Abstract
In many cell types, the rise in cytosolic Ca2+ due to opening of Ca2+ release-activated Ca2+ (CRAC) channels drives a plethora of responses, including secretion, motility, energy production, and gene expression. The amplitude and time course of the cytosolic Ca2+ rise is shaped by the rates of Ca2+ entry into and removal from the cytosol. However, an extended bulk Ca2+ rise is toxic to cells. Here, we show that the plasma membrane Ca2+ ATPase (PMCA) pump plays a major role in preventing a prolonged cytosolic Ca2+ signal following CRAC channel activation. Ca2+ entry through CRAC channels leads to a sustained sub-plasmalemmal Ca2+ rise but bulk Ca2+ is kept low by the activity of PMCA4b. Despite the low cytosolic Ca2+, membrane permeability to Ca2+ is still elevated and Ca2+ continues to enter through CRAC channels. Ca2+-dependent NFAT activation, driven by Ca2+ nanodomains near the open channels, is maintained despite the return of bulk Ca2+ to near pre-stimulation levels. Our data reveal a central role for PMCA4b in determining the pattern of a functional Ca2+ signal and in sharpening local Ca2+ gradients near CRAC channels, whilst protecting cells from a toxic Ca2+ overload.
Keywords: Ca2+, plasma membrane ATPase, calcium channel, Transcription factor
Graphical Abstract
Graphical Abstract.

Introduction
A rise in cytosolic Ca2+ is a universal trigger for activation of a remarkable breadth of cellular responses, including neurotransmitter release, contraction, energy production, gene transcription, and cell death.1 The dynamics of a cytosolic Ca2+ signal is determined by the balance between Ca2+ entry into the cytosol, typically accomplished through opening Ca2+-permeable ion channels that populate organelles and the plasma membrane, mobile and immobile Ca2+ buffers, and Ca2+ removal mechanisms that sequestrate Ca2+ into internal Ca2+ stores or eject the ion from the cell.2
In many cell types, particularly non-excitable cells, Ca2+ release-activated Ca2+ (CRAC) channels play a central and often indispensable role in providing the Ca2+ rise that drives important biological outputs.3,4 Stimulation of cell-surface receptors that couple either to Gq-type heterotrimeric G proteins or tyrosine kinases activate isoforms of phospholipase C to produce the second messenger inositol-1, 4, 5-trisphosphate (InsP3).5 InsP3 binds to and opens InsP3-gated Ca2+ channels in the endoplasmic reticulum (ER).6 Ca2+ depletion from the ER activates two proteins in the ER membrane, stromal interaction molecule 1 (STIM1) and STIM2.7,8 Upon Ca2+ store emptying, the proteins form oligomers and migrate towards the plasma membrane, where they bind to, cluster and gate open plasmalemmal Orai proteins, the pore-forming subunits of CRAC channels.9–11 Ca2+ entry through the channels drives a plethora of spatially and temporally distinct responses, including secretion, ER store refilling, and recruitment of Ca2+-regulated transcription factors such as c-fos, the nuclear factor of activated T cells (NFAT), nuclear factor kappa-light-chain enhancer of activated B cells (NF-κB) and cAMP response element-binding protein (CREB).12,13
Ca2+ entry through CRAC channels is buffered by mitochondria, taken into the ER by SERCA pumps or ejected from the cell by plasma membrane Ca2+ ATPases (PMCA) and, to a lesser extent in non-excitable cells, by the electrogenic Na+–Ca2+ exchanger. Both SERCA pumps and mitochondrial Ca2+ buffering have important and well-characterized consequences on the duration of CRAC channel activity. By refilling stores, SERCA pumps deactivate CRAC channels and thereby terminate Ca2+ entry.14–16 Mitochondria prolong CRAC channel activity because Ca2+ uptake by the mitochondrial Ca2+ uniporter prevents bulk cytosolic Ca2+ from increasing sufficiently or long enough to evoke Ca2+-dependent slow inactivation of the channels.17–21 By contrast, the role of PMCA in shaping Ca2+ signals generated by CRAC channels is less well understood. Four genes encode mammalian PMCAs (PMCA1–4) and alternative splicing generates more than twenty variants.22 The pumps are all activated by Ca2+-calmodulin but differ in their rates of activation and in their affinities for calmodulin.23
Pászty and colleagues have examined the impact of overexpression of various PMCAs on the time course of the cytosolic Ca2+ signal following CRAC channel activation in HeLa cells.24 They demonstrated that different PMCAs dramatically altered the pattern and time course of the Ca2+ signal in a manner dependent on how quickly the PMCA was activated by Ca2+-calmodulin. In the presence of the fast PMCa2b isoform, cytosolic Ca2+ rapidly returned to resting levels whereas slow, oscillatory signals were seen when the slower PMCA4b protein was present instead. These results clearly establish the impact of PMCAs on Ca2+ signals evoked by store-operated Ca2+ entry.
In Jurkat T lymphocytes, local Ca2+ entry through CRAC channels stimulates PMCA activity in two distinct phases: an initial rapid increase is followed by a form of modulation that develops over tens of seconds and enhances pump activity ∼4-fold.25 This slow increase in PMCA activity after store depletion gradually reduces cytosolic Ca2+ and ensures a stable cytosolic Ca2+ elevation is reached. Findings that are contradictory to this have been recently reported, also extracted from Jurkat T cells.26 In this latter study, STIM1 was found to inhibit PMCA, substantially delaying recovery of cytosolic Ca2+ by several tens of seconds following Ca2+ entry through CRAC channels. By prolonging the cytosolic Ca2+ rise, inhibition of Ca2+ clearance was proposed to be essential for Ca2+-dependent NFAT activation.26 A further twist has recently been provided by the same authors who found that STIM1 inhibition was prevented by the protein partner of STIM1 (POST).27 In the presence of overexpressed POST, PMCA activity was not blocked by STIM1 in Jurkat T cells. Cytosolic Ca2+ now rapidly recovered and it was suggested that the rapid decline of cytosolic Ca2+ was essential for NFAT activity, ostensibly by preventing Ca2+-dependent inactivation of CRAC channels.27
In this study, we have investigated the role of PMCA in shaping cytosolic Ca2+ signals generated by CRAC channels. Extending the findings of Bautista and Lewis,28 we find that modulation of the PMCA4b isoform leads to a remarkable increase in PMCA activity such that the pump more than matches Ca2+ entry through CRAC channels, returning cytosolic Ca2+ to pre-stimulation levels despite maintained channel activity. This increase in PMCA activity prevents toxic consequences of a sustained a larger cytosolic Ca2+ rise. By using a Ca2+ sensor tagged directly to Orai1, we reveal that local Ca2+ near the channels remains high despite bulk cytosolic Ca2+ declining rapidly. Finally, we demonstrate that gene expression continues unabated despite the loss of the bulk Ca2+ rise. Our results show that enhanced PMCA activity sharpens spatial Ca2+ gradients emanating from CRAC channels, increasing the signalling power of local Ca2+ signals. Our data reinforce the importance of Ca2+ nanodomains near CRAC and not a bulk Ca2+ rise in driving excitation–transcription coupling.
Materials and Methods
Cell Culture
HEK293 were purchased from ATCC (via the United Kingdom supplier LGC) and were cultured in Dulbecco's modified Eagle's medium (DMEM) (Thermo Scientific). Media were supplemented with 10% fetal bovine serum and 1% penicillin-streptomycin.
Plasmid Constructs and Transfection
Plasmids: PMCA4b-cherry was a kind gift from Dr Agnes Enyedi. Orai1-GECO and cytosolic GECO were purchased from Addgene. NFAT1-cherry was provided by Dr Irene Frischauf (Linz University, Austria). STIM1-YFP, Orai1-GFP, and Orai1-CFP were provided by Dr James Putney (NIEHS, USA). HEK293 cells were transfected with Lipofectamine 2000 (Invitrogen) using 1 μg plasmid, and then incubated in media without penicillin-streptomycin. In the GECO experiments, only 0.6 μg plasmid was used to reduce the levels of overexpression. Experiments were then carried out 24 to 48 h after transfection.
Cytosolic Ca2+ Measurements
Cytosolic Ca2+ was measured as previously described.29 Cells were loaded with 1 μm Fura 2-am for 40 min in the dark at room temperature, washed three times and then kept in the dark for a further 15 min to allow for dye de-esterification prior to recording. Fura-2 fluorescence was measured by alternately exciting the dye at 340 and 380 nm, and emission was collected at 510 nm. Changes in Ca2+ concentration are presented as the ratio = F340/F380. Some experiments were carried in Oxford using an Olympus BX51 × 1 upright microscope with the commercial xcellence rt software. Data shown for Figures 1, 2C and D, 5, and 6A were obtained in the NIEHS using a Nikon TS-100 inverted microscope equipped with a 20 × fluor objective (0.75 NA). Fluorescence images of the cells were recorded and analyzed with a digital fluorescence imaging system (InCyt Im2, Intracellular Imaging Inc., Cincinnati, OH, USA) equipped with a light-sensitive CCD camera (Cooke PixelFly, ASI, Eugene, OR, USA).
Figure 1.
Bulk cytosolic Ca2+ recovers despite prolonged exposure to store depletion. (A) Time courses of cytosolic Ca2+ are compared for cells challenged with thapsigarin in the absence of external Ca2+, presence of external Ca2+, and following pretreatment with the channel blocker BTP2 in the presence of external Ca2+. Each trace is the mean ± SEM of between 29 and 30 cells. In the thapsigargin/0Ca2+ experiment, 1 μm ionomycin was applied at 26 min. (B) Store-operated Ca2+ entry evoked by thapsigargin does not refill the store, as gauged by the loss of the ionomycin response in Ca2+-free solution. Data are mean ± SEM of between 26 and 28 cells.
Figure 2.
CRAC channels remain active during sustained stimulation with thapsigargin. (A) FRET signal between STIM1-YFP and Orai1-CFP was sustained (black trace) despite cytosolic Ca2+ recovering to resting levels (green trace). (B) Aggregate data from 4 independent experiments as in panel A are compared. P < 0.01 between the two groups. (C) Cytosolic Ca2+ falls rapidly when cells are perfused in Ca2+-free solution as shown. Data are mean ± SEM of 29 cells. (D) Raising external Ca2+ to 10 m m increases cytosolic Ca2+ in cells exposed to thapsigargin despite cytosolic Ca2+ having returned close to resting levels (blue trace). Elevating external Ca2+ in cells with replete stores did not increase cytosolic Ca2+ (red trace). Data are mean ± SEM of 27 cells (blue trace) and 27 cells (red trace).
Figure 5.
NFAT migration continues despite cytosolic Ca2+ being low. (A) Cytosolic Ca2+ and NFAT1-cherry dynamics were measured in the same cell. Images below the graph show NFAT distribution at the times indicated (t = 0, 6, and 35 min). Thapsigargin was applied at the arrow. N denotes nucleus. (B) Graph compares cytosolic Ca2+ and nuclear/cytosolic ratio of NFAT at 20 and 35 min. Both parameters have been normalized to their respective maximal responses. P < 0.05 between points for each condition.
Figure 6.
PMCA pump is the major Ca2+ clearance mechanism. (A) A small Ca2+ rise is evoked by FCCP both in control cells and after 15 min of stimulation with thapsigargin. (B) Block of the PMCA with external La3+ enhances the Ca2+ response to thapsigargin and delays its recovery. Data are mean of >20 cells for each condition. (C) The Ca2+ response is increased and sustained in alkaline pH, which inhibits the PMCA pump. Each trace is mean of >16 cells for each condition. (D) Removal of external Na+ has little impact on the recovery of cytosolic Ca2+ to thapsigargin. (E) Removal of external Na+ does not affect the half time for recovery of cytosolic Ca2+. P > 0.2 between the groups.
TIRF Measurements
Fluorescence images were captured on an Andor Dragonfly 505 multi modal confocal system (Oxford Instruments, Abingdon, UK) in Total Internal Refection Fluorescence (TIRF) mode using a Nikon 60X Apo TIRF 60X/1.49 objective lens. The 488 nm laser line was set to 5%, TIRF mode to penetration with a depth of 100 nm while fluorescence emission was collected with a 100 ms exposure of an Andor Zyla camera. A total of 200 time series images were acquired every 10 s during each experiment. The time series images were analyzed in FIJI (ImageJ 1.53q) where individual cells were cropped and then mean fluorescence intensity was measured above a threshold of 200 for all images.
Airyscan Confocal Microscopy
Airyscan confocal images were taken on a Zeiss LSM880 with Airyscan (Carl Zeiss Inc, Oberkochen, Germany) using a C-Apochromat 40X/1.2 Water DIC objective. For the red channel, a 561 nm DPSS laser at 2.0% power was used for excitation of mCherry while a long pass 570 nm filter collected the emission. For the green channel, a 488 nm ArKr laser line at 5.0% power was used for excitation of GFP while a bandpass 495–550 filter was used for collection of the emission signal. The master gain setting of the airyscan detector was held constant for all images of both channels with a setting of 800. Furthermore, all images were taken with a zoom of 3, a 1.47μs pixel dwell time, a 0.05 μm pixel size, and a physical pinhole setting of 124 μm. The imaging experiment was performed as a time series of multiple positions where a total 15 images were acquired every 90 s at each position.
Individual cells were analyzed across the time series from each position. Time lapse images of each cell were imported into Imaris 9.9 (Oxford Instruments plc, Abington, UK) licensed with the Coloc feature. Co-localization thresholds for each channel were manually set and held constant across the entire time series. The number of co-localized voxels for each time point were then exported and subsequently normalized to time 0 where a mean and SE were calculated across all replicates.
FRET Measurements
FRET measurements between STIM1-YFP and Orai1-CFP were performed on an Olympus BX51 × 1 upright microscope equipped with 40× water immersion objective and OBS megaview CCD camera. DV2 beam splitter was used to split donor emission (CFP-480/30) and acceptor emission (YFP-535/40) channels using dichroic mirror at 505 wavelength. The donor and acceptor channels were projected on separate halves of the camera and two halves were aligned using pixel by pixel alignment using the patterned slide provided by Olympus and alignment was also reconfirmed using ImageJ. Analysis of ratiometric FRET (FRET = intensity in YFP channel/intensity in CFP channel) and FRET kinetics was calculated after subtracting background intensity using the FRET module provided with the commercial xcellence rt software by Olympus. Images were recorded with 100 ms of exposure using a cell R excitation system fitted with CFP excitation filter at 0.1 Hertz frequency with no binning.
NFAT Translocation and Cytosolic Ca2+ Measurements
Cytosolic Ca2+ and NFAT translocation were measured at the same time by loading cells expressing NFAT1-cherry with Fluo 4-am (1 μm for 30 min). Images were taken with a Zeiss LSM880 (Carl Zeiss Inc, Oberkochen, Germany) using the 488 and 594 nm laser lines for excitation paired with emission filter settings of 499–588 nm, and 590–735 nm, respectively. A C-Apochromat 40x/1.2 water objective was used for image collection.
Statistical Analysis
All results were expressed as means ± SD unless indicated. Two-tailed Student's t-test was used to compare differences between two groups in all the experiments, using GraphPad Prism and statistical significance was set at a P-value of < 0.05.
Results
Sustained Activation of CRAC Channels Leads to a Transient Rise in Cytosolic Ca2+
Ca2+ store depletion following exposure to the SERCA pump blocker thapsigargin (2 μm) in the absence of external Ca2+ evoked a transient rise in cytosolic Ca2+, which then decayed monotonically to pre-stimulation levels after ∼8 min (Figure 1A, black trace). Because store depletion leads to opening of CRAC channels, we expected a prolonged increase in cytosolic Ca2+ following stimulation with thapsigargin in the presence of external Ca2+. However, this was not the case; the Ca2+ signal was not sustained despite continuous exposure of thapsigargin (Figure 1A, red trace). In ∼70% of cells, cytosolic Ca2+ decayed gradually but continuously and returned to pre-stimulation levels only ∼2.5-fold more slowly than was the case in the absence of external Ca2+ [(Figure 1A), see also Figures 2A, 6B, and 7E]. In the remaining cells, cytosolic Ca2+ fell by ∼75% from the peak value within 10–15 min. Cells from a given preparation tended to fit into one or other of these profiles. CRAC channels were functional in the presence of external Ca2+ because stimulation with thapsigargin in the presence of the channel blocker BTP2 resulted in a Ca2+ signal that was indistinguishable from that evoked by thapsigargin in Ca2+-free solution (Figure 1A, blue trace).
Figure 7.
Overexpression of PMCA4b accelerates Ca2+ removal following store-operated Ca2+ entry. (A) The timecourse of cytosolic Ca2+ is compared for a control cell (mock transfected and labeled WT for wild type) and one expressing PMCA4b-cherry. (B) The box in panel A is shown on an expanded time scale. (C) The rate of Ca2+ entry from 22 cells is compared for the conditions shown. P > 0.3 between the two groups. (D) Aggregate data measuring the decay rate of the Ca2+ signal from 22 cells are compared for the conditions shown. Decay rate was measured as a linear fit to the initial steep decline of cytosolic Ca2+ when external Ca2+ was removed. P < 0.01 between the two groups. (E) Ca2+ signals are compared between wild type cells and cells expressing PMCA4b-cherry. Red trace is the mean of 22 cells and green trace of 26 cells. (F) Ca2+ signals to ionomycin challenge in Ca2+-free solution are compared for wild type cells and for cells expressing PMCA4b-cherry. Each trace is the mean of 16–21 cells. (G) Aggregate data measuring the area under the traces in panel E are compared. P < 0.05 between the two groups.
We considered various explanations for the complete return of cytosolic Ca2+ to resting levels despite sustained activation of CRAC channels. It is possible that stores refilled, leading to deactivation of the channels. However, this is unlikely for three reasons. First, we used a supra-maximal concentration of thapsigargin, which irreversibly inhibits SERCA pumps over the time course of our experiments. Second, we measured store Ca2+ content following application of the Ca2+ ionopore ionomycin in Ca2+-free solution. Control cells not exposed to thapsigargin showed a robust rise in cytosolic Ca2+ following challenge with ionomycin, demonstrating stores were replete with Ca2+ under resting conditions (red trace in Figure 1B). Exposure to thapsigargin in Ca2+-free solution released Ca2+ from stores and subsequent stimulation with ionomycin evoked a much reduced response [Figure 1A, black trace; ionomycin was applied at 1800 s; compare response here with the ionomycin response (red trace) in Figure 1B]. Hence both thapsigargin and ionomycin target the same Ca2+ store. Importantly, the ionomycin response was almost abolished in cells pretreated with thapsigargin in the presence of external Ca2+, revealing little if any store refilling had taken place despite prolonged CRAC channel activity (Figure 1B, blue trace). Third, interaction between STIM1 and Orai1 was sustained in thapsigargin-stimulated cells in the presence of 2 m m external Ca2+ for up to 20 min, a time when cytosolic Ca2+ had returned to, or close to, pre-stimulation levels (see below).
CRAC Channels Remain Active Despite the Recovery of Cytosolic Ca2+
Another explanation for why the Ca2+ signal is transient despite sustained store depletion is that cytosolic Ca2+ inactivates the channels, uncoupling STIM1 from Orai1. CRAC channels exhibit both fast and slow Ca2+-dependent inactivation.4,30,31 Fast inactivation develops over milliseconds, is controlled by Ca2+ binding to sites located within 3 and 4 nm of the pore and, over the physiological range of voltages encountered by a cell, accounts for up to ∼40% inhibition of CRAC channels.32–34 Slow inactivation develops over tens of seconds and requires a rise in bulk cytosolic Ca2+ to the low microMolar range.14,35,36 Ca2+-dependent inactivation of CRAC channels results in dissociation of STIM1 away from the plasma membrane, so the protein is no longer able to bind to and gate Orai1 channels.37 Several lines of evidence suggest Ca2+-dependent inactivation of CRAC channels does not account for the decline in the cytosolic Ca2+ signal despite sustained store depletion (Figure 1A). First, recoveries from fast and slow inactivation are complete within ∼500 ms and ∼100 s, respectively.14,33,34 Therefore, as cytosolic Ca2+ falls over tens of seconds, recovery from inactivation should occur and lead to slow oscillatory Ca2+ signals.38 This was not observed; cytosolic Ca2+ fell continuously with time and did not oscillate (Figure 1A). Second, we measured FRET between STIM1-YFP and Orai1-CFP following stimulation with thapsigargin in the presence of external Ca2+ over several minutes. The FRET signal peaked after ∼4 min and remained strong despite cytosolic Ca2+ signal returning to pre-stimulation levels (Figure 2A). The fractional decrease in the FRET signal at 15 min was 19± 4% even though cytosolic Ca2+ had declined close to basal levels (Figure 2B). The sustained FRET signal in Figure 2A and B provide additional support for the absence of store refilling under these conditions; store refilling would lead to disassociation of STIM1 and Orai1 with a subsequent loss of FRET between the two proteins. A third argument against Ca2+-dependent inactivation is shown in Figure 2C and D. If the decline in cytosolic Ca2+ in Figure 1A is due to loss of CRAC channels as a consequence of strong Ca2+-dependent inactivation, then membrane permeability to Ca2+ should be low as cytosolic Ca2+ declines in the continuous presence of thapsigargin. To test this, we carried out two different experiments. First, removed external Ca2+ once cytosolic Ca2+ had decayed close to pre-stimulation levels. If CRAC channels had inactivated, then cytosolic Ca2+ should be unaltered. However, cytosolic Ca2+ fell quickly, consistent with channel activity (Figure 2C). Second, we raised external Ca2+ to 10 m m, once cytosolic Ca2+ was close to resting levels. A clear increase in cytosolic Ca2+ occurred (Figure 2D). Raising external Ca2+ to 10 m m in cells not exposed to thapsigargin failed to increase cytosolic Ca2+ (Figure 2D, red trace). Therefore, CRAC channels remain open in the continuous presence of thapsigargin, despite bulk cytosolic Ca2+ returning close to pre-stimulation levels.
Measurements of Local Ca2+ Near Orai1 Channels
If CRAC channels are indeed open despite bulk cytosolic Ca2+ being low, then local Ca2+ near CRAC channels should remain elevated even when the bulk cytosolic Ca2+ has fallen substantially. To address this directly, we expressed an Orai1 construct in which the Ca2+-sensitive fluorescent protein GECO had been tagged to the channel N-terminus.39 The GECO moiety faithfully reports Ca2+ close to CRAC channels and has been used to measure single Orai1 channel activity.39 Stimulation with thapsigargin in Ca2+-free solution failed to evoke a detectable Ca2+ rise near Orai1 channels (Figure 3A), reflecting at least in part the relatively low affinity of GECO for Ca2+ compared with fura 2 (∼600 versus 200 n m, respectively) and the low levels of released Ca2+ that reach the plasma membrane. Readmission of external Ca2+ resulted in a rapid rise in the Orai1-GECO signal and this decayed slowly over several minutes, falling by only ∼25% after 15 min of Ca2+ entry (Figure 3A, green trace). We also measured bulk Ca2+ by expressing GECO targeted to the cytosol. Unlike the case with Orai1-GECO, cytosolic GECO did respond to Ca2+ release from the stores (Figure 3A, yellow trace). Readmission of external Ca2+ resulted in a rise in the cytosolic GECO signal but the response was transient, fully recovering to pre-stimulation levels within 15 min (Figure 3A). No such Orai1-GECO signal was seen when BTP2 was present (Figure 3A, red trace). We repeated these experiments but stimulated cells with thapsigargin in the continuous presence of external Ca2+. Cytosolic GECO reported a rapid rise in cytosolic Ca2+, which then decayed back to pre-stimulation levels within 10–15 min (Figure 3B, labeled GECO; 3 cells from 3 independent experiments are presented to show the modest variability between experiments). Orai1-GECO responded after a delay of ∼1 min and the fluorescent signal then increased. The slower kinetics of the Orai1-GECO signal closely reflect the time taken for CRAC channels to activate following passive store depletion; CRAC current develops over several tens of seconds when store are depleted using thapsigargin. The Orai1-GECO signal remained elevated, declining by only 24± 5% after 20 min (Figure 3B). Collectively, these results confirm that CRAC channels remain open despite bulk cytosolic Ca2+ returning to pre-stimulation levels.
Figure 3.
Local Ca2+ near CRAC channels are sustained whereas cytosolic Ca2+ falls quickly. (A) Traces compare Ca2+ signals measured using cytosolic GECO or Orai1-GECO. Cells were stimulated with thapsigargin in Ca2+-free solution and external Ca2+ was readmitted as indicated. Each trace shows the means and SEM of between 12 and 17 cells. (B) As in panel A but cells were challenged with thapsigargin in the continuous presence of external Ca2+. 3 cells from 3 independent experiments are shown, to demonstrate the mild variability in responses, particularly with respect to peak amplitude. Similar data were seen in a further 3 experiments.
TIRF Microscopy Measurements of Local Ca2+ Near CRAC Channels
We used TIRF microcopy to measure Ca2+ near Orai1-GECO channels, an approach that provides an opportunity to image cytosolic Ca2+ signals within ∼100 nm of the plasma membrane. Stimulation with thapsigargin led to a sustained rise in sub-plasmalemmal Ca2+ that was maintained for 20 min (Figure 4A and B). This sustained Ca2+ signal required Ca2+ influx through CRAC channels because it was abolished rapidly by the CRAC channel blocker BTP2, added after ∼20 min stimulation (Figure 4A and B). To ensure we were recording Ca2+ signals close to Orai1-GECO, we loaded cells with the slow chelator EGTA to restrict Ca2+ entry through CRAC channels to within ∼100 nm of the plasma membrane.40,41 In the presence of cytosolic EGTA, the local Ca2+ signal remained relatively sustained and was similar to that seen in non-EGTA-loaded cells (Figure 4C).
Figure 4.
TIRF measurements of subplasmalemmal Ca2+ using Orai1-GECO. (A) Images compare Orai1-GECO Ca2+ signals under resting conditions (labeled NT for non-treated), after 20 min stimulation with thapsigargin in external Ca2+ (2Ca–TG) and then after the addition of 10 μm BTP2. (B) Graph depicts the time course of the Orai1-GECO signal for the treatments shown. Data are the average of 9 cells. (C) Graph shows the averaged time course of Orai1-GECO signal in EGTA-loaded cells.
CRAC Channels Are Functional in the Absence of a Bulk Cytosolic Ca2+ Rise
We asked whether the open CRAC channels retained a functional ability to activate gene expression, despite bulk cytosolic Ca2+ being low. Ca2+ nanodomains near open CRAC channels increase expression of the immediate early gene c-fos,42,43 an integral component of the AP-1 transcription factor complex, and the Ca2+-activated transcription factor NFAT. NFAT1–4 comprise a family of Ca2+-dependent transcription factors, which are stimulated by the Ca2+-activated protein phosphatase calcineurin, the target for immunosuppressants.44 Local Ca2+ entry through CRAC channels stimulate calcineurin.29,45 Active calcineurin then dephosphorylates NFAT, leading to the exposure of a nuclear localization sequence, which enables NFAT to migrate into the nucleus. We measured cytosolic Ca2+ and NFAT1-mCherry dynamics in the same cells at the same time. Cytosolic Ca2+ rose to peak ∼4 min after stimulation and then declined steadily. By contrast, NFAT translocated into the nucleus considerably more slowly (Figure 5A, red trace) and continued to accumulate despite cytosolic Ca2+ falling substantially. After 20 min of stimulation, cytosolic Ca2+ had fallen by 73% (relative to the peak) and by 35 min, it had fallen by 84%. Nevertheless, between 20 and 35 min, NFAT1 translocation into the nucleus increased by 27% (Figure 5A). Aggregate data comparing cytosolic Ca2+ and NFAT nuclear translocation in the same cells are shown in Figure 5B. Despite cytosolic Ca2+ being at a relatively low level and continuing to fall, NFAT activation continued. These results are consistent with earlier work that showed loading the cytosol with EGTA to prevent a bulk cytosolic Ca2+ rise failed to affect the rate or extent of nuclear translocation of NFAT.46
The Plasma Membrane Ca2+ ATPase Pump Prevents CRAC Channels From Raising Bulk Ca2+
The inability of CRAC channels to raise bulk Ca2+ several minutes after their activation suggests the existence of a mechanism that effectively removes Ca2+ from the cytosol. Major Ca2+ clearance mechanisms in non-excitable cells include plasma membrane Ca2+ ATPase pumps and Na+–Ca2+ exchangers, SERCA pumps on the ER and mitochondrial Ca2+ uptake.47 A role for SERCA pumps can be excluded because we used thapsigargin to deplete stores. We assessed the contribution of mitochondria by comparing responses to the protonophore FCCP 15 min after stimulation with thapsigargin in the presence of external Ca2+, or after the same time but without thapsigargin treatment (control). No clear increase in cytosolic Ca2+ was seen when cells pretreated with thapsigargin were challenged with FCCP (Figure 6A). These data show that mitochondria had not accumulated cytosolic Ca2+ at this time, and therefore that another Ca2+ clearance mechanism was operational and which reduced the cytosolic Ca2+ rise evolved by CRAC channels. We focussed on plasma membrane Ca2+ transport.
The plasma membrane Ca2+ATPase pump is inhibited by milliMolar concentrations of La3+. Stimulation with thapsigargin in Ca2+-free solution supplemented with 1 m m La3+ led to a substantially larger and more prolonged cytosolic Ca2+ rise than was the case in either Ca2+-free or Ca2+-containing solution (Figure 6B). The larger increase in cytosolic Ca2+ following stimulation with thapsigargin in Ca2+-free solution in the presence of La3+ has been seen in other studies,48,49 and reflects the fact that the PMCA pump is the dominant Ca2+ removal mechanism under these conditions. Another property of the plasma membrane Ca2+ATPase pump is that transport activity is inhibited by alkaline pH because the transporter exchanges cytosolic Ca2+ for external H+.50,51 Stimulation with thapsigargin in Ca2+-free solution at pH 11 produced a much larger and more sustained Ca2+ response than at pH 7.4, consistent with a major role for the plasma membrane Ca2+ pump (Figure 6C). To address a role for Na+–Ca2+ exchange, we incubated cells in Na+-free external solution for 5 min prior to challenge with thapsigargin in Na+-free solution. The pattern of the Ca2+ signal was similar to that seen in Na+-containing solution (Figure 6D). Cytosolic Ca2+ declined slightly more quickly in Na+-free solution but no difference in the half times of decay of the Ca2+ signal was observed (Figure 6E). Collectively, these results identify a major role for plasma membrane Ca2+ ATPase pump in extruding Ca2+ that has entered through CRAC channels.
Plasma Membrane Ca2+ ATPase Isoform 4b Accelerates Ca2+ Removal From the Cytosol
Mammalian plasma membrane Ca2+ATPase pumps are encoded by four separate genes.23 PMCA4b is a widely expressed member and is the major isoform found in HEK293 cells.23 If PMCA4b was responsible for removing Ca2+ entry through CRAC channels, a prediction would be that overexpression of the pump should lead to more rapid recovery of cytosolic Ca2+. Following expression of PMCA4b-cherry, we found that thapsigargin-evoked Ca2+ release was considerably smaller and more transient than in control cells (labeled WT; Figure 7A). Readmission of external Ca2+ led to a rapid rise in cytosolic Ca2+ due to store-operated Ca2+ entry (Figure 7A). The rate of Ca2+ entry was similar between cells expressing PMCA4b-cherry and mock-transfected control cells (Figure 7A; expanded in Figure 7B; aggregate data shown in Figure 7C). However, whereas control (WT) cells showed a transient plateau once cytosolic Ca2+ had peaked (Figure 7A, green trace), no plateau was evident in PMCA4b-expressing cells (Figure 7A, red trace). The subsequent decay rate of cytosolic Ca2+, measured in Ca2+-free solution, was considerably faster (∼2.5-fold) in the presence of recombinant PMCA4b (Figure 7D). Hence PMCA4b effectively removes Ca2+ that has entered the cytosol via CRAC channels. To examine whether increased expression of PMCA4b accelerated the decay of the cytosolic Ca2+ signal in the continuous presence of external Ca2+, we compared the pattern of the Ca2+ signal following thapsigargin stimulation between control cells and those expressing PMCA4b-cherry (Figure 7E). The cytosolic Ca2+ signal was smaller and returned to pre-stimulation levels ∼2-fold more quickly when the pump was overexpressed (Figure 7E).
To quantify the impact of PMCA4b overexpression on Ca2+ clearance, we stimulated cells with a high concentration of ionomycin (2 μm) in the absence of external Ca2+ and compared the duration of the cytosolic Ca2+ rise (measured as the area under the curve) between control cells and those overexpressing PMCA4b. The Ca2+ signal was smaller and more transient when PMCA4b was overexpressed (Figure 7F and G).
Relative Distribution of Orai1 and PMCA4b
We used Airyscan confocal microscopy to assess the relative spatial distribution of PMCA4B-cherry and Orai1-GFP. In unstimulated cells, both proteins were distributed throughout the cell periphery and this led to significant overlap in their distribution (Figure 8A; upper panel; co-localization is shown in white in the right hand image). Interestingly, PMCA-cherry was also contained in numerous relatively small cytosolic vesicles, and to a much greater extent that Orai1-GFP. Many vesicles seemed to contain only one type of tagged protein. After stimulation with thapsigargin in the presence of external Ca2+, PMCA distribution changed little if at all, whereas Orai1 formed clusters (Figure 8A, lower set of images). The extent of overlap was now considerably less (aggregate data are summarized in Figure 8B). These results suggest that Orai1 does not interact physically with PMCA4b. The overlap in distribution in resting cells reflects the homogenous distribution of both proteins in the plasma membrane, with some overlap by chance. However, after store depletion, Orai1 clusters at endoplasmic reticulum–plasma membrane junctions and, in the absence of a direct interaction between Orai1 and the PMCA4b, the extent of spatial overlap decreases as the pump does not accumulate at these sites.
Figure 8.
Airy scan confocal microscopy shows reduced co-localization of PMCA4b and Orai1 after stimulation. (A) Images compare distribution of Orai1-GFP and PMCA4b-cherry under resting conditions (upper) and after thapsigargin treatment for 20 min (lower). Computed co-localization is indicated in white. (B) Graph shows the change in colocalized voxel of Orai1 and PMCA4b after exposure to thapsigargin. Each point after 2 min (arrow) was significantly different from the value at 2 min. P < 0.05 for the first two points and then P < 0.01 for the rest. Data are presented as mean ± SEM.
Discussion
The widely expressed PMCA4b isoform is considered a house keeping PMCA, maintaining low cytosolic Ca2+ in resting cells and thereby preserving a concentration gradient for Ca2+ entry. Our new results demonstrate the versatility of PMCA4b in shaping Ca2+ signals evoked by store-operated Ca2+ entry. Remarkably, we find the activity of PMCA4b adjusts over time to overcome continuous store-operated Ca2+ entry and return bulk cytosolic Ca2+ to resting levels despite a prolonged increase in membrane permeability to Ca2+. Because store-operated Ca2+ entry takes several seconds to initiate and then develops over tens of seconds when stores are depleted passively with thapsigargin, the initial large increase in cytosolic Ca2+ followed by its recovery is consistent with the finding that modulation of PMCA is acquired slowly,25 taking several tens of seconds to adapt to a new level of activity.
It is noteworthy that cytosolic Ca2+ decayed back to resting levels in native HEK293 cells despite continued Ca2+ entry through CRAC channels. In Jurkat T cells, the Ca2+ signal stabilized at an elevated plateau25 and a similar profile was seen in RBL cells (Supplementary Figure 2 in52). In all three cell types, PMCA4b is expressed. Why therefore is the Ca2+ signal relatively transient in HEK cells? Two factors need to be considered: First, the number of functional CRAC channels and second, the number of active PMCA pumps along with the rate and extent of their modulation. The relative levels of expression of the pump in the different cell types are unknown. However, CRAC channel density is markedly different. Jurkat T lymphocytes and RBL cells exhibit large native CRAC currents, of ∼−15 and −40 pA at −80 mV (both equating to ∼−3 pA/pF). By contrast, the native CRAC current in HEK cell is generally too small to be measured accurately. Since the level of detection of a whole cell current in a cell the size of a HEK cell is ∼−1.5 pA, we assume the endogenous HEK cell macroscopic CRAC current is ≤1.5 pA. Taking a typical non-excitable cell buffering capacity of ∼100 and ignoring Ca2+-dependent inactivation, which would be modest over the physiological range, CRAC current would raise cytosolic Ca2+ in a HEK cell at a rate of ∼50 n m/s. By measuring the initial fast phase of decay of cytosolic Ca2+ on switching to Ca2+-free solution after Ca2+ entry through CRAC channels for 4 min, we estimate Ca2+ clearance by PMCA in HEK293 cells to be ∼25 n m/s, only 2-fold less than that reported in Jurkat T cells.25 Since PMCA modulation develops over minutes, pump activity will develop sufficiently to match the rise in cytosolic Ca2+ achieved by CRAC channel opening in HEK cells. Therefore, the main reason why cytosolic Ca2+ declines fully in HEK293 cells and only partially in Jurkat T lymphocytes most likely reflects the much larger number of functional CRAC channels in the latter cell type. Although our estimate of the rate of rise of cytosolic Ca2+ due to CRAC channel activity is not derived from empirical measurements but is based on calculations that rely on several assumptions such as the Ca2+ buffering capacity of the cytosol, the cytosolic volume and a reasonable estimate of the Ca2+ current, it is not dissimilar from the rate of Ca2+ removal by PMCA, suggesting the processes can be matched.
Our experiments in HEK293 cells are in good agreement with studies in Jurkat T cells that show PMCA4b activity remains high during store-operated Ca2+ entry.25,53 By contrast, STIM1 has been reported to inhibit PMCA4b in Jurkat T cells, dramatically slowing down Ca2+ clearance.26 This effect was attributed to the N-terminus of STIM1 because a construct truncated immediately after the proline-rich region, STIM1Δ597, no longer pulled down with PMCA in co-immunoprecipitation experiments and did not slow Ca2+ removal. The effect of STIM1 on PMCA has been been reported to depend on the presence of the protein POST. Overexpression of POST in Jurkat T cells prevented STIM1 from inhibiting PMCA4 and so Ca2+ entry through CRAC channels was cleared rapidly from the cytosol. Knockdown of endogenous POST slowed Ca2+ clearance.27 These findings conflict with an earlier study by Clapham and colleagues who showed, also in native Jurkat T cells, that siRNA-dependent knockdown of POST increased Ca2+ clearance, leading to the conclusion that POST served as an inhibitor of PMCA.54 These authors also found that POST was critical for STIM1 to bind to and inhibit PMCA.54 Further work is needed to reconcile these contradictory reports on whether and how STIM1 and POST affect PMCA activity in Jurkat T cells.
We took advantage of the genetically encoded GECO fluorescent Ca2+-sensitive protein to measure Ca2+ levels adjacent to CRAC channels and in the bulk cytosol. Whereas bulk Ca2+ returns to pre-stimulation levels within a few minutes of sustained Ca2+ entry through CRAC channels, Orai1-GECO continued to report elevated Ca2+ near the channels for several minutes. Similar results were obtained when we used TIRF microscopy to measure local Ca2+ near CRAC channels. These findings confirm that CRAC channels remain active despite bulk Ca2+ returning to resting levels and highlight the presence of stable, standing Ca2+ gradients between CRAC channels and bulk cytosol. Using a lipid-modified Fura 2 analogue that measures Ca2+ near membranes, Go et al. reported that sub-membranous Ca2+ could be reduced by overexpression of PMCA4b whereas bulk Ca2+ increased under the same conditions in jurkat T cells.27 Our direct measurements of Ca2+ adjacent to CRAC channels confirm that local and bulk Ca2+ signals are markedly different; local Ca2+ near the channels remains elevated despite bulk Ca2+ returning to pre-stimulation levels.
To develop an appropriate immune response, T lymphocytes require prolonged Ca2+ entry through CRAC channels as well as the formation of an immunological synapse with the antigen-presenting cell. STIM and Orai1 are recruited to the synapse, resulting in long-lasting local Ca2+ influx.55 Quintana et al. have demonstrated that mitochondria also accumulate at the immunological synapse and efficiently buffer Ca2+ entry through CRAC channels, outcompeting PMCA4b for Ca2+ removal.53 Mitochondria then release Ca2+ deeper into the cytosol through their Na+–Ca2+ exchanger, resulting in an elevated bulk cytosolic Ca2+. In HeLa cells, where mitochondria do not actively accumulate near CRAC channels, stimulation with thapsigargin evoked high local Ca2+ but a small bulk Ca2+ rise.53 PMCA pump activity in HeLa cells therefore matches Ca2+ entry through CRAC channels to ensure low global Ca2+, as we describe in HEK293 cells.
PMCA pumps relocate to the immunological synapse following T cell receptor stimulation. Detailed modelling studies have shown that the redistribution of PMCA pumps to the synapse results in lower local Ca2+ but a larger sustained global rise.56,57 A more homogeneous distribution of the pump, as occurs with thapsigargin stimulation, leads to a considerably lower global Ca2+ rise but a larger local Ca2+ signal below the plasma membrane. These conclusions are in excellent agreement with our experimental findings. Our Airy scan confocal microscopy data confirm an earlier study by Quintana et al., who also showed that PMCA4b and Orai1 did not colocalize in T cells either before or after stimulation with thapsigargin.53 As predicted by Hoth and colleagues, the relatively homogeneous distribution of PMCA4b pumps compared with Orai1 channels led to a high and sustained local Ca2+ but a much smaller and transient global Ca2+ signal.
Due to the lack of co-localization between PMCA4b pumps and CRAC channels, PMCA4b is unlikely to impact on the size or lateral extent of the Ca2+ nanodomain near each hexameric CRAC channel, but instead will reduce spillover or smearing of the Ca2+ signal at the edges of ER–PM junctions, thereby preventing local Ca2+ entry from developing into a more global Ca2+ rise. The more homogeneous dispersal of the pump will also ensure Ca2+ is removed throughout the cytosol, thus helping maintain a low global rise. Because a bulk rise in cytosolic Ca2+ triggers Ca2+-dependent slow inactivation of CRAC channels, the lowering of cytosolic Ca2+ by the PMCA will reduce inhibition of CRAC channels and thus, sustain Ca2+ entry. Furthermore, by sharpening the local Ca2+ signal, PMCA4b ensures the specificity of the local Ca2+ signal is maintained by restricting activation of signalling pathways to within the realm of the Orai1 Ca2+ nanodomain.
Data Availability
The data underlying this article will be shared on reasonable request to the corresponding author.
ACKNOWLEDGEMENTS
PB was supported by an EMBO Long-Term Fellowship. This work was supported by an MRC Programme Grant to ABP (grant number L01047X) and by the Intramural Research Program of the NIH, National Institute of Environmental Health Sciences.
Contributor Information
Pradeep Barak, Department of Physiology, Anatomy and Genetics, Oxford University, Oxford OX1 3PT, UK; Oxford Nanoimaging, Linacre House, Jordan Hill Business Park Banbury Road, Oxford OX2 8TA, UK.
Suneet Kaur, Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA.
Erica Scappini, Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA.
Charles J Tucker, Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA.
Anant B Parekh, Department of Physiology, Anatomy and Genetics, Oxford University, Oxford OX1 3PT, UK; Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, NIH, Research Triangle Park NC 27709, USA.
Conflict of Interest
A.B.P. holds the position of Executive Editor for Function and is blinded from reviewing or making decisions for the manuscript. The authors declare they have no conflict of interest.
References
- 1.Clapham DE. Calcium signaling. Cell. 2008;131(6):1047–1058. [DOI] [PubMed] [Google Scholar]
- 2.Petersen OH, Tepikin A.. Polarized calcium signalling in exocrine gland cells. Annu Rev Physiol. 2008;70(1):273–299. [DOI] [PubMed] [Google Scholar]
- 3.Parekh AB, Penner R.. Store-operated calcium influx. Physiol Rev. 1997;77(4):901–930. [DOI] [PubMed] [Google Scholar]
- 4.Prakriya M, Lewis RS.. Store-operated calcium channels. Physiol Rev. 2015;95(4):1383–1436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Berridge MJ, Bootman MD, Roderick HL.. Calcium signalling: dynamics, homeostasis and remodelling. Nat Rev Mol Cell Biol. 2003;4(7):517–529. [DOI] [PubMed] [Google Scholar]
- 6.Streb H, Irvine RF, Berridge MJ, Schulz I.. Release of Ca2+ from a non-mitochondrial intracellular store in pancreatic acinar cells. Nature. 1983;306(5938):67–69. [DOI] [PubMed] [Google Scholar]
- 7.Liou J, Kim ML, Heo WDet al. STIM is a calcium sensor essential for calcium-store-depletion-triggered calcium infux. Curr Biol. 2005;15(13):1235–1241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Roos J, DiGregorio PJ, Yeromin AVet al. STIM1, an essential and conserved component of store-operated Ca2+ channel function. J Cell Biol. 2005;169(3):435–445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Prakriya M, Feske S, Gwack Y, Srikanth S, Rao A, Hogan PG.. Orai1 is an essential pore subunit of the CRAC channel. Nature. 2006;443(7108):230–233. [DOI] [PubMed] [Google Scholar]
- 10.Yeromin AV, Zhang SL, Jiang W, Yu Y, Safrina O, Cahalan MD.. Molecular identification of the CRAC channel by altered ion selectivity in a mutant of Orai. Nature. 2006;443(7108):226–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Vig M, Peinelt C, Beck Aet al. CRACM1 is a plasma membrane protein essential for store-operated calcium entry. Science. 2006;312(5777):1220–1223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Bakowski D, Murray F, Parekh AB.. Annu Rev Pharmacol Toxicol. 2021;(61):629–654. [DOI] [PubMed] [Google Scholar]
- 13.Vaeth M, Feske S.. NFAT control of immune function: new frontiers for an abiding trooper. F1000Res. 2018;(7)260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Zweifach A, Lewis RS.. Slow calcium-dependent inactivation of depletion-activated calcium current. Store-dependent and -independent mechanisms. J Biol Chem. 1995;270(24):14445–14451. [DOI] [PubMed] [Google Scholar]
- 15.Glitsch MD, Parekh AB.. Ca2+ store dynamics determines the pattern of activation and inactivation of the store-operated Ca2+ current ICRAC in response to InsP3 in rat basophilic leukaemia cells. J Physiol (Lond). 2000;(523):283–290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Jacob R. Agonist-stimulated divalent cation entry into single cultured human umbilical vein endothelial cells. J Physiol (Lond). 1990;(421):55–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Gilabert J-A, Parekh AB.. Respiring mitochondria determine the pattern of activation and inactivation of the store-operated Ca2+ current ICRAC. EMBO J. 2000;19(23):6401–6407.. http://emboj.oupjournals.org/cgi/content/abstract/19/23/6401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Hoth M, Button D, Lewis RS.. Mitochondrial control of calcium channel gating: a mechanism for sustained signaling and transcriptional activation in T lymphocytes. Proc Natl Acad Sci USA. 2000;97(19):10607–10612. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Samanta K, Douglas S, Parekh AB.. Mitochondrial calcium uniporter MCU supports cytoplasmic Ca2+ oscillations, store-operated Ca2+ entry and Ca2+-dependent gene expression in response to receptor stimulation. PLoS One. 2014;9(7):e101188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Samanta K, Bakowski D, Amin N, Parekh AB.. The whole-cell Ca2+ release-activated Ca2+ current, ICRAC, is regulated by the mitochondrial Ca2+ uniporter channel and is independent of extracellular and cytosolic Na+. J Physiol. 2020;598(9):1753–1773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Yoast R, Emrich SM, Zhang Xet al. The Mitochondrial Ca2+ uniporter is a central regulator of interorganellar Ca2+ transfer and NFAT activation. J Biol Chem. 2021;297(4):101174. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Strehler EE, Caride AJ, Filoteo AG, Xiong Y, Penniston JT, Enyedi A.. Ann NY Acad Sci. 2007;1099: 226–236.. doi:1099:10.1196/annals.1387.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Strehler EE, Zacharias DA.. Role of alternative splicing in generating isoform diversity among plasma membrane calcium pumps. Physiol Rev. 2001;81(1):21–50. [DOI] [PubMed] [Google Scholar]
- 24.Pászty K, Caride AJ, Bajzer Zet al. Plasma membrane Ca2+-ATPases can shape the pattern of Ca2+ transients induced by store-operated Ca2+ entry. Sci Signal. 2015;8(364):ra19. [DOI] [PubMed] [Google Scholar]
- 25.Bautista DM, Hoth M, Lewis RS.. Enhancement of calcium signalling dynamics and stability by delayed modulation of the plasma-membrane calcium-ATPase in human T cells. J Physiol (Lond). 2002;541(3):877–894. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Ritchie MF, Samakai E, Soboloff J. STIM1 is required for attenuation of PMCA-mediated Ca2+ clearance during T-cell activation. EMBO J. 2012;31(5):1123–1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Go CK, Hooper R, Aronson MRet al. The Ca2+ export pump PMCA clears near-membrane Ca2= to facilitate store-operated Ca2+ entry and NFAT activation. Sci Signal. 2019;12(602):eaaw2627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bautista DM, Lewis RS.. Modulation of plasma membrane calcium-ATPase activity by local calcium microdomains near CRAC channels in human T cells. J Physiol (Lond). 2004;556(3):805–817. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kar P, Lin YP, Bhardwaj Ret al. The N terminus of Orai1 couples to the AKAP79 signaling complex to drive NFAT1 activation by local Ca2+ entry. Proc Natl Acad Sci USA. 2021;118(19):e2012908118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Parekh AB, Putney JWJ.. Store-operated calcium channels. Physiol Rev. 2005;85(2):757–810. [DOI] [PubMed] [Google Scholar]
- 31.Parekh AB. Regulation of CRAC channels by Ca2+-dependent inactivation. Cell calcium. 2017;(63):20–23. [DOI] [PubMed] [Google Scholar]
- 32.Hoth M, Penner R.. Calcium release-activated calcium current in rat mast cells. J Physiol (Lond). 1993;(465):359–386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Zweifach A, Lewis RS.. Rapid inactivation of depletion-activated calcium current (ICRAC) due to local calcium feedback. J Gen Physiol. 1995;105(2):209–226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Fierro L, Parekh AB.. Fast calcium-dependent inactivation of calcium release-activated calcium current (CRAC) in RBL-1 cells. J Membr Biol. 1999;168(1):9–17. [DOI] [PubMed] [Google Scholar]
- 35.Ng SW, Bakowski D, Nelson Cet al. Cysteinyl leukotriene type I receptor desensitization sustains Ca2+-dependent gene expression. Nature. 2012;482(7383):111–115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Parekh AB. Slow feedback inhibition of calcium release-activated calcium current by calcium entry. J Biol Chem. 1998;273(24):14925–14932. [DOI] [PubMed] [Google Scholar]
- 37.Malli R, Naghdi S, Romanin C, Graier WF.. Cytosolic Ca2+ prevents the subplasmalemmal clustering of STIM1: an intrinsic mechanism to avoid Ca2+ overload. J Cell Sci. 2008;121(19):3133–3139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lewis RS, Dolmetsch R, Zweifach A.. Positive and negative feedback of depletion-activated calcium channels by calcium. New York (NY): The Rockfeller University Press; 1996. [PubMed] [Google Scholar]
- 39.Dynes JL, Amcheslavsky A, Cahalan MD.. Genetically targeted single-channel optical recording reveals multiple Orai1 gating states and oscillations in calcium influx. Proc Natl Acad Sci USA. 2016;113(113):440–445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Neher E. Vesicle pools and Ca2+ microdomains: new tools for understanding their roles in neurotransmitter release. Neuron. 1998;20(3):389–399. [DOI] [PubMed] [Google Scholar]
- 41.Parekh AB. Ca2+ microdomains near plasma membrane Ca2+ channels: impact on cell function. J Physiol. 2008;586(Pt13):3043–3054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Di Capite J, Ng S-W, Parekh AB.. Decoding of cytoplasmic Ca2+ oscillations through the spatial signature drives gene expression. Curr Biol. 2009;19(10):853–858. [DOI] [PubMed] [Google Scholar]
- 43.Ng S-W, Nelson C, Parekh AB.. Coupling of Ca2+ microdomains to spatially and temporally distinct cellular responses by the tyrosine kinase Syk. J Biol Chem. 2009;284(37):24767–24772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Hogan PG, Chen L, Nardone J, Rao A.. Transcriptional regulation by calcium, calcineurin, and NFAT. Genes Dev. 2003;17(18):2205–2232. [DOI] [PubMed] [Google Scholar]
- 45.Kar P, Samanta K, Kramer H, Morris O, Bakowski D, Parekh AB.. Dynamic assembly of a membrane signaling complex enables selective activation of NFAT by orai1. Curr Biol. 2014;24(12):1361–1368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Kar P, Parekh AB.. Distinct spatial Ca2+ signatures selectively activate different NFAT transcription factor isoforms. Mol Cell. 2015;58(2):232–243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Carafoli E. Calcium signalling: a tale for all seasons. Proc Natl Acad Sci USA. 2002;99:1115–1122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Bird GS, Putney JWJ.. Capacitative calcium entry supports calcium oscillations in human embryonic kidney cells. J Physiol (Lond). 2005;562(3):697–706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Chang WC, Di Capite J, Singaravelu K, Nelson C, Halse V, Parekh AB.. Local calcium influx through calcium release-activated calcium (CRAC) channels stimulates production of an intracellular messenger and an intercellular pro-inflammatory signal. J Biol Chem. 2008;283(8):4622–4631. [DOI] [PubMed] [Google Scholar]
- 50.Niggli V, Sigel E, Carafoli E.. The purified Ca2+ pump of human-erythrocyte membranes catalyzes an electroneutral Ca2+–H+ exchange in reconstituted liposomal systems. J Biol Chem. 1982;257(5):2350–2356. [PubMed] [Google Scholar]
- 51.Schwiening CJ, Kennedy HJ, Thomas RC.. Calcium–hydrogen exchange by the plasma membrane Ca-ATPase of voltage-clamped snail neurons. Proc R Soc Lond B. 1993;253(1338):285–289. [DOI] [PubMed] [Google Scholar]
- 52.Lin Y-P, Bakowski D, Mirams GR, Parekh AB.. Selective recruitment of different Ca2+-dependent transcription factors by STIM1-Orai1 channel clusters. Nat Commun. 2019;10(1):2516. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Quintana A, Pasche M, Junker Cet al. Calcium microdomains at the immunological synapse: how Orai channels, mitochondria and calcium pumps generate local calcium signals for efficient T-cell activation. EMBO J. 2011;30(19):3895–3912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Krapivinsky G, Krapivinsky L, Stotz SC, Manasian Y, Clapham DE.. POST, partner of stromal interaction molecule 1 (STIM1), targets STIM1 to multiple transporters. Proc Natl Acad Sci USA. 2011;108(48):19234–19239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Lioudyno MI, Kozak JA, Penna Aet al. Orai1 and STIM1 move to the immunological synapse and are up-regulated during T cell activation. Proc Natl Acad Sci USA. 2008;105(6):2011–2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Maccari I, Zhao R, Peglow Met al. Cytoskeleton rotation relocates mitochondria to the immunological synapse and increases calcium signals. Cell calcium. 2016;60(5):309–321. [DOI] [PubMed] [Google Scholar]
- 57.Peglow M, Niemeyer BA, Hoth M, Rieger H.. Interplay of channels, pumps and organelle location in calcium microdomain formation. New J Phys. 2013;15(5):055022. [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
The data underlying this article will be shared on reasonable request to the corresponding author.








