9301-6.
16. Spassova MA, Soboloff J, He LP, et al. STIM1 has a plasma membrane role in the activation 50. Liou J, Fivaz M, Inoue T, et al. Oligomerization of STIM1 couples ER calcium depletion
of store-operated Ca2+ channels. Proc Natl Acad Sci USA 2006; 103:4040-5. to CRAC channel activation. Nature 2008; 454:538-42.
17. Feske S, Gwack Y, Prakriya M, et al. A mutation in Orai1 causes immune deficiency by 51. Muik M, Frischauf I, Derler I, et al. Dynamic coupling of the putative coiled-coil
abrogating CRAC channel function. Nature 2006; 441:179-85. domain of ORAI1 with STIM1 mediates ORAI1 channel activation. J Biol Chem 2008;
283:8014-22.
18. Vig M, Peinelt C, Beck A, et al. CRACM1 is a plasma membrane protein essential for store-
operated Ca2+ entry. Science 2006; 312:1220-3. 52. Mignen O, Thompson JL, Shuttleworth TJ. STIM1 regulates Ca2+ entry via arachidonate-
regulated Ca2+-selective (ARC) channels without store depletion or translocation to the
19. Zhang SL, Yeromin AV, Zhang XH, et al. Genome-wide RNAi screen of Ca2+ influx identi-
plasma membrane. J Physiol 2007; 579:703-15.
fies genes that regulate Ca2+ release-activated Ca2+ channel activity. Proc Natl Acad Sci USA
2006; 103:9357-62. 53. López JJ, Salido GM, Pariente JA, et al. Interaction of STIM1 with endogenously expressed
human canonical TRP1 upon depletion of intracellular Ca2+ stores. J Biol Chem 2006;
20. Manji SS, Parker NJ, Williams RT, et al. STIM1: a novel phosphoprotein located at the cell
281:28254-64.
surface. Biochim Biophys Acta 2000; 1481:147-55.
54. Hauser CT, Tsien RY. A hexahistidine-Zn2+-dye label reveals STIM1 surface exposure. Proc
21. Zhang SL, Yu Y, Roos J, et al. STIM1 is a Ca2+ sensor that activates CRAC channels and
Natl Acad Sci USA 2007; 104:3693-7.
migrates from the Ca2+ store to the plasma membrane. Nature 2005; 437:902-5.
55. Dziadek MA, Johnstone LS. Biochemical properties and cellular localisation of STIM pro-
22. Smyth JT, Dehaven WI, Jones BF, et al. Emerging perspectives in store-operated Ca2+ entry:
teins. Cell Calcium 2007; 42:123-32.
roles of Orai, Stim and TRP. Biochim Biophys Acta 2006; 1763:1147-60.
56. Stathopulos PB, Li GY, Plevin MJ, et al. Stored Ca2+ depletion-induced oligomerization of
23. Baba Y, Hayashi K, Fujii Y, et al. Coupling of STIM1 to store-operated Ca2+ entry through stromal interaction molecule 1 (STIM1) via the EF-SAM region: An initiation mechanism
its constitutive and inducible movement in the endoplasmic reticulum. Proc Natl Acad Sci for capacitive Ca2+ entry. J Biol Chem 2006; 281:35855-62.
USA 2006; 103:16704-9.
57. Williams RT, Senior PV, Van Stekelenburg L, et al. Stromal interaction molecule 1
24. Huang GN, Zeng W, Kim JY, et al. STIM1 carboxyl-terminus activates native SOC, I(crac) (STIM1), a transmembrane protein with growth suppressor activity, contains an extracel-
and TRPC1 channels. Nat Cell Biol 2006; 8:1003-10. lular SAM domain modified by N-linked glycosylation. Biochim Biophys Acta 2002;
25. Zheng L, Stathopulos PB, Li GY, et al. Biophysical characterization of the EF-hand and 1596:131-7.
SAM domain containing Ca2+ sensory region of STIM1 and STIM2. Biochem Biophys Res 58. Várnai P, Tóth B, Tóth DJ, et al. Visualization and manipulation of plasma membrane-
Commun 2008; 369:240-6. endoplasmic reticulum contact sites indicates the presence of additional molecular compo-
26. Cahalan MD, Zhang SL, Yeromin AV, et al. Molecular basis of the CRAC channel. Cell nents within the STIM1-Orai1 Complex. J Biol Chem 2007; 282:29678-90.
Calcium 2007; 42:133-44. 59. Barr VA, Bernot KM, Srikanth S, et al. Dynamic movement of the xalcium sensor STIM1
27. Prakriya M, Feske S, Gwack Y, et al. Orai1 is an essential pore subunit of the CRAC chan- and the calcium channel Orai1 in activated T cells: Puncta and distal caps. Mol Biol Cell
nel. Nature 2006; 443:230-3. 2008; 19:2802-17.
28. Gwack Y, Srikanth S, Feske S, et al. Biochemical and functional characterization of Orai 60. Csutora P, Peter K, Kilic H, et al. Novel role for STIM1 as a trigger for calcium influx factor
proteins. J Biol Chem 2007; 282:16232-43. production. J Biol Chem 2008; 283:14524-31.
29. Li Z, Lu J, Xu P, et al. Mapping the interacting domains of STIM1 and Orai1 in Ca2+ 61. Bolotina VM. Orai1, STIM1 and iPLA2b: A view from a different perspective. J Physiol
release-activated Ca2+ channel activation. J Biol Chem 2007; 282:29448-56. 2008; 586:3035-42.
30. Zhang SL, Kozak JA, Jiang W, et al. Store-dependent and -independent modes regulating 62. Smani T, Zakharov SI, Csutora P, et al. A novel mechanism for the store-operated calcium
Ca2+ release-activated Ca2+ channel activity of human Orai1 and Orai3. J Biol Chem 2008; influx pathway. Nat Cell Biol 2004; 6:113-20.
283:17662-71. 63. Smani T, Zakharov SI, Leno E, et al. Ca2+-independent phospholipase A2 is a novel deter-
31. Peinelt C, Vig M, Koomoa DL, et al. Amplification of CRAC current by STIM1 and minant of store-operated Ca2+ entry. J Biol Chem 2003; 278:11909-15.
CRACM1 (Orai1). Nat Cell Biol 2006; 8:771-3. 64. Bauer MC, O’Connell D, Cahill DJ, et al. Calmodulin binding to the polybasic
32. Yeromin AV, Zhang SL, Jiang W, et al. Molecular identification of the CRAC channel by C-termini of STIM proteins involved in store-operated calcium entry. Biochemistry 2008;
altered ion selectivity in a mutant of Orai. Nature 2006; 443:226-9. 47:6089-91.
33. Lis A, Peinelt C, Beck A, et al. CRACM1, CRACM2 and CRACM3 are store-operated 65. Takahashi Y, Murakami M, Watanabe H, et al. Essential role of the N-terminus of murine
Ca2+ channels with distinct functional properties. Curr Biol 2007; 17:794-800. Orai1 in store-operated Ca2+ entry. Biochem Biophys Res Commun 2007; 356:45-52.

7 Channels 2008; Vol. 2 Issue 4


The STIM/Orai coupling machinery

66. Green WN, Millar NS. Ion-channel assembly. Trends Neurosci 1995; 18:280-7.
67. Mignen O, Thompson JL, Shuttleworth TJ. Orai1 subunit stoichiometry of the mamma-
lian CRAC channel pore. J Physiol 2008; 586:419-25.
68. Yamashita M, Navarro-Borelly L, McNally BA, et al. Orai1 Mutations Alter Ion Permeation
and Ca2+-dependent Fast Inactivation of CRAC Channels: Evidence for Coupling of
Permeation and Gating. J Gen Physiol 2007; 130:525-40.
69. Prakriya M, Lewis RS. Regulation of CRAC channel activity by recruitment of silent chan-
nels to a high open-probability gating mode. J Gen Physiol 2006; 128:373-86.
70. Lioudyno MI, Kozak JA, Penna A, et al. Orai1 and STIM1 move to the immunologi-
cal synapse and are upregulated during T cell activation. Proc Natl Acad Sci USA 2008;
105:2011-6.
71. Peinelt C, Lis A, Beck A, et al. 2-Aminoethoxydiphenyl borate directly facilitates and indi-
rectly inhibits STIM1-dependent gating of CRAC channels. J Physiol 2008; 586:3061-73.
72. Spassova MA, Hewavitharana T, Fandino RA, et al. Voltage-gating at the selectivity filter
of the CRAC channel induced by mutation of the Orai1 protein. J Biol Chem 2008;
283:14938-45.
73. Gross SA, Wissenbach U, Philipp SE, et al. Murine ORAI2 splice variants form functional
Ca2+ release-activated Ca2+ (CRAC) channels. J Biol Chem 2007; 282:19375-84.
74. Tao L, Harris AL. 2-aminoethoxydiphenyl borate directly inhibits channels composed of
connexin26 and/or connexin32. Mol Pharmacol 2007; 71:570-9.
75. Hogan PG, Chen L, Nardone J, et al. Transcriptional regulation by calcium, calcineurin and
NFAT. Genes Dev 2003; 17:2205-32.
76. Macian F. NFAT proteins: key regulators of T-cell development and function. Nat Rev
Immunol 2005; 5:472-84.
77. Oh-Hora M, Yamashita M, Hogan PG, et al. Dual functions for the endoplasmic reticulum
calcium sensors STIM1 and STIM2 in T cell activation and tolerance. Nat Immunol 2008;
9:432-43.
78. Gwack Y, Srikanth S, Oh-hora M, et al. Hair loss and defective T and B cell function in
mice lacking ORAI1. Mol Cell Biol 2008; in press.
79. Baba Y, Nishida K, Fujii Y, et al. Essential function for the calcium sensor STIM1 in mast
cell activation and anaphylactic responses. Nat Immunol 2008; 9:81-8.
80. Vig M, DeHaven WI, Bird GS, et al. 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.
81. Soboloff J, Spassova MA, Tang XD, et al. Orai1 and STIM reconstitute store-operated
calcium channel function. J Biol Chem 2006; 281:20661-5.
82. Peel SE, Liu B, Hall IP. A key role for STIM1 in store operated calcium channel activation
in airway smooth muscle. Respir Res 2006; 7:119.
83. Williams RT, Manji SS, Parker NJ, 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.
84. Wissenbach U, Philipp SE, Gross SA, et al. Primary structure, chromosomal localization
and expression in immune cells of the murine ORAI and STIM genes. Cell Calcium 2007;
42:439-46.
85. Stiber J, Hawkins A, Zhang ZS, 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.
86. Varga-Szabo D, Braun A, Kleinschnitz C, et al. The calcium sensor STIM1 is an essen-
tial mediator of arterial thrombosis and ischemic brain infarction. J Exp Med 2008;
205:1583-91.
87. Grosse J, Braun A, Varga-Szabo D, et al. An EF hand mutation in Stim1 causes premature
platelet activation and bleeding in mice. J Clin Invest 2007; 117:3540-50.
88. Aromataris EC, Castro J, Rychkov GY, et al. Store-operated Ca2+ channels and Stromal
Interaction Molecule 1 (STIM1) are targets for the actions of bile acids on liver cells.
Biochim Biophys Acta 2008; 1783:874-85.
89. Carroll HP, McNaull BB, Gadina M. Immunodeficiency is a tough nut to CRAC: the
importance of calcium flux in T cell activation. Mol Interv 2006; 6:253-6.
90. Liao Y, Erxleben C, Yildirim E, et al. Orai proteins interact with TRPC channels and confer
responsiveness to store depletion. Proc Natl Acad Sci USA 2007; 104:4682-7.
91. Liao Y, Erxleben C, Abramowitz J, et al. Functional interactions among Orai1, TRPCs and
STIM1 suggest a STIM-regulated heteromeric Orai/TRPC model for SOCE/Icrac chan-
nels. Proc Natl Acad Sci USA 2008; 105:2895-900.
92. Ong HL, et al. Dynamic assembly of TRPC1-STIM1-Orai1 ternary complex is involved
in store-operated calcium influx. Evidence for similarities in store-operated and calcium
release-activated calcium channel components. J Biol Chem 2007; 282:9105-16.
93. Ambudkar IS, Ong HL, Liu X, et al. TRPC1: the link between functionally distinct store-
operated calcium channels. Cell Calcium 2007; 42:213-23.
94. Worley PF, Zeng W, Huang GN, et al. TRPC channels as STIM1-regulated store-operated
channels. Cell Calcium 2007; 42:205-11.
95. Yuan JP, Zeng W, Huang GN, et al. STIM1 heteromultimerizes TRPC channels to deter-
mine their function as store-operated channels. Nat Cell Biol 2007; 9:636-45.

www.landesbioscience.com Channels 8

You might also like