ArticlePDF AvailableLiterature Review

TRPC1: The link between functionally distinct store-operated calcium channels

Authors:

Abstract and Figures

Although store-operated calcium entry (SOCE) was identified more that two decades ago, understanding the molecular mechanisms that regulate and mediate this process continue to pose a major challenge to investigators in this field. Thus, there has been major focus on determining which of the models proposed for this mechanism is valid and conclusively establishing the components of the store-operated calcium (SOC) channel(s). The transient receptor potential canonical (TRPC) proteins have been suggested as candidate components of the elusive store-operated Ca(2+) entry channel. While all TRPCs are activated in response to agonist-stimulated phosphatidylinositol 4,5, bisphosphate (PIP(2)) hydrolysis, only some display store-dependent regulation. TRPC1 is currently the strongest candidate component of SOC and is shown to contribute to SOCE in many cell types. Heteromeric interactions of TRPC1 with other TRPCs generate diverse SOC channels. Recent studies have revealed novel components of SOCE, namely the stromal interacting molecule (STIM) and Orai proteins. While STIM1 has been suggested to be the ER-Ca(2+) sensor protein relaying the signal to the plasma membrane for activation of SOCE, Orai1 is reported to be the pore-forming component of CRAC channel that mediates SOCE in T-lymphocytes and other hematopoetic cells. Several studies now demonstrate that TRPC1 also associates with STIM1 suggesting that SOC and CRAC channels are regulated by similar molecular components. Interestingly, TRPC1 is also associated with Orai1 and a TRPC1-Orai1-STIM1 ternary complex contributes to SOC channel function. This review will focus on the diverse SOC channels formed by TRPC1 and the suggestion that TRPC1 might serve as a molecular link that determines their regulation by store-depletion.
Content may be subject to copyright.
Cell Calcium 42 (2007) 213–223
TRPC1: The link between functionally distinct
store-operated calcium channels
Indu S. Ambudkar, Hwei Ling Ong, Xibao Liu,
Bidhan Bandyopadhyay, Kwong Tai Cheng
Secretory Physiology Section, GTTB, NIDCR, NIH, Bethesda, MD 20892, USA
Received 25 January 2007; received in revised form 30 January 2007; accepted 31 January 2007
Available online 12 March 2007
Abstract
Although store-operated calcium entry (SOCE) was identified more that two decades ago, understanding the molecular mechanisms that
regulate and mediate this process continue to pose a major challenge to investigators in this field. Thus, there has been major focus on
determining which of the models proposed for this mechanism is valid and conclusively establishing the components of the store-operated
calcium (SOC) channel(s). The transient receptor potential canonical (TRPC) proteins have been suggested as candidate components of
the elusive store-operated Ca2+ entry channel. While all TRPCs are activated in response to agonist-stimulated phosphatidylinositol 4,5,
bisphosphate (PIP2) hydrolysis, only some display store-dependent regulation. TRPC1 is currently the strongest candidate component of
SOC and is shown to contribute to SOCE in many cell types. Heteromeric interactions of TRPC1 with other TRPCs generate diverse SOC
channels. Recent studies have revealed novel components of SOCE, namely the stromal interacting molecule (STIM) and Orai proteins. While
STIM1 has been suggested to be the ER-Ca2+ sensor protein relaying the signal to the plasma membrane for activation of SOCE, Orai1 is
reported to be the pore-forming component of CRAC channel that mediates SOCE in T-lymphocytes and other hematopoetic cells. Several
studies now demonstrate that TRPC1 also associates with STIM1 suggesting that SOC and CRAC channels are regulated by similar molecular
components. Interestingly, TRPC1 is also associated with Orai1 and a TRPC1-Orai1-STIM1 ternary complex contributes to SOC channel
function. This review will focus on the diverse SOC channels formed by TRPC1 and the suggestion that TRPC1 might serve as a molecular
link that determines their regulation by store-depletion.
© 2007 Elsevier Ltd. All rights reserved.
Keywords: Transient receptor potential canonical 1; Store-operated calcium entry; Endoplasmic reticulum; Calcium signaling; Regulatory protein complex
1. The conundrum of store-operated calcium entry
The endoplasmic reticulum (ER) provides a large intra-
cellular Ca2+ store from which Ca2+ can be released under
a number of different conditions. The ER, as well as plasma
membrane (PM) in resting cells have low permeability to
Ca2+ which increases following stimulation of cells by a
wide variety of extracellular ligands as well as physical and
chemical stimuli. Store emptying is most often evoked by
an increase in the level of inositol trisphosphate (IP3)by
Corresponding author at: Building 10, Room 1N-113, NIH, Bethesda,
MD 20892, USA. Tel.: +1 301 496 5298; fax: +1 301 402 1228.
E-mail address: indu.ambudkar@nih.gov (I.S. Ambudkar).
stimulation of a variety of cell surface receptors that are
associated with the activation of PLC and hydrolysis of the
plasma membrane lipid phosphatidylinositol bisphosphate,
PIP2.IP
3induces release of Ca2+ from the ER via activa-
tion of IP3receptor, IP3R[1–4]. While it was recognized
almost two decades ago that this Ca2+ signaling process is
accompanied by activation of plasma membrane Ca2+-entry,
understanding the molecular basis of this mechanism remains
somewhat of a conundrum. This Ca2+ entry pathway, which
was first identified in non-excitable cells, was termed store-
operated Ca2+ entry (SOCE) since it is stimulated in response
to depletion of Ca2+ from intracellular Ca2+ stores in ER
(and inactivated by refilling of these stores), and not due to
PIP2hydrolysis per se. SOCE has been subsequently found
0143-4160/$ see front matter © 2007 Elsevier Ltd. All rights reserved.
doi:10.1016/j.ceca.2007.01.013
214 I.S. Ambudkar et al. / Cell Calcium 42 (2007) 213–223
to be present in neuronal as well as muscle cells. Although
it is clear that other Ca2+ entry pathways are also activated
by agonist-stimulated PIP2hydrolysis, via direct effects of
diacylglycerol or PIP2, this review will focus on SOCE. It is
important to note that other mechanisms can also induce inter-
nal Ca2+ store release and activate Ca2+ entry [3,4]. Recently,
protein synthesis has been associated with release of ER
Ca2+ and activation of SOCE. Termination of protein syn-
thesis and release of nascent polypeptide is associated with
a change in the ion permeability of the ribosome-translocon
complex resulting in Ca2+ release from the ER [5–7]. Thus,
depletion of ER by several different conditions, converge on
the activation of SOCE. In addition to regulating a num-
ber of neurotransmitter-dependent physiological processes,
e.g. T-lymphocyte activation, smooth muscle contraction,
endothelial cell permeability, salivary gland fluid secretion,
etc., SOCE, therefore, also sustains optimal levels of protein
synthesis by maintaining ER-[Ca2+]. It can be suggested that
SOCE is a mechanism devised by the cell to ensure optimal
refilling of the internal Ca2+ stores which is critical for protein
synthesis, cell growth, and proliferation.
SOCE is mediated via the activation of specific plasma
membrane channels, termed store-operated calcium (SOC)
channels [1,2,4], which are ubiquitously expressed in all cell
types. However, and somewhat uniquely, the channel char-
acteristics are diverse in different cell types. The first SOC
channel to be identified, calcium-release-activated calcium
(CRAC) channel, mediates a highly Ca2+-selective calcium
current detected in T-lymphocytes and RBL cells [3,8]. Based
on the methodology and criteria used for measuring and iden-
tifying CRAC channel function, SOC channels with different
biophysical characteristics were subsequently identified in
various cell types [1–3,9,10]. These channels range from non-
selective for Ca2+ to those that are relatively selective. While
the molecular basis for these channels is not clear, it can be
suggested that type of channel formed in a cell is determined
by the physiological function that it serves. An important
question that arises from these significant findings is how all
these diverse channels sense internal Ca2+ store depletion;
i.e. are they all activated by the same ER-PM signal?
Understanding the mechanism by which the status of Ca2+
in the ER is transmitted to the PM, to activate or inacti-
vate SOC, has presented a major challenge to investigators
in this field. Among the many models proposed to describe
this elusive pathway, three have received most attention [4].
(i) Conformational coupling hypothesis which suggests that
ER signal is relayed to the SOC via a direct interaction of the
channel with IP3R in the ER. (ii) The diffusible factor hypoth-
esis suggests that a factor is released or generated in response
to internal Ca2+ store depletion. (iii) The secretion coupling
model proposes regulated recruitment of channels by fusion
of intracellular vesicles. A major roadblock towards under-
standing this mechanism has been the lack of knowledge of
the channel or accessory regulatory components. Over the
past decade, several essential components of SOCE havebeen
identified, including members of transient receptor potential
canonical (TRPC) sub-family of cation channels that have
emerged as central players in this process (further discussed
below) [1,3,11]. However, studies within the last 2 years have
identified new components of SOCE that provide insight into
the possible mechanism by which this process is regulated
and account for the diversity in SOC channels.
2. TRPC1: a molecular component of diverse SOC
channels
Studies with Drosophila TRP channel, which is activated
by a phospholipase C (PLC)-dependent mechanism, led to the
search for mammalian TRP channels, which were suggested
as candidate components of the elusive SOC, store-operated
channel. Consistent with the initial prediction, seven mam-
malian channels (TRPC1–TRPC7) were identified which
were activated in response to stimulation of PIP2hydroly-
sis by stimulation of various plasma membrane receptors in
different tissues [3,11]. One of the major criteria for identifi-
cation of SOC channels has been activation by ER depletion
per se; in absence of receptor-mediated PIP2hydrolysis [3,4].
Thus, only some TRPCs consistently display store-dependent
regulation while others have been suggested to primarily con-
tribute to agonist-stimulated non-store-operated Ca2+ entry
mechanisms. It is important to recognize that there is consid-
erable conflict regarding the mode of activation of a number
of TRPC channels. Part of this discrepancy is due to the use
of heterologously expressed proteins in different cell types
where the background SOC channels could confound the
observations. Relatively less information is available regard-
ing the status of endogenous TRPC channels.
TRPC1, the first mammalian TRPC protein to be identi-
fied, is widely expressed in neuronal as well as non-neuronal
tissues including muscle [3,11–14]. Based on all the studies
reported until now, data demonstrating endogenous TRPC1
as a SOC component have been most consistent. TRPC1 has
been reported to contribute to SOCE in a variety of cell types,
including salivary gland, keratinocytes, platelets, smooth,
skeletal, and cardiac muscle, DT40, HEK293, neuronal,
intestinal, and endothelial cells [15–25]. In contrast, some
studies, primarily with the heterologously expressed pro-
tein describe store-independent regulation of TRPC1 [3,11].
TRPC1 forms diverse SOCs ranging from relatively selective
to non-selective (Ca2+ versus Na+)[26,27]. Some TRPC1-
SOCs display strong anomalous mole-fraction behavior for
Ca2+/Na+permeability which renders them more permeable
to Ca2+ under physiological conditions. Such diversity is pro-
posed to be a result of differences in the composition of the
SOC channels generated by homomeric or heteromeric inter-
actions of TRPC1 with other TRPC monomers. Although
there is no conclusive evidence so far of a native homo-
meric TRPC1 channel, selective interactions of TRPC1 with
other TRPCs, e.g. TRPC4/TRPC5 or TRPC3/TRPC7 have
been reported [23,25,27–32]. TRPC1–TRPC1 multimers are
generated by interaction of the N-terminal coiled–coiled
I.S. Ambudkar et al. / Cell Calcium 42 (2007) 213–223 215
domain interactions [33] while TRPC1–TRPC3 heteromers
are formed by interaction of the first ankyrin repeat region
in TRPC1with an as yet unknown region in the TRPC3
N-terminus [27]. Of these, only interaction with TRPC4,
TRPC3, or TRPC7 has been linked with generation of SOC
channels. There is convincing evidence to show that het-
eromeric TRPC3 + TRPC1 channels are involved in SOCE
in HSY cells [27] and hippocampal neuronal cells [34];
TRPC1 + TRPC4 channels mediate SOCE in endothelial
cells [28] and TRPC1 + TRPC3 + TRPC7 in HEK-293 cells
[27]. These studies provide a molecular basis by which
the previously described diversity in SOC channels can be
explained. Although the exact molecular signal(s) that dic-
tates the interaction of TRPC1 with other TRPCs is not yet
known, it can be suggested that this is likely determined by
the type of channel that is required by the cell to fulfill a
specific physiological function.
In the following section, we will discuss well-studied
TRPC1-SOC channels in various cell types (see Table 1)
including salivary gland cells, platelets, endothelial cells,
smooth and skeletal muscle cells. The voluminous amount of
data reported in the last 8–10 years which describe function
of heterologously expressed TRPC, and the controversies in
those observations, will not be discussed here due to inherent
problems associated with such expression systems [13,35].
2.1. TRPC1-SOC in salivary gland cells
SOCE has been suggested to be critical for salivary fluid
secretion since [Ca2+]iincreases regulate ion channels that
drive water flow out of the gland. Studies regarding the role
of TRPC1 in the SOCE mechanism of salivary gland cells
have primarily been reported by us over the last 8–10 years.
Presently most of the studies have been done with cell lines
and primary cells. We first used a knockdown strategy to
show that endogenous TRPC1 contributes to SOCE in the
human submandibular gland cell line, HSG [19]. We fur-
ther identified a TRPC1-SOC channel in these cells that is
distinct from the CRAC channel in that it is less selective
for Ca2+ (Ca2+/Na+permeability was 40 for SOC versus
400 for CRAC). Activation of this channel by depletion of
internal Ca2+ stores with either IP3or inhibition of SERCA
pump by thapsigargin generates relatively inwardly rectify-
ing currents (Erev = +20 mV). The channel displays strong
anomalous mole fraction behavior for Ca2+ versus Na+per-
meability demonstrating it is relatively more permeable to
Ca2+ under physiological conditions [10]. Single channel
measurements demonstrated single channel conductance of
20 pS. The contribution of TRPC1 to the SOC channel was
confirmed by (i) demonstrating that exogenously expressed
TRPC1 generated channel activity similar to the endogenous
activity, (ii) more importantly, by mutating the proposed-pore
region of the protein, confirmed by topology studies [36],
which resulted in change in the calcium permeability of the
channel, (iii) deletion of the pore region which induced a
dominant suppression of the endogenous current [37], and
(iv) by expressing TRPC1 encoded by an adenovirus in vivo
in rat salivary gland which not only increased salivary flow
in the animal but also increased SOCE in the salivary gland
cells [38]. Yuan et al. [39] also reported a similar TRPC1-
dependent SOC current in HSG cells and further identified
a regulation of TRPC1 by IP3R and the scaffolding protein,
Homer. Together, these important findings provided strong
evidence that TRPC1 is a critical component of the SOC
channel in HSG cells and contributes to the Ca2+ selectivity
of the channel. Whether this is a TRPC1-monomeric channel
or not is not yet known. However, TRPC4 and TRPC3 do not
appear to contribute to SOC channel function in these cells.
Surprisingly, but consistent with the ability of TRPC1 to
form diverse SOC channels, we identified a non-selective
Table 1
Presence and function of TRPC1-SOC channels
Cell type Channels Functions References
Salivary gland C1-C1 (?) Ion channel regulation/fluid secretion [10,19,27,37,38]
C1-C3
Endothelial C1-C1 (?) Permeability/proliferation/inflammation [20,23,28,40,41,43,45–47]
C1-C4
Smooth muscle C1-C1 (?) Contraction, proliferation [15,17,50–53]
C1-C5
Neuronal C1-C3 Post-synaptic signaling/growth/apoptosis [18,30,34]
C1-C1 (?)
C1-C5 (?)
Keratinocytes C1-? Wound healing/proliferation/differentiation/ER stress [16,73]
Cardiac myocytes C1-? Hypertrophy [57]
Skeletal myocytes C1-? Role in muscular dystrophy [24]
Intestinal epithelial cells C1-? Wound healing [22]
HEK-293 C1-C3-C7 ? [25,65–68]
DT-40 C1-? ? [21]
Platelets C1-C4 (?) ? [58–60,62–64]
216 I.S. Ambudkar et al. / Cell Calcium 42 (2007) 213–223
SOC channel in human parotid gland cell line, HSY [27].In
this case, TRPC1 interacts with TRPC3 via its N-terminal
domain. Expression of either the N-terminus of TRPC1 or
TRPC3 in these cells induces dominant suppression of SOC
channel function. In addition to minimal selectivity of Ca2+
versus Na+or Na+versus Cs+, this channel does not display
anomalous mole fraction behavior for Ca2+ versus Na+. The
relative amount of Na+or Ca2+ permeating via this chan-
nel is not yet determined. Neither is it clear why cells that are
functionally quite similar require such diverse SOC channels.
Mouse submandibular gland cells also display a linear ISOC,
although TRPC1 contributes to this channel other TRPC com-
ponents have not yet been identified (Liu et al. unpublished
observations).
2.2. TRPC1-SOC in endothelial cells
Store-depletion-activated Ca2+ entry is a critical deter-
minant of endothelial permeability [23,40–44]. Endothelial
cells express almost all TRPC proteins, specifically TRPC1
and TRPC4 have been identified in all the cells studied thus far
including: HUVEC, human umbilical vein endothelial cells;
HPAEC, human pulmonary artery endothelial cells; HMEC,
human dermal microvascular endothelial cell line; RPAEC,
rat pulmonary artery endothelial cells; BAEC, bovine aor-
tic endothelial cells; MAEC, mouse aortic endothelial cells;
MLEC, mouse lung endothelial cells. There is consider-
able evidence for the contribution of TRPC1 to SOCE in
endothelial cells. TRPC1 is shown to form SOC channels
in HUVEC and HMEC. Increased TRPC1 expression in
HMEC increases thrombin-induced Ca2+ influx and IP3-
sensitive store-operated cationic current [45]. In HUVEC,
VEGF-activated Ca2+ entry is blocked by anti-TRPC1 anti-
body directed against the external pore domain of the protein,
suggesting a role for TRPC1 in regulation of endothelial per-
meability [46]. TRPC1 expression is regulated by TNF-
and this accounts for the enhanced Ca2+ influx in response
to thrombin in HPAEC [45]. Further, this overexpression of
TRPC1 is associated with modulation of the permeability
change induced by thrombin. Thus, TRPC1 expression in
microvascular endothelial cells may contribute to pulmonary
edema associated with acute respiratory distress syndrome.
Groschner et al. [47] have demonstrated the expression
of TRPC1, TRPC3, and TRPC4 in HUVEC utilizing RT-
PCR and on the basis of the inhibition of SOCE induced
by the N-terminal fragment of hTRPC3 have suggested
that SOCE is mediated via TRPC1 + TRPC3 heteromer. A
role for TRPC1 in SOC has also been reported in the
A549 cell line as well as pulmonary artery endothelial cells
[26].
Heteromeric TRPC1 + TRPC4 SOC channels have also
been described in endothelial cells. Brough et al. [26]
have reported that transfection of a TRPC1 specific anti-
sense oligonucleotide in HPAEC markedly reduced the
thapsigargin-induced increase in [Ca2+]iand store-operated
Ca2+ entry current. This current is inwardly rectifying
(Erev = +30 mV), suggesting the presence of a relatively Ca2+
selective channel. This channel is reported to be generated
by heteromeric association of TRPC1 and TRPC4. It is sen-
sitive to changes in actin status since it is anchored to the
cytoskeleton by an interaction of TRPC4 to protein 4.1 [48]
which in turn is tethered to spectrin. SOCE in aortic and lung
endothelial cells is decreased in TRPC4/mice [49] sug-
gesting that TRPC1 cannot function alone and compensate
for the loss of TRPC4. Interestingly, TRPC4 has been asso-
ciated with generation of a channel with high selectivity for
Ca2+ (highest of all known TRPCs) which does not display
any increase in outward currents at mV more positive than
the Erev. Although, a similar highly Ca2+-selective current is
detected in aortic endothelial cells, it is unclear whether it is
mediated via a homomeric TRPC4 channel.
2.3. TRPC1-SOC in smooth muscle cells
TRPC1 is expressed in most types of smooth muscle cells
with the exception of a few [3,13,15] and in various species
such as human, dog, mouse, rabbit, and rat. An antibody to the
pore region of the protein as well as TRPC1-siRNA decrease
SOCE and arterial smooth muscle function, both in primary
and cultured cells. A role for TRPC1 in SOCE has been shown
in smooth muscle cells of caudal, basal, pulmonary artery, and
cerebral arterioles. However, the effect of TRPC1 on SOCE
is not always correlated with changes in function. TRPC1
regulates smooth muscle cell proliferation since TRPC1-
antisense RNA decreased pulmonary artery smooth muscle
cell growth [50]. Furthermore, TRPC1 is upregulated under
conditions that initiate smooth muscle regulation and prolifer-
ation [51,52]. TRPC4 also appears to contribute to the SOCE
together with TRPC1 in pulmonary artery myocytes in cul-
ture [53]. The likelihood of interaction between these TRPC
proteins has been demonstrated convincingly by FRET-based
assays. It has also been suggested that TRPC4 might facilitate
the trafficking and surface expression of TRPC1 [54]. TRPC6
knockdown decreases SOCE in rat pulmonary arterial smooth
muscle cells [55], however, the available data do not support
a heteromeric TRPC6 + TRPC1 SOC channel. How TRPC6
contributes to this SOCE is not known, although it is impor-
tant to note that TRPC6 (/) mice have been associated
with increased TRPC3 expression and exhibit functional sec-
ondary effects due to TRPC3-dependent channel activity.
Thus, the role of TRPC6 in SOCE needs to be examined
in further detail.
Rat aortic smooth muscle cell line, A7r5 express TRPC1,
C4, and C6 [56]. Knockdown of TRPC1 decreased ISOC in
these cells suggesting that TRPC1 contributes to the SOC
channel in these cells. Notably, ISOC in these cells was quite
similar to that seen in HSG and smooth muscle cells; i.e. rel-
atively inwardly rectifying. While the role of TRPC4 in this
SOC cannot be ruled out, this study rules out a contribution of
TRPC6 based on the differences in the currents that are asso-
ciated with the two proteins. It is important to note that TRPCs
are not uniformly distributed in all types of SMCs. Further,
I.S. Ambudkar et al. / Cell Calcium 42 (2007) 213–223 217
TRPC1 transcript is not expressed in some of them (e.g. from
mouse and rat cerebral artery, human trachea, canine pul-
monary artery, mouse and canine colon, and rat mesenteric
artery). Thus, it will be important to determine what other
TRPC contribute to SOCE in these cells. In terms of the
properties, SOC channels in smooth muscle cells vary from
highly selective to relatively non-selective. The SOC current
in rabbit pial arteriole smooth muscle has been recently been
proposed to be generated via a TRPC1 + TRPC5 heteromeric
channel based on the characteristic outward rectification pre-
viously described for this heteromer. This current is quite
distinct from the inwardly rectifying found in A7r5 cells
[56] as well as the non-selective currents reported earlier
in rabbit and rat aortic smooth muscles [9]. Recently roles
for TRPC1 and TRPC3 have been reported in skeletal and
cardiac muscle, respectively [24,57].
2.4. TRPC1-SOC in platelets
Studies by Sage and co-workers strongly suggest the
involvement of TRPC1 in the SOCE mechanism in platelets
[58–60]. Activation of platelets has been associated with
increased interaction of TRPC1 with IP3Rs. In addition,
conditions that disrupt this interaction have been shown to
attenuate SOCE. SOCE is also directly linked with regulation
of platelet aggregation induced by a number of different stim-
uli. However, there is some discrepancy in the data reported
for platelets which primarily arises due to the differences
observed in the localization and expression of different TRPC
proteins in platelets by two groups. While Sage and co-
workers have suggested plasma membrane localization of
TRPC1, Authi reported that TRPC1 is primarily localized
intracellularly [61]. Since TRPC1 trafficking to the plasma
membrane as well as its retention there is shown to depend
on several factors [61], it is unclear whether differences in
any of these can account for the observed discrepancies.
TRPC6 is expressed in platelets in relatively high abundance
and forms DAG-activated channels, although it is unclear
whether it contributes to SOC. Thus, the present data sug-
gest the SOC channels in platelets are made up of TRPC1,
possibly together possibly with other TRPCs, e.g. TRPC4,
[59–64]. There is presently no information on the biophys-
ical properties of this channel, which could provide some
insight and potential resolution of the controversies regarding
its components.
2.5. TRPC1-SOC in HEK293 cells
Despite the fact that this is the most widely used cell
for study of TRP channel function, there is little data
regarding its endogenous SOC channels. This is possi-
bly an important contributing factor to the contradictory
findings in studies using heterologous expression of var-
ious TRPCs in these cells. The available data regarding
the molecular components of endogenous SOCE in HEK-
293 cells has been reported primarily by Villereal and
co-workers [25,65–67]. In a systematic study using a knock-
down approach they have demonstrated that TRPC1, TRPC3,
and TRPC7 contribute to the SOCE in these cells [25]. The
role of TRPC1 + TRPC3 in generating a non-selective SOC
was suggested earlier by Klaus Groschner’s group [68].On
the other hand over expression systems have not always
been consistent with these findings. TRPC3, when expressed
at relatively low levels, TRPC2, as well as TRPC7 have
been reported to generate SOC channels in these cells [3].
It is unclear whether these functions are dictated by the
interaction of the expressed TRPCs with an endogenous
protein; e.g. TRPC1. This should be given serious consid-
eration since other studies have shown that heterologously
expressed TRPCs can co-IP with each other and although
TRPC1/TRPC4/TRPC5 or TRPC3/TRPC6/TRPC7 form two
groups whose members seem to associate with each other,
exceptions have been reported. Importantly, such exceptions;
e.g. TRPC1/TRPC3/TRPC7 appear to require the presence of
TRPC1 in the channel [30].
2.6. TRPC1-SOC in neuronal cells
Several studies demonstrate a role for TRPC1 in neuronal
cells. A role for TRPC1 in the proliferation of embryonic
rat neural stem cells was recently reported. Store deple-
tion generated a non-selective current that was attenuated
by antisense-TRPC1 [18]. While TRPC2, C3, C4, C6 tran-
scripts were detected in these cells, their involvement in
SOC channel function was not assessed. A TRPC1 + TRPC3-
SOC channel was suggested in proliferating hippocampal
neurons [34]. In these cells, expression of TRPC1- and
TRPC3-mRNA and protein dramatically increased, while the
expression of TRPC4 and TRPC7 mRNA and protein dramat-
ically decreased in parallel with a 3.4-fold increase in the level
of SOCE. siRNA-induced knockdown of TRPC1 and TRPC3
indicate they are involved in mediating SOCE, TRPC4 and
TRPC7 were not involved. Inhibition of SOCE also decreased
cell proliferation suggesting a role for TRPC1 + TRPC3-
SOC channel in cell proliferation. TRPC1 also contributes
to SOCE in SH-SY5Y human neuroblastoma cells which
have been associated with a pro-survival effect on the
cells [69,70] and mediates mGluR1-dependent Ca2+ entry
[71].
2.7. TRPC1-SOCs in other cell types
In addition to the role of TRPC1 in SOCE in the cell
types discussed above, it is also shown to generate store-
dependent non-selective cation channels in CHO cells [72].
In gingival keratinocytes [16,73,74] where SOCE regulates
cell differentiation, two SOC currents have been described
which are activated sequentially by store depletion; an ICRAC-
like Ca2+ selective current that is initially activated followed
by a larger non-selective cation current. The former has been
associated with TRPC4 and the latter with TRPC1. Thus, in
these cells TRPC1 and TRPC4 appear to form independent
218 I.S. Ambudkar et al. / Cell Calcium 42 (2007) 213–223
SOC channels. TRPC1 is also involved in SOCE in LNCaP
human prostate cancer epithelial cells [75–77]. The currents
associated with TRPC1 in these cells are relatively inwardly
rectifying and dependent on IP3Rs as well as the cytoskele-
tal status. There is only one report showing that TRPC1
contributes to ICRAC. This study in DT40 cells reveals that
deletion of TRPC1 protein expression induces almost com-
plete attenuation of the current in about 80% of the cells,
and normal SOCE is restored in these cells by expression
of hTRPC1. In the remaining 20% cells, loss of TRPC1 is
not associated with any changes in ICRAC [21]. The reason
for this diversity in the cell function is unclear. Interestingly,
we have recently reported that TRPC1 is not endogenously
involved in ICRAC in RBL-2H3 cells [78]. However, heterolo-
gous expression of TRPC1 in these cells alters the properties
of CRAC, shifting the Erev to the left. Mutant TRPC1 lacking
the pore region exerts dominant suppression of ICRAC. These
data suggest that TRPC1 can interact heteromerically with
the CRAC channel components in RBL-2H3 cells.
Together the studies discussed above provide strong evi-
dence that endogenous TRPC1 contributes to SOC channels.
Further, they illustrate the remarkable diversity in the char-
acteristics and functions of TRPC1-SOC channels. TRPC1
forms SOC channels by interactions with TRPCs that are sug-
gested to form SOC (e.g. TRPC1 and TRPC4) as well as those
which are not typically associated with generation of SOCs
(e.g. TRPC3, TRPC7). The involvementof the latter is consis-
tent with a previous suggestion that novel interaction between
TRPC proteins requires the presence of TRPC1. An important
question that arises is whether typically store-independent
TRPCs gain store-dependent regulation by interacting with
TRPC1; i.e. is TRPC1 the molecular link in diverse SOC
channels? (see Fig. 1). To address this question, the mecha-
nisms involved in store-dependent activation of TRPC1 have
to be considered.
Fig. 1. Contribution of TRPC1 to diverse SOC channels. Homomeric and
heteromeric SOC channels can be generated by interaction of TRPC1 with
other TRPC monomers, as indicated in the Figure. Some of these TRPCs
are ER-coupled in that they interact with STIM1 while others are not. The
latter can still contribute to generation of SOC via interaction with a TRPC
monomer that is regulated by store-depletion, e.g. TRPC1. For sake of keep-
ing the figure simple, only dimeric channels have been depicted and other
proteins implicated in TRPC1 function; such as IP3R, HOMER, caveolin,
etc., have not been shown.
3. Store-dependent regulation of TRPC1-SOC
The mechanism(s) involved in regulation of SOC chan-
nels continues to be the focus of a large number of studies.
The activity of TRPC1-SOC channels are determined by store
depletion and refilling, by feedback inhibition of Ca2+ entry
which modulates Ca2+ entry after it has been activated, and
by regulation of its surface expression. Plasma membrane
localization of TRPC1 is dependent on its interactions with
other TRPCs (e.g. TRPC1, TRPC4) [54], or other proteins
such as caveolin-1 [79–81],-tubulin (microtubule struc-
tures) [82], and RhoA (remodeling of the cytoskeleton) [20].
TRPC1 interaction with caveolin-1 and RhoA mediates its
localization in caveolar lipid raft domains. Disruption of
these domains abrogates SOCE leading to the suggestion that
TRPC1-SOC channels are assembled within such domains.
A number of reports show that TRPC1 is assembled in a
signalplex that contains key Ca2+ signaling proteins such
PLC, Gq/11 and IP3R[12,79,81] and depending on the cell
type, neurotransmitter and growth factor receptors, such as
bradykinin [83] and FGF receptors [18], and mGluR [71]
which are involved upstream events, leading to PIP2hydrol-
ysis, IP3generation, and release of Ca2+ from the ER; i.e.
events that result in activation of Ca2+ entry. Despite exten-
sive studies, the mechanism that is involved in the final step
of SOC activation, i.e. sensing ER depletion and transmis-
sion of the signal to the PM, is not yet established. IP3R
was the first ER protein to be proposed as a candidate pro-
tein for this function and thus a major focus has been placed
on possible interactions between IP3Rs and TRPC1 [4].In
fact the “conformational coupling” model for activation of
SOCE was based on the ryanodine receptor-dependent regu-
lation of voltage-gated Ca2+ channel in excitation-contraction
coupling in muscle cells which is reminiscent of ER–PM
interactions during regulation of SOCE. Consistent with this,
TRPC channels including TRPC1 have been shown to inter-
act with IP3R. Further, there is convincing evidence that IP3R
regulates TRPC1 function. The scaffolding protein HOMER
was shown to mediate interaction between TRPC1 and IP3R
and disruption of this interaction was associated with TRPC1
activation by ER-Ca2+ depletion [39]. In contrast, an increase
in association of IP3R with TRPC1 has been seen in platelets
and endothelial cells suggesting that the IP3R-TRPC1 inter-
action is required for SOCE [20,84]. CaM, which mediates
Ca2+-dependent feedback inhibition of TRPC1 has also been
proposed to be involved in the regulation of channel function
by IP3R. IP3R and CaM have been proposed to have antag-
onistic effects in the activation of TRPC1, with CaM being
inhibitory and IP3R activating the channel by displacing CaM
[85].
TRPC1 trafficking to the membrane and its assembly in
aCa
2+ channel complex is key to its function and regula-
tion. Within this complex, TRPC1 interacts with scaffolding
as well as regulatory proteins [86–88]. Plasma membrane
expression of TRPC1 is increased following stimulation of
endothelial cells, via a mechanism that is mediated via local
I.S. Ambudkar et al. / Cell Calcium 42 (2007) 213–223 219
cytoskeletal changes. In endothelial cells, channel activa-
tion is also associated with recruitment of TRPC1, IP3R,
and RhoA in a complex [20]. Further, specialized plasma
membrane domains appear to be involved in regulating the
assembly, function, as well as trafficking of TRPC1 [1]. These
domains, lipid raft domains, are enriched in PIP2as well as
sphingolipids, both of which have been implicated in regula-
tion of TRPC channels. Cholesterol and cholesterol-binding
proteins are also concentrated in lipid raft domains. This
lipid structure in the plasma membrane provides a platform
for assembly of TRPC1 channel complexes and facilitates
the regulation of these channels [81]. Local remodeling of
cytoskeleton has also been suggested occur in these regions
since actin-binding proteins such as ezrin are found here.
Thus, despite significant progress in our understanding of
SOC channels and identification of several “candidates” for
the mechanism of regulation, how TRPC1-SOC channels are
actually gated still remains unclear.
4. New findings: insights into old mechanisms?
As discussed above, a considerable amount of data provide
strong evidence that TRPC1 is a component of SOC chan-
nels in a large variety of cells. However, the fact that none
of the TRPCs generate ICRAC has been a source of concern.
The exceptions, as noted above, are TRPC4 in aortic endothe-
lial and smooth muscle cells [42,49] and keratinocytes [74],
TRPC1 in DT40 cells [21], and TRPC3 in T lympho-
cytes [89]. Thus, considerable effort has been made towards
identifying the components of the CRAC channel. Two lab-
oratories using siRNA strategies independently identified
stromal interacting protein1, STIM1, a single transmembrane
domain protein found both in the plasma membrane and
the ER, as a critical protein involved in SOCE, including
CRAC channel function [90,91] Although overexpression of
the protein does not increase SOCE, knockdown effectively
decreases SOCE in several cell types. STIM1 has an EF-
hand domain at its N-terminal end that is localized within
the ER lumen. Thus it has been proposed that STIM1 can
function as a sensor for ER-[Ca2+][92–94]. Depletion of
ER-Ca2+ causes a relocation of STIM1 from a diffuse ER
localization to clusters in peripheral ER-plasma membrane
junctions which have been proposed as the centers where
SOCE occurs [95]. Importantly, TRPC1 also interacts with
STIM1 [63,78,96]. Huang et al. report that the mechanism
involved in STIM1-dependent activation of SOCE and CRAC
channels are identical to those involved in the binding and
activation of STIM1 and TRPC1 [96]. Activity of STIM1
requires an ERM domain, which mediates the selective bind-
ing of STIM1 to TRPC1, 2 and 4, but not to TRPC3, 6 or 7.
Further, TRPC1 forms clusters that coincide with the STIM1
clusters in the sub-plasma membrane region after internal
Ca2+ store depletion. In platelets, store depletion induces an
increase in TRPC1-STIM1 association which is required for
activation of SOCE and depends on the cytoskeletal changes
[63]. Our recent studies [78] are consistent with these find-
ings. We have shown that TRPC1-C terminus interacts with
the C terminus of STIM1 in HSG cells and that this interaction
is involved in regulating TRPC1-SOC function. TRPC1 and
STIM1 co-localize and can be co-immunoprecipitated from
extracts of HSG, mouse submandibular gland cells, HEK-
293 cells, and A7r5 cells. Association between the proteins
is increased by thapsigargin treatment. It is important to note
that IP3Rs also cluster in response to cell stimulation [97].
Whether the IP3R clusters also coincide with the STIM1 and
TRPC1 clusters is not yet known. These observations reveal
similar regulation of CRAC, and TRPC-SOC channels by
STIM1.
Similar siRNA screen as well as genetic linkage stud-
ies in SCID patients identified another protein family (Orai,
which presently has three members). These proteins were ini-
tially suggested as possible regulators of the CRAC channel
since knockdown of their expression decreased ICRAC. More
significantly, mutations in Orai1, found in T lymphocytes
from SCID patients, were associated with defective ICRAC
[92,98]. Subsequently, overexpression of Orai1 and STIM1
was shown to induce large increases in SOCE and ICRAC
and mutations in negatively charged aa residues in the trans-
membrane domains caused changes in the Ca2+ permeability
of CRAC channel [93,99–102]. Thus, Orai1 has been now
been suggested to be a pore-forming component of CRAC
channel. Further, it was reported that STIM1 and Orai1 are
sufficient for generation of functional CRAC channels. How-
ever, studies reported presently suggest that these proteins are
not exclusive to CRAC channels. In addition to the associa-
tion of TRPC1 with STIM1, we have now reported that Orai1
and STIM1 interact with and contribute to TRPC1-SOC chan-
nel. Treatment with siSTIM1 and siOrai1 reduced SOCE
and SOC channel function in HSG cells [78] and HSY cells
(Cheng et al., unpublished observations). Furthermore, we
show that TRPC1, Orai1, and STIM1 are colocalized in the
plasma membrane region of cells and form a ternary complex.
Association of the three proteins is increased upon ER-Ca2+
store depletion. These studies reveal that TRPC1, Orai1, and
STIM1 concertedly determine SOCE in cells where TRPC1 is
an essential pore-forming component of SOC channels. We
have not yet determined whether Orai1 also contributes to
the SOC channel pore. Interestingly, the effects of the Orai1
mutants on channel function are very similar to what we have
previously reported with TRPC1 mutants. Clearly, further
studies are required to asses the role of Orai1 and STIM1
in SOC channels in other cell types. Furthermore, it is quite
possible that other TRPCs might also associate with these
two proteins. In an interesting study, it was observed that
Orai1 can interact with TRPC3 and TRPC6 to mediate store-
dependent regulation of these channels (Liao et al., PNASc, in
press. Personal communication from Dr. Lutz Birnbaumer).
By measuring the effect of Orai1 on the single channel proper-
ties of TRPC3, these investigators have proposed that Orai1
acts as a regulatory, , subunit of TRPC channels. Similar
studies are required for TRPC1-SOC channels to resolve the
220 I.S. Ambudkar et al. / Cell Calcium 42 (2007) 213–223
functional interaction between TRPC1 and Orai1. Part of the
problem with the studies with Orai1 is they have all been car-
ried out with overexpression systems and furthermore, there
are no antibodies for detecting endogenous Orai1. Thus the
status of endogenous Orai1, e.g. the level of expression in
different cells, is yet unclear. Despite this, the compelling
effect of siOrai1 on TRPC1-SOC channels provides strong
evidence that these proteins converge on the same mecha-
nism. Whether Orai1 also contributes to the pore of the SOC
channels or acts as a regulatory subunit, such as those of
ionotropic GluR channels as well as voltage-dependent cal-
cium channels, is yet to be determined. It is interesting that
the subunits affect the trafficking as well as gating of the
“excitable” cell Ca2+ channels.
We propose (see Fig. 1) that TRPC1 is the STIM1 bind-
ing component in TRPC-containing SOC channels. While
TRPC2 and TRPC7 can also link plasma membrane SOC to
ER, TRPC7 is not found in majority of the SOC channels
and TRPC2 is not expressed in human cells. Furthermore,
SOC channels in which TRPC1 involvement has been con-
clusively excluded appear to be rare. Thus a reasonable
hypothesis for how diverse SOC channels are regulated by
the same intracellular signal (ER Ca2+ depletion) is that
TRPC1 links the plasma membrane SOC channel to the ER-
Ca2+ sensor, STIM1. We suggest that TRPC1 and STIM1
can provide a molecular basis for TRPC-dependent SOCE.
TRPC1, together with other TRPC monomers forms the pore-
forming component of the SOC channel while STIM1 serves
as the ER-Ca2+ sensor that relays the ER “signal” to the
channel. Whether this directly results in gating the SOC chan-
nel or other proteins are also involved in this relay is not
yet known. Furthermore, our suggestion does not exclude
the involvement of the other proteins (discussed above) in
SOCE although clearly further studies will be required to
establish the relative contributions of proteins such as IP3R,
HOMER, and STIM1 to SOC channel function. There are no
data available presently to establish whether Orai1 interacts
directly with either STIM1 or TRPC1. Thus, how Orai1 con-
tributes to TRPC1-SOC or even other SOC channels needs
to be further evaluated. We suggest the following possible
models for the role of TRPC1 and Orai1 in SOC channels
(see Fig. 2):
Orai1 and TRPC1 form independent channels: This is not
supported by the activity seen in cells after TRPC1 or Orai1
knockdown (e.g. TRPC1 knockdown does not result in
residual ICRAC-like current); also the activities due to the
two proteins are not additive.
Orai1 and TRPC1 contribute to a single SOC channel:
Conclusive evidence for this is lacking although the knock-
down data do suggest that both proteins contribute to
SOCE. How a 4-TM and 6-TM domain proteins can con-
tribute to one channel pore requires further clarification.
Orai1 is the pore-forming subunit and TRPC1 is a
regulator: Mutations in the charged aa residues in the trans-
membrane domain alters selectivity of Orai1. Although
Fig. 2. Possible role of TRPC1 and Orai1 in SOCE. The figure illustrates
the four possibilities discussed in the text. (i) TRPC1 and Orai1 form inde-
pendent channel, (ii) TRPc1 and Orai1 contribute to the same channel, (iii)
TRPC1 is a regulator of Orai1-channel (indicated by orange arrow at the
top), and (iv) Orai1 is a regulator of TRPC1 channel.
these studies are promising further structure-functional
confirmation is needed to establish whether this is the pore-
region of Orai1. Regulatory subunits of other calcium
channels such as GluRs or VDCCs, which have 4-TMs,
regulate gating, current amplitudes, and trafficking of the
pore-forming subunits. Thus detailed single channel
studies are required to resolve the role of Orai1.
TRPC1 is the pore-forming subunit and Orai1 is the
regulator: There are considerable data defining the pore
properties of TRPC channels. The proposed topology of
TRPC1 is similar to that of other 6-TM channels. Topology
and mutational studies have identified the region spanning
the 5th and 6th TM domain to be the site of the pore.
Thus, there is considerable evidence presently available
to support TRPC1 as the pore-forming unit of SOC chan-
nels. However, the possibility that Orai1 regulates SOC
channels (as described above for TRPC3) needs to be
investigated more thoroughly.
Since STIM1 and Orai1 are recently described proteins,
there are relatively few reports. For example there is no infor-
mation on endogenous Orai1 other than the effects of siRNA
on ICRAC, even these are not substantiated by assessment of
the protein. Further, more detailed studies are required to
substantiate the suggestion that a direct interaction between
Orai1 and STIM1 is involved in generation of CRAC chan-
nel. Despite the lack of such information, identification of
these proteins has provided new directions in the study of
SOCE which could potentially lead to an understanding of
its regulation. There is no doubt that future studies will reveal
important aspects of the role of TRPC1, Orai1, and STIM1
in generation of SOC channel.
5. Concluding remarks
Much effort has been focused on answering the ques-
tion of whether TRPC1 is store-operated or not. Majority
of the studies reported until now suggest that TRPC1 is func-
I.S. Ambudkar et al. / Cell Calcium 42 (2007) 213–223 221
tional component of a variety of native SOCE pathways in
different cell types. TRPC1 generates SOC channels by inter-
action with other TRPC monomers. SOC channels in which
TRPC1 involvement has been conclusively excluded appear
to be rare. Thus, we propose that TRPC1 is a common com-
ponent of diverse SOC channels. Most likely STIM1 is the
ER-regulator of a number of different TRPC1-SOC channels,
whether it regulates all of them is not known. Furthermore,
Orai1 might also interact with TRPC1 and contribute to
its function. While the exact mode by which Orai1 affects
TRPC1-SOC channel function needs to be clarified it appear
that Orail is required for TRPC1 function. Knowledge that
TRPC1 is part of SOCE represents a significant advancement
in our understanding of SOCE. Future studies should address
the important question of how the signal linked to store deple-
tion is actually relayed to TRPC1 for activation of SOCE.
Conflict of interest
None.
References
[1] I.S. Ambudkar, Ca2+ signaling microdomains: platforms for the
assembly and regulation of TRPC channels, Trends Pharmacol. Sci.
27 (2006) 25–32.
[2] G.S. Bird, O. Aziz, J.P. Lievremont, B.J. Wedel, M. Trebak, G.
Vazquez, J.W. Putney Jr., Mechanisms of phospholipase C-regulated
calcium entry, Curr. Mol. Med. 4 (2004) 291–301.
[3] A.B. Parekh, J.W. Putney Jr., Store-operated calcium channels, Phys-
iol. Rev. 85 (2005) 757–810.
[4] J.W. Putney Jr., L.M. Broad, F.J. Braun, J.P. Lievremont, G.S. Bird,
Mechanisms of capacitative calcium entry, J. Cell. Sci. 114 (2001)
2223–2229.
[5] C. Camello, R. Lomax, O.H. Petersen, A.V. Tepikin, Calcium leak
from intracellular stores—the enigma of calcium signalling, Cell Cal-
cium 32 (2002) 355–361.
[6] R. Lomax, C. Camello, F.V. Coppenolle, O.H. Petersen, A.V. Tepikin,
Basal and physiological Ca2+ leak from the endoplasmic reticulum of
pancreatic acinar cells, J. Biol. Chem. 277 (2002) 26479–26485.
[7] H.L. Ong, X. Liu, A. Sharma, R.S. Hegde, I.S. Ambudkar, Intracellu-
lar Ca2+ release via the ER translocon activates store-operated calcium
entry, Pflugers Arch. 453 (2007) 797–808.
[8] A.B. Parekh, R. Penner, Store depletion and calcium influx, Physiol.
Rev. 77 (1997) 901–930.
[9] V.M. Bolotina, Store-operated channels: diversity and activation
mechanisms, Sci. STKE (2004) pe34.
[10] X. Liu, K. Groschner, I.S. Ambudkar, Distinct Ca2+-permeable cation
currents are activated by internal Ca2+-store depletion in RBL-2H3
cells and human salivary gland cells, HSG and HSY, J. Membr. Biol.
200 (2004) 93–104.
[11] C. Montell, The TRP superfamily of cation channels, Sci. STKE 272
(2005) re3.
[12] I.S. Ambudkar, TRPC1: a core component of store-operated calcium
channels, Biochem. Soc. Trans. 35 (2007) 96–100.
[13] G. Rychkov, G.J. Barritt, TRPC1 Ca2+-permeable channels in animal
cells, Handb. Exp. Pharmacol. (2007) 23–52.
[14] X. Zhu, P.B. Chu, M. Peyton, L. Birnbaumer, Molecular cloning of
a widely expressed human homologue for the Drosophila trp gene,
FEBS Lett. 373 (1995) 193–198.
[15] D.J. Beech, Emerging functions of 10 types of TRP cationic channel
in vascular smooth muscle, Clin. Exp. Pharmacol. Physiol. 32 (2005)
597–603.
[16] S. Cai, S. Fatherazi, R.B. Presland, C.M. Belton, F.A. Roberts, P.C.
Goodwin, M.M. Schubert, K.T. Izutsu, Evidence that TRPC1 con-
tributes to calcium-induced differentiation of human keratinocytes,
Pflugers Arch. 452 (2006) 43–52.
[17] A. Dietrich, V.Chubanov, H. Kalwa, B.R. Rost, T. Gudermann, Cation
channels of the transient receptor potential superfamily: their role
in physiological and pathophysiological processes of smooth muscle
cells, Pharmacol. Ther. 112 (2006) 744–760.
[18] A. Fiorio Pla, D. Maric, S.C. Brazer, P. Giacobini, X. Liu, Y.H.Chang,
I.S. Ambudkar, J.L. Barker, Canonical transient receptor potential 1
plays a role in basic fibroblast growth factor (bFGF)/FGF receptor-1-
induced Ca2+ entry and embryonic rat neural stem cell proliferation,
J. Neurosci. 25 (2005) 2687–2701.
[19] X. Liu, W. Wang, B.B. Singh, T. Lockwich, J. Jadlowiec, B.
O’Connell, R. Wellner, M.X. Zhu, I.S. Ambudkar, Trp1, a candi-
date protein for the store-operated Ca2+ influx mechanism in salivary
gland cells, J. Biol. Chem. 275 (2000) 3403–3411.
[20] D. Mehta, G.U. Ahmmed, B.C. Paria, M. Holinstat, T. Voyno-
Yasenetskaya, C. Tiruppathi, R.D. Minshall, A.B. Malik, RhoA
interaction with inositol 1,4,5-triphosphate receptor and transient
receptor potential channel-1 regulates Ca2+ entry, J. Biol. Chem. 278
(2003) 33492–33500.
[21] Y. Mori, M. Wakamori, T. Miyakawa, M. Hermosura, Y. Hara, M.
Nishida, K. Hirose, A. Mizushima, M. Kurosaki, E. Mori, K. Gotoh,
T.Okada, A. Fleig, R. Penner, M. Iino, T. Kurosaki, Transientreceptor
potential 1 regulates capacitative Ca2+ entry and Ca2+ release from
endoplasmic reticulum in B lymphocytes, J. Exp. Med. 195 (2002)
673–681.
[22] J.N. Rao, O. Platoshyn, V.A. Golovina, L. Liu, T. Zou, B.S. Marasa,
D.J. Turner, J.X. Yuan, J.Y. Wang, TRPC1 functions as a store-
operated Ca2+ channel in intestinal epithelial cells and regulates early
mucosal restitution after wounding, Am. J. Physiol. Gastrointest Liver
Physiol. 290 (2006) G782–G792.
[23] C. Tiruppathi, G.U. Ahmmed, S.M. Vogel, A.B. Malik, Ca2+ signal-
ing, TRP channels, and endothelial permeability, Microcirculation 13
(2006) 693–708.
[24] C. Vandebrouck, D. Martin, M. Colson-Van Schoor, H. Debaix, P.
Gailly, Involvement of TRPC in the abnormal calcium influx observed
in dystrophic (mdx) mouse skeletal muscle fibers, J. Cell Biol. 158
(2002) 1089–1096.
[25] T.K. Zagranichnaya, X. Wu, M.L. Villereal, Endogenous TRPC1,
TRPC3, and TRPC7 proteins combine to form native store-operated
channels in HEK-293 cells, J. Biol. Chem. 280 (2005) 29559–
29569.
[26] G.H. Brough, S. Wu, D. Cioffi, T.M. Moore, M. Li, N. Dean, T.
Stevens, Contribution of endogenously expressed Trp1 to a Ca2+ -
selective, store-operated Ca2+ entry pathway, FASEB J. 15 (2001)
1727–1738.
[27] X. Liu, B.C. Bandyopadhyay, B.B. Singh, K. Groschner, I.S. Ambud-
kar, Molecular analysis of a store-operated and 2-acetyl-sn-glycerol-
sensitive non-selective cation channel. Heteromeric assembly of
TRPC1-TRPC3, J. Biol. Chem. 280 (2005) 21600–21606.
[28] D.L. Cioffi, S. Wu, T. Stevens, On the endothelial cell ISOC, Cell
Calcium 33 (2003) 323–336.
[29] T.D. Plant, M. Schaefer, Receptor-operated cation channels formed
by TRPC4 and TRPC5, Naunyn Schmiedebergs Arch. Pharmacol.
371 (2005) 266–276.
[30] C. Strubing, G. Krapivinsky, L. Krapivinsky, D.E. Clapham, For-
mation of novel TRPC channels by complex subunit interactions in
embryonic brain, J. Biol. Chem. 278 (2003) 39014–39019.
[31] S.Z. Xu, G. Boulay, R. Flemming, D.J. Beech, E3-targeted anti-
TRPC5 antibody inhibits store-operated calcium entry in freshly
isolated pial arterioles, Am. J. Physiol. Heart Circ. Physiol. 291 (2006)
H2653–H2659.
222 I.S. Ambudkar et al. / Cell Calcium 42 (2007) 213–223
[32] F. Zeng, S.Z. Xu, P.K. Jackson, D. McHugh, B. Kumar, S.J. Fountain,
D.J. Beech, Human TRPC5 channel activated by a multiplicity of
signals in a single cell, J. Physiol. 559 (2004) 739–750.
[33] M. Engelke, O. Friedrich, P. Budde, C. Schaefer, U. Niemann, C.
Zitt, E. Jungling, O. Rocks, A. Luckhoff, J. Frey, Structural domains
required for channel function of the mouse transient receptor potential
protein homologue TRP1, FEBS Lett. 523 (2002) 193–199.
[34] X. Wu, T.K. Zagranichnaya, G.T. Gurda, E.M. Eves, M.L. Villereal,
A TRPC1/TRPC3-mediated increase in store-operated calcium entry
is required for differentiation of H19-7 hippocampal neuronal cells,
J. Biol. Chem. 279 (2004) 43392–43402.
[35] B. Nilius, From TRPs to SOCs, CCEs, and CRACs: consensus and
controversies, Cell Calcium 33 (2003) 293–298.
[36] Y. Dohke, Y.S. Oh, I.S. Ambudkar, R.J. Turner, Biogenesis and topol-
ogy of the transient receptor potential Ca2+ channel TRPC1, J. Biol.
Chem. 279 (2004) 12242–12248.
[37] X. Liu, B.B. Singh, I.S. Ambudkar, TRPC1 is required for functional
store-operated Ca2+ channels. Role of acidic amino acid residues in
the S5-S6 region, J. Biol. Chem. 278 (2003) 11337–11343.
[38] B.B. Singh, C. Zheng, X. Liu, T. Lockwich, D. Liao, M.X. Zhu, L.
Birnbaumer, I.S. Ambudkar, Trp1-dependent enhancement of salivary
gland fluid secretion: role of store-operated calcium entry, FASEB J.
15 (2001) 1652–1654.
[39] J.P. Yuan, K. Kiselyov, D.M. Shin, J. Chen, N. Shcheynikov, S.H.
Kang, M.H. Dehoff, M.K. Schwarz, P.H. Seeburg, S. Muallem, P.F.
Worley, Homer binds TRPC family channels and is required for gating
of TRPC1 by IP3 receptors, Cell 114 (2003) 777–789.
[40] D.L. Cioffi, T. Stevens, Regulation of endothelial cell barrier function
by store-operated calcium entry,Microcirculation 13 (2006) 709–723.
[41] B. Nilius, G. Droogmans, Ion channels and their functional role in
vascular endothelium, Physiol. Rev. 81 (2001) 1416–1459.
[42] M. Freichel, S.H. Suh, A. Pfeifer, U. Schweig, C. Trost, P. Weigerber,
M. Biel, S. Philipp, D. Freise, G. Droogmans, F. Hofmann, V. Flock-
erzi, B. Nilius, Lack of an endothelial store-operated Ca2+ current
impairs agonist-dependent vasorelaxation in TRP4/mice, Nat.
Cell Biol. 3 (2001) 121–127.
[43] C. Tiruppathi, R.D. Minshall, B.C. Paria, S.M. Vogel, A.B. Malik,
Role of Ca2+ signaling in the regulation of endothelial permeability,
Vascul. Pharmacol. 39 (2002) 173–185.
[44] C. Tiruppathi, M. Freichel, S.M. Vogel, B.C. Paria, D. Mehta, V.
Flockerzi, A.B. Malik, Impairment of store-operated Ca2+ entry in
TRPC4(/) mice interferes with increase in lung microvascular
permeability, Circ. Res. 91 (2002) 70–76.
[45] B.C. Paria, S.M. Vogel, G.U. Ahmmed, S. Alamgir, J. Shroff, A.B.
Malik, C. Tiruppathi, Tumor necrosis factor-alpha-induced TRPC1
expression amplifies store-operated Ca2+ influx and endothelial per-
meability, Am. J. Physiol. Lung Cell Mol. Physiol. 287 (2004)
L1303–L1313.
[46] G.U. Ahmmed, D. Mehta, S. Vogel,M. Holinstat, B.C. Paria, C. Tirup-
pathi, A.B. Malik, Protein kinase Calpha phosphorylates the TRPC1
channel and regulates store-operated Ca2+ entry in endothelial cells,
J. Biol. Chem. 279 (2004) 20941–20949.
[47] K. Groschner, S. Hingel, B. Lintschinger, M. Balzer, C. Romanin,
X. Zhu, W. Schreibmayer, Trp proteins form store-operated cation
channels in human vascular endothelial cells, FEBS Lett. 347 (1998)
101–106.
[48] D.L. Cioffi, S. Wu, M. Alexeyev, S.R. Goodman, M.X. Zhu, T.
Stevens, Activationof the endothelial store-operated ISOC Ca2+ chan-
nel requires interaction of protein 4.1 with TRPC4, Circ. Res. 97
(2005) 1164–1172.
[49] M. Freichel, S. Philipp, A. Cavalie, V. Flockerzi, TRPC4 and
TRPC4-deficient mice, Novartis Found Symp. 258 (2004) 189–199
(discussion 199–203, 263–266).
[50] A. Bergdahl, M.F. Gomez, A.K. Wihlborg, D. Erlinge, A. Eyjolf-
son, S.Z. Xu, D.J. Beech, K. Dreja, P. Hellstrand, Plasticity of TRPC
expression in arterial smooth muscle: correlation with store-operated
Ca2+ entry, Am. J. Physiol. Cell Physiol. 288 (2005) C872–C880.
[51] B. Kumar, K. Dreja, S.S. Shah, A. Cheong, S.Z. Xu, P. Sukumar, J.
Naylor, A. Forte, M. Cipollaro, D. McHugh, P.A. Kingston, A.M.
Heagerty, C.M. Munsch, A. Bergdahl, A. Hultgardh-Nilsson, M.F.
Gomez, K.E. Porter, P. Hellstrand, D.J. Beech, Upregulated TRPC1
channel in vascular injury in vivo and its role in human neointimal
hyperplasia, Circ. Res. 98 (2006) 557–563.
[52] M.J. Lin, G.P. Leung, W.M. Zhang, X.R. Yang, K.P. Yip, C.M. Tse,
J.S. Sham, Chronic hypoxia-induced upregulation of store-operated
and receptor-operated Ca2+ channels in pulmonary arterial smooth
muscle cells: a novel mechanism of hypoxic pulmonary hypertension,
Circ. Res. 95 (2004) 496–505.
[53] S. Zhang, H.H. Patel, F. Murray, C.V. Remillard, C. Schach, P.
Thistlethwaite, P.A. Insel, J.X. Yuan, Pulmonary artery smooth mus-
cle cells from normal subjects and IPAH patients with idiopathic
pulmonary arterial hypertension show divergent cAMP-mediated
effects on TRPC expression and capacitative Ca2+ entry, Am. J. Phys-
iol. Lung Cell Mol. Physiol. 292 (2007) L1202–L1210.
[54] T. Hofmann, M. Schaefer, G. Schultz, T. Gudermann, Subunit com-
position of mammalian transient receptor potential channels in living
cells, Proc. Natl. Acad. Sci. U.S.A. 99 (2002) 7461–7466.
[55] Y. Yu, M. Sweeney, S. Zhang, O. Platoshyn, J. Landsberg, A. Roth-
man, J.X.-J. Yuan, PDGF stimulates pulmonary vascular smooth
muscle cell proliferation by upregulating TRPC6 expression, Am.
J. Physiol. 284 (2003) C316–C330.
[56] L.I. Brueggemann, D.R. Markun, K.K. Henderson, L.L. Cribbs, K.L.
Byron, Pharmacological and electrophysiological characterization of
store-operated currents and capacitative Ca2+ entry in vascularsmooth
muscle cells, J. Pharmacol. Exp. Ther. 317 (2006) 488–499.
[57] H. Nakayama, B.J. Wilkin, I. Bodi, J.D. Molkentin, Calcineurin-
dependent cardiomyopathy is activated by TRPC in the adult mouse
heart, FASEB J. 20 (2006) 1660–1670.
[58] J.A. Rosado, S.L. Brownlow, S.O. Sage, Endogenously expressed
Trp1 is involved in store-mediated Ca2+ entry by conformational
coupling in human platelets, J. Biol. Chem. 277 (2002) 42157–42163.
[59] S.L. Brownlow, S.O. Sage, Transient receptor potential protein sub-
unit assembly and membrane distribution in human platelets, Thromb.
Haemost. 94 (2005) 839–845.
[60] S.L. Brownlow, A.G. Harper, M.T. Harper, S.O. Sage, A role for
hTRPC1 and lipid raft domains in store-mediated calcium entry in
human platelets, Cell Calcium 35 (2004) 107–113.
[61] K.S. Authi, TRP channels in platelet function, Handb. Exp. Pharma-
col. (2007) 425–443.
[62] S.L. Brownlow, S.O. Sage, Rapid agonist-evoked coupling of type
II Ins(1,4,5)P3receptor with human transient receptor potential
(hTRPC1) channels in human platelets, Biochem. J. 375 (2003)
697–704.
[63] J.J. Lopez, G.M. Salido, J.A. Pariente, J.A. Rosado, Interaction of
STIM1 with endogenously expressed human canonical TRP1 upon
depletion of intracellular Ca2+ stores, J. Biol. Chem. 281 (2006)
28254–28264.
[64] I. Wakabayashi, M. Marumo, A. Graziani, M. Poteser, K. Groschner,
TRPC4 expression determines sensitivity of the platelet-type capaci-
tative Ca2+ entry channel to intracellular alkalosis, Platelets 17 (2006)
454–461.
[65] X. Wu, G. Babnigg, M.L. Villereal, Functional significance of human
trp1 and trp3 in store-operated Ca2+ entry in HEK-293 cells, Am. J.
Physiol. 278 (2000) C526–C536.
[66] G. Babnigg, B. Heller, M.L. Villereal, Cell-to-cell variation in store-
operated calcium entry in HEK-293 cells and its impact on the
interpretation of data from stable clones expressing exogenous cal-
cium channels, Cell Calcium 27 (2000) 61–73.
[67] X. Wu, G. Babnigg, T. Zagranichnaya, M.L. Villereal, The role
of endogenous human Trp4 in regulating carbachol-induced cal-
cium oscillations in HEK-293 cells, J. Biol. Chem. 277 (2002)
13597–13608.
[68] B. Lintschinger, M. Balzer-Geldsetzer, T. Baskaran, W.F. Graier, C.
Romanin, M.X. Zhu, K. Groschner, Coassembly of Trp1 and Trp3
I.S. Ambudkar et al. / Cell Calcium 42 (2007) 213–223 223
proteins generates diacylglycerol- and Ca2+-sensitive cation channels,
J. Biol. Chem. 275 (2000) 27799–27805.
[69] S. Bollimuntha, M. Ebadi, B.B. Singh, TRPC1 protects human
SH-SY5Y cells against salsolinol-induced cytotoxicity by inhibiting
apoptosis, Brain Res. 1099 (2006) 141–149.
[70] S. Bollimuntha, B.B. Singh, S. Shavali, S.K. Sharma, M. Ebadi,
TRPC1-mediated inhibition of 1-methyl-4-phenylpyridinium ion
neurotoxicity in human SH-SY5Y neuroblastoma cells, J. Biol. Chem.
280 (2005) 2132–2140.
[71] S.J. Kim, Y.S. Kim, J.P. Yuan, R.S. Petralia, P.F. Worley, D.J. Linden,
Activation of the TRPC1 cation channel by metabotropic glutamate
receptor mGluR1, Nature 426 (2003) 285–291.
[72] C. Zitt, A.G. Obukhov, C. Strubing, A. Zobel, F. Kalkbrenner, A.
Luckhoff, G. Schultz, Expression of TRPC3 in Chinese Hamster
Ovary cells results in calcium-activated cations current not related
to store depletion, J. Cell Biol. 138 (1997) 1333–1341.
[73] S. Cai, S. Fatherazi, R.B. Presland, C.M. Belton, K.T. Izutsu, TRPC
channel expression during calcium-induced differentiation of human
gingival keratinocytes, J. Dermatol. Sci. 40 (2005) 21–28.
[74] S. Fatherazi, R.B. Presland, C.M. Belton, P. Goodwin, M. Al-Qutub,
Z. Trbic, G. Macdonald, M.M. Schubert, K.T. Izutsu, Evidence that
TRPC4 supports the calcium selective I(CRAC)-likecurrent in human
gingival keratinocytes, Pflugers Arch. (2006).
[75] D. Pigozzi, T. Ducret, N. Tajeddine, J.L. Gala, B. Tombal, P. Gailly,
Calcium store contents control the expression of TRPC1, TRPC3 and
TRPV6 proteins in LNCaP prostate cancer cell line, Cell Calcium
(2006).
[76] F. Vanden Abeele, L. Lemonnier, S. Thebault, G. Lepage, J.B. Parys,
Y. Shuba, R. Skryma, N. Prevarskaya, Two types of store-operated
Ca2+ channels with different activation modes and molecular origin
in LNCaP human prostate cancer epithelial cells, J. Biol. Chem. 279
(2004) 30326–30337.
[77] F. Vanden Abeele, Y. Shuba, M. Roudbaraki, L. Lemonnier, K.
Vanoverberghe, P. Mariot, R. Skryma, N. Prevarskaya, Store-operated
Ca2+ channels in prostate cancer epithelial cells: function, regu-
lation, and role in carcinogenesis, Cell Calcium 33 (2003) 357–
373.
[78] H.L. Ong, K.T. Cheng, X. Liu, B.C. Bandyopadhyay, B.C. Paria,
J. Soboloff, B. Pani, Y. Gwack, S. Srikanth, B.B. Singh, D. Gill,
I.S. Ambudkar, Dynamic assembly of TRPC1/STIM1/Orai1 ternary
complex is involved in store operated calcium influx: evidence for
similarities in SOC and CRAC channel components, J. Biol. Chem.
282 (2007) 9105–9116.
[79] I.S. Ambudkar, Cellular domains that contribute to Ca2+ entry events,
Sci. STKE 243 (2004) pe32.
[80] S.C. Brazer, B.B. Singh, X. Liu, W. Swaim, I.S. Ambudkar, Caveolin-
1 contributes to assembly of store-operated Ca2+ influx channels by
regulating plasma membrane localization of TRPC1, J. Biol. Chem.
278 (2003) 27208–27215.
[81] T.P. Lockwich, X. Liu, B.B. Singh, J. Jadlowiec, S. Weiland, I.S.
Ambudkar, Assembly of Trp1 in a signaling complex associated with
caveolin-scaffolding lipid raft domains, J. Biol. Chem. 275 (2000)
11934–11942.
[82] S. Bollimuntha, E. Cornatzer, B.B. Singh, Plasma membrane local-
ization and function of TRPC1 is dependent on its interaction with
beta-tubulin in retinal epithelium cells, Vis. Neurosci. 22 (2005)
163–170.
[83] P. Delmas, Assembly and gating of TRPC channels in signalling
microdomains, Novartis Found Symp. 258 (2004) 75–89 (discussion
89–102, 263–266).
[84] J.A. Rosado, P.C. Redondo, S.O. Sage, J.A. Pariente, G.M. Salido,
Store-operated Ca2+ entry: vesicle fusion or reversible trafficking
and de novo conformational coupling? J. Cell Physiol. 205 (2005)
262–269.
[85] M.X. Zhu, J. Tang, TRPC channel interactions with calmodulin and
IP3 receptors, Novartis Found Symp. 258 (2004) 44–58 (discussion
58–62, 98–102, 263–266).
[86] I.S. Ambudkar, Trafficking of TRP channels: determinants of channel
function, Handb. Exp. Pharmacol. (2007) 541–557.
[87] I.S. Ambudkar, B.C. Bandyopadhyay,X. Liu, T.P. Lockwich, B. Paria,
H.L. Ong, Functional organization of TRPC-Ca2+ channels and reg-
ulation of calcium microdomains, Cell Calcium 40 (2006) 495–504.
[88] K. Kiselyov, D.M. Shin, J.Y. Kim, J.P. Yuan,S. Muallem, TRPC chan-
nels: interacting proteins, Handb. Exp. Pharmacol. (2007) 559–574.
[89] S. Philipp, B. Strauss, D. Hirnett, U. Wissenbach, L. Mery, V.
Flockerzi, M. Hoth, TRPC3 mediates T-cell receptor-dependent cal-
cium entry in human T-lymphocytes, J. Biol. Chem. 278 (2003)
26629–26638.
[90] J. Liou, M.L. Kim, W.D. Heo, J.T. Jones, J.W. Myers, J.E. Ferrell Jr.,
T. Meyer, STIM is a Ca2+ sensor essential for Ca2+-store-depletion-
triggered Ca2+ influx, Curr. Biol. 15 (2005) 1235–1241.
[91] J. Roos, P.J. DiGregorio, A.V. Yeromin, K. Ohlsen, M. Lioudyno,
S. Zhang, O. Safrina, J.A. Kozak, S.L. Wagner, M.D. Cahalan, G.
Velicelebi, K.A. Stauderman, STIM1, an essential and conserved
component of store-operated Ca2+ channel function, J. Cell Biol. 169
(2005) 435–445.
[92] J.T. Smyth, W.I. Dehaven, B.F. Jones, J.C. Mercer, M. Trebak, G.
Vazquez, J.W. Putney Jr., Emerging perspectives in store-operated
Ca2+ entry: Roles of Orai, Stim and TRP, Biochim. Biophys. Acta
1763 (2006) 1147–1160.
[93] J. Soboloff, M.A. Spassova, X.D. Tang, T. Hewavitharana, W. Xu,
D.L. Gill, Orai1 and STIM reconstitute store-operated calcium chan-
nel function, J. Biol. Chem. 281 (2006) 20661–20665.
[94] M.A. Spassova, J. Soboloff, L.P. He, W. Xu, M.A. Dziadek, D.L. Gill,
STIM1 has a plasma membrane role in the activation of store-operated
Ca2+ channels, Proc. Natl. Acad. Sci. U.S.A. 103 (2006) 4040–4045.
[95] R.M. Luik, M.M. Wu, J. Buchanan, R.S. Lewis, The elementary unit
of store-operated Ca2+ entry: local activation of CRAC channels by
STIM1 at ER-plasma membrane junctions, J. Cell Biol. 174 (2006)
815–825.
[96] G.N. Huang, W. Zeng, J.Y. Kim, J.P. Yuan, L. Han, S. Muallem, P.F.
Worley, STIM1 carboxyl-terminus activates native SOC, I(crac) and
TRPC1 channels, Nat. Cell Biol. 8 (2006) 1003–1010.
[97] C. Hisatsune, K. Mikoshiba, Novel compartment implicated in cal-
cium signaling—is it an “induced coupling domain”? Sci. STKE
(2005) pe53.
[98] S. Feske, Y. Gwack, M. Prakriya, S. Srikanth, S.H. Puppel, B. Tanasa,
P.G. Hogan, R.S. Lewis, M. Daly, A. Rao, A mutation in Orai1 causes
immune deficiency by abrogating CRAC channel function, Nature
441 (2006) 179–185.
[99] C. Peinelt, M. Vig, D.L. Koomoa, A. Beck, M.J. Nadler, M. Koblan-
Huberson, A. Lis, A. Fleig, R. Penner, J.P. Kinet, Amplification of
CRAC current by STIM1 and CRACM1 (Orai1), Nat. Cell Biol. 8
(2006) 771–773.
[100] M. Prakriya, S. Feske, Y. Gwack, S. Srikanth, A. Rao, P.G. Hogan,
Orai1 is an essential pore subunit of the CRAC channel, Nature 443
(2006) 230–233.
[101] A.V. Yeromin, S.L. Zhang, W. Jiang, Y. Yu, O. Safrina, M.D. Caha-
lan, Molecular identification of the CRAC channel by altered ion
selectivity in a mutant of Orai, Nature 443 (2006) 226–229.
[102] M. Vig, A. Beck, J.M. Billingsley, A. Lis, S. Parvez, C. Peinelt,
D.L. Koomoa, J. Soboloff, D.L. Gill, A. Fleig, J.P. Kinet, R. Pen-
ner, CRACM1 multimers form the ion-selective pore of the CRAC
channel, Curr. Biol. 16 (2006) 2073–2079.
... mammalian TRPC protein, exhibits widespread expression in both neuronal and nonneuronal tissues and is currently the principal candidate component of the SOCE system, contributing significantly to SOCE in various cell types.112 Cumulative evidence underscores the indispensable role of the TRPC1-SOCE pathway in the initiation and progression of neurodegenerative disorders. ...
Article
Full-text available
Background Alzheimer's disease (AD) is a neurodegenerative disorder distinguished by a swift cognitive deterioration accompanied by distinctive pathological hallmarks such as extracellular Aβ (β‐amyloid) peptides, neuronal neurofibrillary tangles (NFTs), sustained neuroinflammation, and synaptic degeneration. The elevated frequency of AD cases and its proclivity to manifest at a younger age present a pressing challenge in the quest for novel therapeutic interventions. Numerous investigations have substantiated the involvement of C/EBPβ in the progression of AD pathology, thus indicating its potential as a therapeutic target for AD treatment. Aims Several studies have demonstrated an elevation in the expression level of C/EBPβ among individuals afflicted with AD. Consequently, this review predominantly delves into the association between C/EBPβ expression and the pathological progression of Alzheimer's disease, elucidating its underlying molecular mechanism, and pointing out the possibility that C/EBPβ can be a new therapeutic target for AD. Methods A systematic literature search was performed across multiple databases, including PubMed, Google Scholar, and so on, utilizing predetermined keywords and MeSH terms, without temporal constraints. The inclusion criteria encompassed diverse study designs, such as experimental, case–control, and cohort studies, restricted to publications in the English language, while conference abstracts and unpublished sources were excluded. Results Overexpression of C/EBPβ exacerbates the pathological features of AD, primarily by promoting neuroinflammation and mediating the transcriptional regulation of key molecular pathways, including δ‐secretase, apolipoprotein E4 (APOE4), acidic leucine‐rich nuclear phosphoprotein‐32A (ANP32A), transient receptor potential channel 1 (TRPC1), and Forkhead BoxO (FOXO). Discussion The correlation between overexpression of C/EBPβ and the pathological development of AD, along with its molecular mechanisms, is evident. Investigating the pathways through which C/EBPβ regulates the development of AD reveals numerous multiple vicious cycle pathways exacerbating the pathological progression of the disease. Furthermore, the exacerbation of pathological progression due to C/EBPβ overexpression and its molecular mechanism is not limited to AD but also extends to other neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD), and multiple sclerosis (MS). Conclusion The overexpression of C/EBPβ accelerates the irreversible progression of AD pathophysiology. Additionally, C/EBPβ plays a crucial role in mediating multiple pathways linked to AD pathology, some of which engender vicious cycles, leading to the establishment of feedback mechanisms. To sum up, targeting C/EBPβ could hold promise as a therapeutic strategy not only for AD but also for other degenerative diseases.
... Apart from STIM-regulated Orai channels that contribute to Ca 2+ release-activated Ca 2+ current (I CRAC )-mediated SOCE, recent studies indicated that STIM-operated TRPC1 activity mediates I SOC (Liu et al., 2003;Ambudkar et al., 2017). Among six known TRPC proteins, TRPC1, TRPC3, and TRPC4 are SOCE partners that are activated by the depletion of Ca 2+ from stores, whereas the operation of TRPC5, TRPC6, and TRPC7 is store-independent (Ambudkar et al., 2007;Liu et al., 2007;Venkatachalam and Montell, 2007;Trebak et al., 2009;Putney and Tomita, 2012). Phospholipase C (PLC)-mediated phosphatidylinositol 4,5-bisphosphate (PI( 4,5 )P 2 ) hydrolysis upon the (S)-3,5-dihydroxyphenylglycine (DHPG)-induced activation of metabotropic glutamate receptor 1/5 (mGluR1/5) may activate TRPCs in a store-independent manner with diacylglycerol (DAG) (Itsuki et al., 2014). ...
Article
Full-text available
Huntington's disease (HD) is a progressive neurodegenerative disorder that is characterized by motor, cognitive, and psychiatric problems. It is caused by a polyglutamine expansion in the huntingtin protein that leads to striatal degeneration via the transcriptional dysregulation of several genes, including genes that are involved in the calcium (Ca²⁺) signalosome. Recent research has shown that one of the major Ca²⁺ signaling pathways, store-operated Ca²⁺ entry (SOCE), is significantly elevated in HD. SOCE refers to Ca²⁺ flow into cells in response to the depletion of endoplasmic reticulum Ca²⁺ stores. The dysregulation of Ca²⁺ homeostasis is postulated to be a cause of HD progression because the SOCE pathway is indirectly and abnormally activated by mutant huntingtin (HTT) in γ-aminobutyric acid (GABA)ergic medium spiny neurons (MSNs) from the striatum in HD models before the first symptoms of the disease appear. The present review summarizes recent studies that revealed a relationship between HD pathology and elevations of SOCE in different models of HD, including YAC128 mice (a transgenic model of HD), cellular HD models, and induced pluripotent stem cell (iPSC)-based GABAergic medium spiny neurons (MSNs) that are obtained from adult HD patient fibroblasts. SOCE in MSNs was shown to be mediated by currents through at least two different channel groups, Ca²⁺ release-activated Ca²⁺ current (ICRAC) and store-operated Ca²⁺ current (ISOC), which are composed of stromal interaction molecule (STIM) proteins and Orai or transient receptor potential channel (TRPC) channels. Their role under physiological and pathological conditions in HD are discussed. The role of Huntingtin-associated protein 1 isoform A in elevations of SOCE in HD MSNs and potential compounds that may stabilize elevations of SOCE in HD are also summarized. Evidence is presented that shows that the dysregulation of molecular components of SOCE or pathways upstream of SOCE in HD MSN neurons is a hallmark of HD, and these changes could lead to HD pathology, making them potential therapeutic targets.
... Two different types of ionic current are evoked by store-depletion: 1) a high selectivity Ca 2+ current mediated by a Ca 2+ release-activated channel (CRAC) called Orai1 [16] and 2) NSCC current [17,18]. Several members of the transient receptor potential canonical (TRPC) channel family have been proposed to act as SOCs (reviewed in [19,20]), although this topic continues to be widely debated [21][22][23]. Previous work from our laboratory has shown that inhibition of acid-sensing ion channel 1a (ASIC1a) diminishes SOCE and associated vasoconstriction in pulmonary VSMC [24]. ...
Article
Full-text available
Although voltage-gated Ca²⁺ channels (VGCC) are a major Ca²⁺ entry pathway in vascular smooth muscle cells (VSMCs), several other Ca²⁺-influx mechanisms exist and play important roles in vasoreactivity. One of these is store-operated Ca²⁺ entry (SOCE), mediated by an interaction between STIM1 and Orai1. Although SOCE is an important mechanism of Ca²⁺ influx in non-excitable cells (cells that lack VGCC); there is debate regarding the contribution of SOCE to regulate VSMC contractility and the molecular components involved. Our previous data suggest acid-sensing ion channel 1a (ASIC1a) is a necessary component of SOCE and vasoconstriction in small pulmonary arteries. However, it is unclear if ASIC1a similarly contributes to SOCE and vascular reactivity in systemic arteries. Considering the established role of Orai1 in mediating SOCE in the systemic circulation, we hypothesize the involvement of ASIC1a in SOCE and resultant vasoconstriction is unique to the pulmonary circulation. To test this hypothesis, we examined the roles of Orai1 and ASIC1a in SOCE- and endothelin-1 (ET-1)-induced vasoconstriction in small pulmonary and mesenteric arteries. We found SOCE is coupled to vasoconstriction in pulmonary arteries but not mesenteric arteries. In pulmonary arteries, inhibition of ASIC1a but not Orai1 attenuated SOCE- and ET-1-induced vasoconstriction. However, neither inhibition of ASIC1a nor Orai1 altered ET-1-induced vasoconstriction in mesenteric arteries. We conclude that SOCE plays an important role in pulmonary, but not mesenteric, vascular reactivity. Furthermore, in contrast to the established role of Orai1 in SOCE in non-excitable cells, the SOCE response in pulmonary VSMCs is largely mediated by ASIC1a.
... Several activation mechanisms have been proposed for TRPC channels, the most frequently described being the activation by G-protein-coupled receptors or by receptor tyrosine kinases via the phospholipase C (PLC)-induced formation of diacylglycerol and inositol 1, 4, 5-trisphosphate [8][9][10]. TRPC1 in particular can also be involved in store-operated calcium entry in cooperation with Orai1 channel and activated by STIM1, a sensor of Ca 2+ contents in the endoplasmic reticulum [11][12][13][14]. It can be directly activated by phosphatidylinositol 3,4,5-trisphosphate, by phosphorylation by the protein kinase C (PKC), or through the hydrolysis of phosphatidylinositol 4,5-bisphosphate and/or the concomitant generation of diacylglycerol [15] or other lipids, such as sphingosine-1-P [16]. ...
Article
Full-text available
Group I metabotropic glutamate receptors (mGluR) are involved in various forms of synaptic plasticity that are believed to underlie declarative memory. We previously showed that mGluR5 specifically activates channels containing TRPC1, an isoform of the canonical family of Transient Receptor Potential channels highly expressed in the CA1-3 regions of the hippocampus. Using a tamoxifen-inducible conditional knockout model, we show here that the acute deletion of the Trpc1 gene alters the extinction of spatial reference memory. mGluR-induced long-term depression, which is partially responsible for memory extinction, was impaired in these mice. Similar results were obtained in vitro and in vivo by inhibiting the channel by its most specific inhibitor, Pico145. Among the numerous known postsynaptic pathways activated by type I mGluR, we observed that the deletion of Trpc1 impaired the activation of ERK1/2 and the subsequent expression of Arc, an immediate early gene that plays a key role in AMPA receptors endocytosis and subsequent long-term depression.
Article
Oxidative stress is a predisposing factor in Chronic Obstructive Pulmonary Disease (COPD). Specifically, pulmonary epithelial (PE) cells reduce antioxidant capacity during COPD because of the continuous production of reactive oxygen species (ROS). However, the molecular pathogenesis that governs such ROS activity is unclear. Here we show that the dysregulation of intracellular calcium concentration ([Ca2+]i) in PE cells from COPD patients, compared to the healthy PE cells, is associated with the robust functional expressions of Transient Receptor Potential Canonical (TRPC)1 and TRPC3 channels, and Ca2+ entry (SOCE) components, Stromal Interaction Molecule 1 (STIM1) and ORAI1 channels. Additionally, the elevated expression levels of fibrotic, inflammatory, oxidative, and apoptotic markers in cells from COPD patients suggest detrimental pathway activation, thereby reducing the ability of lung remodeling. To further delineate the mechanism, we used human lung epithelial cell line, A549, since the behavior of SOCE and the expression patterns of TRPC1/C3, STIM1, and ORAI1 were much alike PE cells. Notably, the knockdown of TRPC1/C3 in A549 cells substantially reduced the SOCE-induced [Ca2+]i rise, and reversed the ROS-mediated oxidative, fibrotic, inflammatory, and apoptotic responses, thus confirming the role of TRPC1/C3 in SOCE driven COPD-like condition. Higher TRPC1/C3, STIM1, and ORAI1 expressions, along with a greater Ca2+ entry, via SOCE in ROS-induced A549 cells, led to the rise in oxidative, fibrotic, inflammatory, and apoptotic gene expression, specifically through the extracellular signal-regulated kinase (ERK) pathway. Abatement of TRPC1 and/or TRPC3 reduced the mobilization of [Ca2+]i and reversed apoptotic gene expression and ERK activation, signifying the involvement of TRPC1/C3. Together these data suggest that TRPC1/C3 and SOCE facilitate the COPD condition through ROS-mediated cell death, thus implicating their likely roles as potential therapeutic targets for COPD. SUMMARY: Alterations in Ca2+ signaling modalities in normal pulmonary epithelial cells exhibit COPD through oxidative stress and cellular injury, compromising repair, which was alleviated through inhibition of store-operated calcium entry. SUBJECT AREA: Calcium, ROS, Cellular signaling, lung disease.
Article
Full-text available
The neural cell adhesion molecule (NCAM) plays important functional roles in the developing and mature nervous systems. Here, we show that the transient receptor potential canonical (TRPC) ion channels TRPC1, −4, and −5 not only interact with the intracellular domains of the transmembrane isoforms NCAM140 and NCAM180, but also with the glycan polysialic acid (PSA) covalently attached to the NCAM protein backbone. NCAM antibody treatment leads to the opening of TRPC1, −4, and −5 hetero- or homomers at the plasma membrane and to the influx of Ca2+ into cultured cortical neurons and CHO cells expressing NCAM, PSA, and TRPC1 and −4 or TRPC1 and −5. NCAM-stimulated Ca2+ entry was blocked by the TRPC inhibitor Pico145 or the bacterial PSA homolog colominic acid. NCAM-stimulated Ca2+ influx was detectable neither in NCAM-deficient cortical neurons nor in TRPC1/4- or TRPC1/5-expressing CHO cells that express NCAM, but not PSA. NCAM-induced neurite outgrowth was reduced by TRPC inhibitors and a function-blocking TRPC1 antibody. A characteristic signaling feature was that extracellular signal-regulated kinase 1/2 phosphorylation was also reduced by TRPC inhibitors. Our findings indicate that the interaction of NCAM with TRPC1, −4, and −5 contributes to the NCAM-stimulated and PSA-dependent Ca2+ entry into neurons thereby influencing essential neural functions.
Article
Full-text available
Pre-mRNA processing factor 4B (PRP4) has previously been shown to induce epithelial-mesenchymal transition (EMT) and drug resistance in cancer cell lines. As melanin plays an important photoprotective role in the risk of sun-induced skin cancers, we have investigated whether PRP4 can induce drug resistance and regulate melanin biosynthesis in a murine melanoma (B16F10) cell line. Cells were incubated with a crucial melanogenesis stimulator, alpha-melanocyte-stimulating hormone, followed by transfection with PRP4. This resulted in the inhibition of the production of melanin via the downregulation of adenylyl cyclase-cyclic adenosine 3′,5′-monophosphate (AC)–(cAMP)–tyrosinase synthesis signaling pathway. Inhibition of melanin production by PRP4 leads to the promotion of carcinogenesis and induced drug resistance in B16F10 cells. Additionally, PRP4 overexpression upregulated the expression of β-arrestin 1 and desensitized the extracellular calcium-sensing receptor (CaSR), which in turn, inhibited the influx of extracellular Ca2+ ions. The decreased influx of Ca2+ was confirmed by a decreased expression level of calmodulin. We have demonstrated that transient receptor potential cation channel subfamily C member 1 was involved in the influx of CaSR-induced Ca2+ via a decreasing level of its expression. Furthermore, PRP4 overexpression downregulated the expression of AC, decreased the synthesis of cAMP, and modulated the actin cytoskeleton by inhibiting the expression of Ras homolog family member A (RhoA). Our investigation suggests that PRP4 inhibits the production of melanin in B16F10 cells, blocks the influx of Ca2+ through desensitization of CaSR, and modulates the actin cytoskeleton through downregulating the AC–cAMP pathway; taken together, these observations collectively lead to the promotion of skin carcinogenesis.
Article
Full-text available
Sustained imbalance in intracellular calcium (Ca2+) entry and clearance alters cellular integrity, ultimately leading to cellular homeostasis disequilibrium and cell death. Alzheimer’s disease (AD) is the most common cause of dementia. Beside the major pathological features associated with AD-linked toxic amyloid beta (Aβ) and hyperphosphorylated tau (p-tau), several studies suggested the contribution of altered Ca2+ handling in AD development. These studies documented physical or functional interactions of Aβ with several Ca2+ handling proteins located either at the plasma membrane or in intracellular organelles including the endoplasmic reticulum (ER), considered the major intracellular Ca2+ pool. In this review, we describe the cellular components of ER Ca2+ dysregulations likely responsible for AD. These include alterations of the inositol 1,4,5-trisphosphate receptors’ (IP3Rs) and ryanodine receptors’ (RyRs) expression and function, dysfunction of the sarco-endoplasmic reticulum Ca2+ ATPase (SERCA) activity and upregulation of its truncated isoform (S1T), as well as presenilin (PS1, PS2)-mediated ER Ca2+ leak/ER Ca2+ release potentiation. Finally, we highlight the functional consequences of alterations of these ER Ca2+ components in AD pathology and unravel the potential benefit of targeting ER Ca2+ homeostasis as a tool to alleviate AD pathogenesis.
Article
Full-text available
Transient receptor potential canonical (TRPC) proteins constitute a group of receptor-operated calcium-permeable nonselective cationic membrane channels of the TRP superfamily. They are largely expressed in the hippocampus and are able to modulate neuronal functions. Accordingly, they have been involved in different hippocampal functions such as learning processes and different types of memories, as well as hippocampal dysfunctions such as seizures. This review covers the mechanisms of activation of these channels, how these channels can modulate neuronal excitability, in particular the after-burst hyperpolarization, and in the persistent activity, how they control synaptic plasticity including pre- and postsynaptic processes and how they can interfere with cell survival and neurogenesis.
Article
Calcium influx in nonexcitable cells regulates such diverse processes as exocytosis, contraction, enzyme control, gene regulation, cell proliferation, and apoptosis. The dominant Ca2+ entry pathway in these cells is the store-operated one, in which Ca2+ entry is governed by the Ca2+ content of the agonist-sensitive intracellular Ca2+ stores. Only recently has a Ca2+ current been described that is activated by store depletion. The properties of this new current, called Ca2+ release-activated Ca2+ current (ICRAC), have been investigated in detail using the patch-clamp technique. Despite intense research, the nature of the signal that couples Ca2+ store content to the Ca2+ channels in the plasma membrane has remained elusive. Although ICRAC appears to be the most effective and widespread influx pathway, other store-operated currents have also been observed. Although the Ca2+ release-activated Ca2+ channel has not yet been cloned, evidence continues to accumulate that the Drosophila trp gene might encode a store-operated Ca2+ channel. In this review, we describe the historical development of the field of Ca2+ signaling and the discovery of store-operated Ca2+ currents. We focus on the electrophysiological properties of the prototype store-operated current ICRAC, discuss the regulatory mechanisms that control it, and finally consider recent advances toward the identification of molecular mechanisms involved in this ubiquitous and important Ca2+ entry pathway.
Article
Endothelial cells (EC) form a unique signal-transducing surface in the vascular system. The abundance of ion channels in the plasma membrane of these nonexcitable cells has raised questions about their functional role. This review presents evidence for the involvement of ion channels in endothelial cell functions controlled by intracellular Ca ²⁺ signals, such as the production and release of many vasoactive factors, e.g., nitric oxide and PGI 2 . In addition, ion channels may be involved in the regulation of the traffic of macromolecules by endocytosis, transcytosis, the biosynthetic-secretory pathway, and exocytosis, e.g., tissue factor pathway inhibitor, von Willebrand factor, and tissue plasminogen activator. Ion channels are also involved in controlling intercellular permeability, EC proliferation, and angiogenesis. These functions are supported or triggered via ion channels, which either provide Ca ²⁺ -entry pathways or stabilize the driving force for Ca ²⁺ influx through these pathways. These Ca ²⁺ -entry pathways comprise agonist-activated nonselective Ca ²⁺ -permeable cation channels, cyclic nucleotide-activated nonselective cation channels, and store-operated Ca ²⁺ channels or capacitative Ca ²⁺ entry. At least some of these channels appear to be expressed by genes of the trp family. The driving force for Ca ²⁺ entry is mainly controlled by large-conductance Ca ²⁺ -dependent BK Ca channels ( slo), inwardly rectifying K ⁺ channels (Kir2.1), and at least two types of Cl ⁻ channels, i.e., the Ca ²⁺ -activated Cl ⁻ channel and the housekeeping, volume-regulated anion channel (VRAC). In addition to their essential function in Ca ²⁺ signaling, VRAC channels are multifunctional, operate as a transport pathway for amino acids and organic osmolytes, and are possibly involved in endothelial cell proliferation and angiogenesis. Finally, we have also highlighted the role of ion channels as mechanosensors in EC. Plasmalemmal ion channels may signal rapid changes in hemodynamic forces, such as shear stress and biaxial tensile stress, but also changes in cell shape and cell volume to the cytoskeleton and the intracellular machinery for metabolite traffic and gene expression.
Conference Paper
The TRPC (transient receptor potential canonical) proteins are activated in response to agonist-stimulated PIP2 (phosphatidylinositol 4,5-bisphosphate) hydrolysis and have been suggested as candidate components of the elusive SOC (store-operated calcium channel). TRFC1 is currently the strongest candidate component of SOC. Enclogenous TRPC1 has been shown to contribute to SOCE (store-operated calcium entry) in several different cell types. However, the mechanisms involved in the regulation of TRFC1 and its exact physiological function have yet to be established. Studies from our laboratory and several others have demonstrated that TRPC1 is assembled in a signalling complex with key calcium signalling proteins in functionally specific plasma membrane microdomains. Furthermore, critical interactions between TRPC1 monomers as well as interactions between TRPC1 and other proteins determine the surface expression and function of TRFC1-containing channels. Recent studies have revealed novel regulators of TRFC1 -containing SOCs and have demonstrated a common molecular basis for the regulation of CRAC (calcium-release-activated calcium) and SOC channels. In the present paper, we will revisit the role of TRPC1 in SOCE and discuss how studies with TRPC1 provide an experimental basis for validating the mechanism of SOCE.
Article
This study examined the involvement of store-operated Ca 2+ entry in agonist-stimulation of salivary gland fluid secretion. A recombinant adenovirus (AdCMV-hTrp1) encoding the storeoperated Ca 2+ channel protein, human transient receptor potential 1 (hTrp1), was used to direct expression of HA (hemaglutinin )-tagged hTrp1 in vivo in rat submandibular glands (SMG) and in vitro in the human submandibular gland cell line (HSG). Studies with HSG cells demonstrated that AdCMV-hTrp1 was successful in directing the expression of functional hTrp1 and that it did not affect early Ca 2+ signaling events. AdCMV-hTrp1-infected SMG displayed an increase in the level of Trp1 and a fivefold increase in pilocarpine-stimulated fluid secretion, compared with glands infected with a control adenovirus encoding luciferase (AdCMV-Luc). The expressed hTrp1 demonstrated polarized localization in the basolateral plasma membrane region of SMG acinar cells and was co-immunoprecipitated with IP 3Rs. Further, acinar cells isolated from AdCMV-hTrp1-infected glands demonstrated a significant increase in carbachol- and Tgstimulated Ca 2+ entry compared with cells isolated from AdCMV-Luc-infected glands. We conclude that in vivo expression of Trp1 in SMG induces an enhancement of agonist-stimulated fluid secretion via increasing store-operated Ca 2+ entry into acinar cells. These data suggest that store-operated Ca 2+ entry has a role in agonist-stimulated fluid secretion from salivary glands.
Article
Capacitative Ca2+ entry (CCE) activated by release/depletion of Ca2+ from internal stores represents a major Ca2+ influx mechanism in lymphocytes and other nonexcitable cells. Despite the importance of CCE in antigen-mediated lymphocyte activation, molecular components constituting this mechanism remain elusive. Here we demonstrate that genetic disruption of transient receptor potential (TRP)1 significantly attenuates both Ca2+ release-activated Ca2+ currents and inositol 1,4,5-trisphosphate (IP3)-mediated Ca2+ release from endoplasmic reticulum (ER) in DT40 B cells. As a consequence, B cell antigen receptor–mediated Ca2+ oscillations and NF-AT activation are reduced in TRP1-deficient cells. Thus, our results suggest that CCE channels, whose formation involves TRP1 as an important component, modulate IP3 receptor function, thereby enhancing functional coupling between the ER and plasma membrane in transduction of intracellular Ca2+ signaling in B lymphocytes.
Article
Members of the Trp protein family have been suggested as the structural basis of store-operated cation conductances. With this study, we provide evidence for the expression of three isoforms of Trp (hTrp1, 3 and 4) in human umbilical vein endothelial cells (HUVEC). The role of Trp proteins in store regulation of endothelial membrane conductances was tested by expression of an N-terminal fragment of hTrp3 (N-TRP) which exerts a dominant negative effect on Trp channel function presumably due to suppression of channel assembly. Depletion of intracellular Ca2+ stores with IP3 (100 microM) or thapsigargin (100 nM) induced a substantial cation conductance in sham-transfected HUVEC as well as in HUVEC transfected with hTrp3. In contrast, HUVEC transfected with N-TRP failed to exhibit store-operated currents. Our results suggest the involvement of Trp related proteins in the store-operated cation conductance of human vascular endothelial cells.