Plasma membrane Ca2+ ATPases (PMCAs, also known as ATP2B1–ATP2B4) are known targets of phosphatidylinositol 4,5-bisphosphate [PtdIns(4,5)P2], but if and how they control the PtdIns(4,5)P2 pool has not been considered. We demonstrate here that PMCAs protect PtdIns(4,5)P2 in the plasma membrane from hydrolysis by phospholipase C (PLC). Comparison of active and inactive PMCAs indicates that the protection operates by two mechanisms; one requiring active PMCAs, the other not. It appears that the mechanism requiring activity is the removal of the Ca2+ required for sustained PLC activity, whereas the mechanism not requiring activity is PtdIns(4,5)P2 binding. We show that in PMCA overexpressing cells, PtdIns(4,5)P2 binding can lead to less inositol 1,4,5-triphosphate (InsP3) and diminished Ca2+ release from intracellular Ca2+ pools. Inspection of a homology model of PMCA suggests that PMCAs have a conserved cluster of basic residues forming a ‘blue collar’ at the interface between the membrane core and the cytoplasmic domains. By molecular dynamics simulation, we found that the blue collar forms four binding pockets for the phosphorylated inositol head group of PtdIns(4,5)P2; these pockets bind PtdIns(4,5)P2 strongly and frequently. Our studies suggest that by having the ability to bind PtdIns(4,5)P2, PMCAs can control the accessibility of PtdIns(4,5)P2 for PLC and other PtdIns(4,5)P2-mediated processes.

The plasma membrane Ca2+ ATPase (PMCA) belongs to the P-type ATPase family. Its major role is to remove excess Ca2+ from the cytosol and maintain intracellular Ca2+ homeostasis essential for the living cell (Brini and Carafoli, 2009; Strehler et al., 2007). The primary structure of PMCA shows about 30% identity with that of the sarco/endoplasmic reticulum Ca2+ ATPase (SERCA), and it has highly conserved sequence motifs that are common to most P-type ATPases (Verma et al., 1988). Information about the tertiary structure is not available for the PMCAs because it is difficult to crystallize them; therefore, crystal structures of other members of P-type ATPases (Bublitz et al., 2010; Toyoshima et al., 2000) are used to make structural predictions (Bublitz et al., 2011). According to these predictions, the overall cytoplasmic domain arrangement of the PMCAs is similar to that of SERCA except that PMCAs have two additional relatively large inserts; one connects the A domain with transmembrane helix 3, whereas the other is a long C-terminal tail that has binding sites for Ca2+-calmodulin and other regulatory elements.

There are four isoforms of PMCA (PMCA1–PMCA4, also known as ATP2B1–ATP2B4). Alternative splicing at splice sites ‘A’ (variants x, y, z, w) and ‘C’ (variants a, b, c) result in more than 20 PMCA variants (Strehler and Zacharias, 2001). According to their activation kinetics with Ca2+-CaM, the PMCAs are being classified as ‘slow’ and ‘fast’ pumps (Caride et al., 2001). It has long been known that, besides the activation with Ca2+-CaM, the activity of the PMCA is also increased by acidic phospholipids. Compared with other acidic lipids, however, lower concentrations of PtdIns(4,5)P2 are required for their activation (Choquette et al., 1984; Lehotsky et al., 1992; Missiaen et al., 1989a; Missiaen et al., 1989b). Early proteolysis (Enyedi et al., 1987; Zvaritch et al., 1990) and peptide binding (Brodin et al., 1992; Filoteo et al., 1992) studies identified an acidic lipid binding sequence, the so-called AL region, close to transmembrane segment M3. This sequence has eight candidate residues that, if exposed, could provide positively charged side-chains for the electrostatic interaction with the lipids. Recent studies have shown that deletions and/or mutations within/or close to this putative lipid-binding region activate the pump in the same manner as do acidic lipids (Brini et al., 2010; Pinto and Adamo, 2002).

PtdIns(4,5)P2 is a second messenger of the phospholipase C (PLC) signaling pathway (Berridge, 2009) and regulates many cellular functions, including membrane targeting (Schill and Anderson, 2009), cytoskeletal reorganization (Moss, 2012), endocytosis and exocytosis (Koch and Holt, 2012), as well as a variety of ion channels and receptors (Lemonnier et al., 2008; Michailidis et al., 2007; Michailidis et al., 2011; Suh and Hille, 2008; Trebak et al., 2009; Zhao et al., 2007). PtdIns(4,5)P2 also interacts with many different proteins through either unstructured basic-residue-rich regions or more-structured domains, such as the pleckstrin homology (PH), Tubby, Fab1, YOTB, Vac1 and EEA1 (FYVE); Phox homology (PX), epsin N-terminal homology (ENTH), clathrin assembly lymphoid myeloid (CALM); phosphotyrosine binding (PTB), and 4.1, ezrin, radixin, moiesin (FERM) domains, and regulates their activity and/or localization (Balla, 2005; McLaughlin and Murray, 2005; McLaughlin et al., 2002).

In the last three decades we have learned how the overall level of PtdIns(4,5)P2 in the membrane is modulated by phosphoinositol phosphate kinases and phospholipase C. However, it has become evident that specific membrane microdomains of high PtdIns(4,5)P2 levels are formed by complexes of proteins with PtdIns(4,5)P2. Such proteins have been called ‘PIPmodulins’ (Gamper and Shapiro, 2007; Lanier and Gertler, 2000). In the brain the myristoylated-alanine-rich C-kinase substrate (MARCKS) proteins have been implicated as major collectors of PtdIns(4,5)P2 molecules (Kwiatkowska, 2010). Recent experiments have suggested that syntaxin-1A could also induce PtdIns(4,5)P2 segregation and that PtdIns(4,5)P2 could sequester syntaxin-1A into distinct lipid–protein clusters (van den Bogaart et al., 2011).

Here, we show for the first time that PMCAs can serve as PIPmodulins because they attract PtdIns(4,5)P2 molecules and protect them from excessive depletion by phospholipase C. We also show that PMCA expression decreases receptor-mediated inositol 1,4,5-triphosphate (InsP3) formation and, hence, Ca2+ release from intracellular pools. Molecular dynamics simulation revealed a region of positively charged side chains that appears to interact more strongly with PtdIns(4,5)P2 than does the previously described AL region.

Experimental design to study the role of PMCAs in PtdIns(4,5)P2 signaling

Previously a fluorescence resonance energy transfer (FRET)-based direct binding assay has suggested that there is a strong interaction between the purified erythrocyte PMCA pump and PtdIns(4,5)P2 (Lehotsky et al., 1992). Here, we wanted to see how PMCA affected PtdIns(4,5)P2 signaling in live cells. In order to study this we used three species of PMCA and two types of HeLa cells in five distinct experimental setups: (1) sh-HeLa cells, in which the endogenous PMCA4 was knocked down to achieve the lowest possible PMCA level (supplementary material Fig. S1); (2) sh-HeLa cells overexpressing PMCA2wb, the ‘w’ splice variant of PMCA2b, which is a rapidly activated ‘fast’ PMCA isoform encoded by the gene ATP2B2 (Caride et al., 2001); (3) wild-type HeLa cells, with small amounts of endogenous PMCA4 and PMCA1; (4) wild-type HeLa cells overexpressing PMCA4b-LA, which is a localization mutant of the slowly activated PMCA4b encoded by gene ATP2B4 [the designation LA indicates mutation of three leucine residues to alanine residues at the extreme C-terminus of PMCA4b at positions 1167–1169 (highlighted in red letters on a gray background in the sequence of supplementary material Fig. S4) – this mutant had the same enzymatic characteristics as the wild-type PMCA4b pump, but showed enhanced plasma membrane localization (Antalffy et al., 2013)]; and (5) wild-type HeLa cells overexpressing a non-functional (dead) PMCA4b-(D672E)-LA mutant (hereafter referred to as dPMCA4b-LA; the location of the D672E mutation is shown in supplementary material Fig. S4, as a white letter on purple). Given that this mutant cannot transport Ca2+ (Adamo et al., 1995), it allowed us to separate the Ca2+ extrusion from the PtdIns(4,5)P2 binding characteristics of the pump. This set of five conditions, covered the range from cells almost lacking PMCAs (1 and 3), through cells containing intact but inactive PMCAs (5), cells containing large amounts of a ‘slow’ PMCA (4), to cells containing large amounts of a ‘fast’ PMCA (2). The relative levels of PMCA expression in the different cell types and the different experimental setups are shown in the supplementary material Fig. S1. It is important to note that the expression levels of the different PMCA constructs are comparable, and ∼100 times higher than that of the endogenous pumps in wild-type HeLa cells.

In our studies, we also used four fluorescent reporter molecules. GFP or mCherry was covalently attached to PMCA and allowed visualization of the PMCA location. The third reporter molecule, PHPLCδ1–RFP [which has the pleckstrin homology (PH) domain of PLCδ1 fused to RFP] (Szentpetery et al., 2009; Várnai and Balla, 2008) is a sensor of PtdIns(4,5)P2 and of InsP3 molecules. We refer to this construct as the PtdIns(4,5)P2 sensor. The fourth reporter, GCaMP2, is a sensor of the intracellular Ca2+ concentration.

PMCAs protect PtdIns(4,5)P2 molecules from ionomycin-induced hydrolysis

Fig. 1 shows how the intracellular distribution of the PtdIns(4,5)P2 sensor was affected by expression of PMCA when Ca2+ was introduced into the cell. Before introduction of Ca2+ by addition of ionomycin, the PtdIns(4,5)P2 sensor localized mainly in the plasma membrane either when expressed alone or together with PMCA2wb or PMCA4b-LA. The graphs show the PtdIns(4,5)P2 sensor (red) and PMCA (green) signal intensity profiles along the line of interest marked on the confocal images.

Fig. 1.

The presence of PMCA proteins prevents the translocation of the PtdIns(4,5)P2 sensor during ionomycin treatments. The PtdIns(4,5)P2 sensor, PHPLCδ1–RFP was expressed alone or together with GFP–PMCA2wb in sh-HeLa cells; or together with GFP-PMCA4b-LA in HeLa cells. (A) The distribution of the PtdIns(4,5)P2 sensor and the PMCA constructs 1 day after transfection in resting cells. Graphs display the PHPLCδ1–RFP (red) and PMCA (green) signal intensity profiles along the line of interest marked on the confocal images. (B) The same cells as in A 2 minutes after the addition of 10 µM ionomycin. Scale bars: 10 µm.

Fig. 1.

The presence of PMCA proteins prevents the translocation of the PtdIns(4,5)P2 sensor during ionomycin treatments. The PtdIns(4,5)P2 sensor, PHPLCδ1–RFP was expressed alone or together with GFP–PMCA2wb in sh-HeLa cells; or together with GFP-PMCA4b-LA in HeLa cells. (A) The distribution of the PtdIns(4,5)P2 sensor and the PMCA constructs 1 day after transfection in resting cells. Graphs display the PHPLCδ1–RFP (red) and PMCA (green) signal intensity profiles along the line of interest marked on the confocal images. (B) The same cells as in A 2 minutes after the addition of 10 µM ionomycin. Scale bars: 10 µm.

Ionomycin treatment increases [Ca2+]i, which leads to the activation of PLC and then PtdIns(4,5)P2 hydrolysis, resulting in the translocation of the PtdIns(4,5)P2 sensor from the plasma membrane to the cytosol (Szentpetery et al., 2009). When the PtdIns(4,5)P2 sensor moves from the plasma membrane to the cytosol, this reflects both the reduction of PtdIns(4,5)P2 in the plasma membrane and the generation of InsP3 in the cytoplasm (Hirose et al., 1999). Fig. 1B shows that PMCAs dramatically reduce the movement of the PtdIns(4,5)P2 sensor during ionomycin treatment. In the absence of PMCA, the PtdIns(4,5)P2 sensor was distributed homogeneously over the entire cell, whereas in cells expressing GFP–PMCA4b-LA or GFP–PMCA2wb, a high level of the PtdIns(4,5)P2 sensor signal was detected in the plasma membrane even after the ionomycin treatment (see the graphs in Fig. 1).

To study further the effect of PMCAs on the distribution of PtdIns(4,5)P2, cells were exposed to increasing concentrations of ionomycin (Fig. 2). Images in Fig. 2A show the location of the PtdIns(4,5)P2 sensor before treatments and at 5 minutes after the addition of 2, 5 and 10 µM ionomycin, as indicated. The images of Fig. 2A compare the effect of PMCA4b-LA (bottom) with control wild-type HeLa cells (middle) and cells in which PMCA4 is suppressed (sh-HeLa cells; top). The graphs of Fig. 2B show time courses of the changes in the cytosolic fluorescence intensities of the PtdIns(4,5)P2 sensor, and the bar graph in Fig. 2C shows the frequency of cells responding to a given ionomycin concentration with complete translocation of the sensor. Experiments of this sort allowed us to classify cells according to their sensitivity to ionomycin. In Fig. 2B, the signal measured in sh-HeLa cells are shown in gray color. The light grey curve shows the averaged responses of highly ionomycin-sensitive cells (18% of total sh-HeLa cells) in which addition of 2 µM ionomycin induced full translocation of the PtdIns(4,5)P2 sensor from the plasma membrane to the cytosol; 24% of cells (Fig. 2B, medium-gray curve) responded to 5 µM ionomycin and the remaining cells (Fig. 2B, dark gray curve) responded to 10 µM ionomycin resulting in full PtdIns(4,5)P2 sensor translocation in all sh-HeLa cells (see also supplementary material Movie 1). Expression of GFP–PMCA2wb in sh-HeLa cells protected the PtdIns(4,5)P2 pool, so that the sensor remained in the plasma membrane region during the ionomycin treatment even at high (10 µM) ionomycin concentration (Fig. 2B, red curve). Similarly, ionomycin could not destroy PtdIns(4,5)P2, and the PtdIns(4,5)P2 sensor remained in the plasma membrane when GFP–PMCA4b-LA was expressed in wild-type HeLa cells (Fig. 2B, blue curve; supplementary material Movie 2).

Fig. 2.

The effect of active and inactive PMCA constructs on the translocation of the PtdIns(4,5)P2 sensor during ionomycin treatments. (A) sh-HeLa (top images) or HeLa (middle images) cells were transfected with PHPLCδ1–RFP alone or together with GFP-tagged PMCA4b-LA (bottom images), as in Fig. 1. The cells were treated with 2 µM, 5 µM and 10 µM ionomycin (iono) at 0, 6 and 12 minutes, respectively. The fluorescence of the PtdIns(4,5)P2 sensor is shown before treatments, and 5 minutes after the indicated concentration of ionomycin was added. The translocation of the PtdIns(4,5)P2 sensor is reflected as an increase of the RFP fluorescence in the cytosol. Scale bars: 10 µm. (B) The graphs show the changes of PHPLCδ1–RFP cytosolic fluorescence intensity during the treatments. The fluorescence intensity ratio (Ft/F0) is plotted as a function of time. Arrows indicate the times (0, 6 and 12 minutes) when 2 µM, 5 µM and 10 µM ionomycin were added. The gray curves (left panel) show changes of cytosolic PHPLCδ1–RFP intensity values in sh-HeLa cells expressing the sensor without PMCA (supplementary material Movie 1). These cells were distinguished depending on their sensitivity to ionomycin. The red curve shows the fluorescence of the PHPLCδ1–RFP sensor in sh-HeLa cells expressing GFP-PMCA2wb. The right panel shows the intensity values of the PHPLCδ1–RFP sensor in HeLa cells expressing GFP-PMCA4b-LA (blue; supplementary material Movie 2) and the inactive GFP–PMCA4b-LA (dPMCA4b-LA) pump (green). (C) Frequency of cells responding to a given concentration of ionomycin with complete translocation of PHPLCδ1–RFP. The percentage of cells not responding to ionomycin is not shown. Three independent experiments were performed (n = 30–60 cells were counted). Statistical significance of differences between cells expressing or not the different PMCA construct was analyzed using the χ2 test. *P<0.05; ***P<0.0001.

Fig. 2.

The effect of active and inactive PMCA constructs on the translocation of the PtdIns(4,5)P2 sensor during ionomycin treatments. (A) sh-HeLa (top images) or HeLa (middle images) cells were transfected with PHPLCδ1–RFP alone or together with GFP-tagged PMCA4b-LA (bottom images), as in Fig. 1. The cells were treated with 2 µM, 5 µM and 10 µM ionomycin (iono) at 0, 6 and 12 minutes, respectively. The fluorescence of the PtdIns(4,5)P2 sensor is shown before treatments, and 5 minutes after the indicated concentration of ionomycin was added. The translocation of the PtdIns(4,5)P2 sensor is reflected as an increase of the RFP fluorescence in the cytosol. Scale bars: 10 µm. (B) The graphs show the changes of PHPLCδ1–RFP cytosolic fluorescence intensity during the treatments. The fluorescence intensity ratio (Ft/F0) is plotted as a function of time. Arrows indicate the times (0, 6 and 12 minutes) when 2 µM, 5 µM and 10 µM ionomycin were added. The gray curves (left panel) show changes of cytosolic PHPLCδ1–RFP intensity values in sh-HeLa cells expressing the sensor without PMCA (supplementary material Movie 1). These cells were distinguished depending on their sensitivity to ionomycin. The red curve shows the fluorescence of the PHPLCδ1–RFP sensor in sh-HeLa cells expressing GFP-PMCA2wb. The right panel shows the intensity values of the PHPLCδ1–RFP sensor in HeLa cells expressing GFP-PMCA4b-LA (blue; supplementary material Movie 2) and the inactive GFP–PMCA4b-LA (dPMCA4b-LA) pump (green). (C) Frequency of cells responding to a given concentration of ionomycin with complete translocation of PHPLCδ1–RFP. The percentage of cells not responding to ionomycin is not shown. Three independent experiments were performed (n = 30–60 cells were counted). Statistical significance of differences between cells expressing or not the different PMCA construct was analyzed using the χ2 test. *P<0.05; ***P<0.0001.

To show the effect of an inactive PMCA in sequestering and protecting the PtdIns(4,5)P2 molecules in the plasma membrane, we used the GFP-tagged inactive dPMCA4b-LA mutant. Fig. 2B,C shows that in HeLa cells expressing the inactive pump, high (10 µM) ionomycin concentration was required to induce substantial movement of the PtdIns(4,5)P2 sensor into the cytosol, whereas lower ionomycin concentrations caused translocation of the PtdIns(4,5)P2 sensor in only a few cells (Fig. 2C). In contrast, 27% of wild-type HeLa cells expressing the endogenous low amounts of PMCA4b and PMCA1b responded to 5 µM ionomycin. Unlike sh-HeLa cells, however, none of the wild-type HeLa cells responded to 2 µM ionomycin, indicating that the endogenous pumps also provide some protection from PLC. Addition of 10 µM ionomycin, by contrast, induced translocation of the sensor in all wild-type or sh-HeLa cells. From these experiments, it is evident that functional PMCAs can protect the PtdIns(4,5)P2 pool from an excess Ca2+ overload and PLC activation. The comparison of the dPMCA4b-LA-expressing cells with the wild-type HeLa cells in Fig. 2C suggests that even the inactive PMCA mutant could provide a significant shielding of PtdIns(4,5)P2 from hydrolysis.

We also examined how the different PMCA variants affected the Ca2+ transients during the ionomycin treatments (Fig. 3) by repeating the same experiments as above using the GCaMP2 Ca2+ sensor to monitor [Ca2+]i and mCherry-tagged PMCAs (supplementary material Fig. S1). In sh-HeLa cells, ionomycin induced long lasting Ca2+ increases and a high final signal value (Fig. 3B, black curve). In cells expressing mCherry–PMCA2wb, addition of ionomycin induced only small and short-lived Ca2+ signals (Fig. 3B, red curve) explaining why the PtdIns(4,5)P2 sensor was not translocated in the corresponding experiments. Expression of the mCherry–PMCA4b-LA pump resulted in much larger ionomycin-evoked Ca2+ signals (Fig. 3B, blue curve) than in the PMCA2wb-expressing cells, although they were somewhat less pronounced than in the sh-HeLa cells. In cells expressing the inactive dPMCA4b-LA, the [Ca2+]i response upon ionomycin stimulation (Fig. 3B, green curve) was greater than that measured in PMCA4b-LA expressing cells, as expected. The response of the dPMCA4b-LA-expressing cells was similar to that of the wild-type HeLa cells in this respect (data not shown).

Fig. 3.

Ca2+ transients in HeLa cells expressing active and inactive PMCA constructs treated with increasing concentrations of ionomycin. (A) sh-HeLa and HeLa cells were transfected with the Ca2+ sensor, GCaMP2 alone or together with mCherry-tagged PMCA constructs. The cells were treated with 2 µM, 5 µM and then 10 µM ionomycin in the same way as in the experiment shown in Fig. 2. The fluorescence of the GCaMP2 sensor is shown before the treatment and after the addition of 10 µM ionomycin. Scale bars: 10 µm. (B) The graphs show the changes of the cytosolic fluorescence intensity of the GCaMP2 sensor, during the treatments. The fluorescence intensity ratio (Ft/F0) was plotted as a function of time. Arrows indicate times when 2 µM, 5 µM and 10 µM ionomycin were added. The black curve (left panel) shows the changes in the GCaMP2 intensity values in sh-HeLa cells expressing the GCaMP2 sensor without PMCA. The red curve shows the fluorescence of the GCaMP2 sensor in sh-HeLa cells expressing mCherry-PMCA2wb. The right panel shows the intensity values of GCaMP2 in HeLa cells expressing mCherry-PMCA4b-LA (blue) and the inactive mCherry–PMCA4b-LA (dPMCA4b-LA) pump (green). Values represent mean±s.e.m. of 15–30 cells from two or three independent experiments.

Fig. 3.

Ca2+ transients in HeLa cells expressing active and inactive PMCA constructs treated with increasing concentrations of ionomycin. (A) sh-HeLa and HeLa cells were transfected with the Ca2+ sensor, GCaMP2 alone or together with mCherry-tagged PMCA constructs. The cells were treated with 2 µM, 5 µM and then 10 µM ionomycin in the same way as in the experiment shown in Fig. 2. The fluorescence of the GCaMP2 sensor is shown before the treatment and after the addition of 10 µM ionomycin. Scale bars: 10 µm. (B) The graphs show the changes of the cytosolic fluorescence intensity of the GCaMP2 sensor, during the treatments. The fluorescence intensity ratio (Ft/F0) was plotted as a function of time. Arrows indicate times when 2 µM, 5 µM and 10 µM ionomycin were added. The black curve (left panel) shows the changes in the GCaMP2 intensity values in sh-HeLa cells expressing the GCaMP2 sensor without PMCA. The red curve shows the fluorescence of the GCaMP2 sensor in sh-HeLa cells expressing mCherry-PMCA2wb. The right panel shows the intensity values of GCaMP2 in HeLa cells expressing mCherry-PMCA4b-LA (blue) and the inactive mCherry–PMCA4b-LA (dPMCA4b-LA) pump (green). Values represent mean±s.e.m. of 15–30 cells from two or three independent experiments.

PMCAs reduce the receptor-mediated PtdIns(4,5)P2 signals

To study further the role of the PMCAs in PtdIns(4,5)P2 signaling we induced activation of PLC through G-protein-coupled receptors. In this case, initial activation of PLC does not need an increase in [Ca2+]i. Instead, PLC is activated through G-proteins to hydrolyze PtdIns(4,5)P2 and release InsP3, which then stimulates Ca2+ release from the endoplasmic reticulum (ER) (Berridge, 2009). An example of the type of protocol used in our experiments is shown in supplementary material Fig. S2. First, the PLC signaling pathways were activated with a saturating concentration of extracellular ATP. After stimulation, purinergic receptors quickly desensitize (Mundell and Kelly, 2011), therefore we used histamine, another G-protein-coupled agonist, to test how effectively the cells can respond to a second stimulus. Both receptor stimuli were added in a Ca2+-free medium to show only the InsP3-mediated signal. It is clear from supplementary material Fig. S2 that in response to ATP stimulation only a fraction (10–20%; see also Fig. 4A1,B1) of total PtdIns(4,5)P2 is degraded, inducing robust Ca2+ release from the internal stores in good accordance with recent findings (Dickson et al., 2013). After 5 minutes, Ca2+ was added back to the medium to allow the entry of extracellular Ca2+ into the cells through channels that open in response to the low concentration of Ca2+ in the lumen of the ER (store-operated Ca2+ entry, SOCE) (Bird et al., 2008). Finally, we added a high concentration of ionomycin to evoke a maximal response of the system.

Fig. 4.

Effect of PMCA constructs on PtdIns(4,5)P2 and Ca2+ signaling during receptor-mediated PLC activation. sh-HeLa cells were transfected with PHPLCδ1–RFP or the GCaMP2 sensor alone or together with GFP- or mCherry-tagged PMCA2wb. HeLa cells were co-transfected with either PHPLCδ1–RFP or GCaMP2 and the appropriate PMCA4b-LA or inactive PMCA4b-LA constructs. PHPLCδ1–RFP and the GFP-tagged PMCA constructs were used in panels A1 and B1, and GCaMP2 and the mCherry-tagged PMCA constructs in panels A2 and B2, as indicated. Cells were stimulated with 100 µM ATP and 100 µM histamine (2 minutes after ATP) in the absence of Ca2+. The extracellular medium was replenished by 2 mM Ca2+ after 5 minutes. More details of the experimental protocol are described in supplementary material Fig. S2. (A,B) A1 and B1 show the PHPLCδ1–RFP signals, A2 and B2 show the Ca2+ transients measured in cells expressing the sensor alone (black), together with GFP–PMCA4b-LA (blue), GFP-dPMCA4b-LA (green) or GFP–PMCA2wb (red), as indicated. Values represent mean±s.e.m. of 10–30 cells from two or three independent experiments. (C) Areas of Ca2+ (GCaMP2) signals under the ATP peaks (C1) and Ca2+ entry peaks (C2) (arbitrary units). Data are mean±s.e.m. of 15–60 cells analyzed from three independent experiments. Asterisks represent significant difference between bars indicated with brackets. **P<0.01; ***P<0.0001, Student's t-test.

Fig. 4.

Effect of PMCA constructs on PtdIns(4,5)P2 and Ca2+ signaling during receptor-mediated PLC activation. sh-HeLa cells were transfected with PHPLCδ1–RFP or the GCaMP2 sensor alone or together with GFP- or mCherry-tagged PMCA2wb. HeLa cells were co-transfected with either PHPLCδ1–RFP or GCaMP2 and the appropriate PMCA4b-LA or inactive PMCA4b-LA constructs. PHPLCδ1–RFP and the GFP-tagged PMCA constructs were used in panels A1 and B1, and GCaMP2 and the mCherry-tagged PMCA constructs in panels A2 and B2, as indicated. Cells were stimulated with 100 µM ATP and 100 µM histamine (2 minutes after ATP) in the absence of Ca2+. The extracellular medium was replenished by 2 mM Ca2+ after 5 minutes. More details of the experimental protocol are described in supplementary material Fig. S2. (A,B) A1 and B1 show the PHPLCδ1–RFP signals, A2 and B2 show the Ca2+ transients measured in cells expressing the sensor alone (black), together with GFP–PMCA4b-LA (blue), GFP-dPMCA4b-LA (green) or GFP–PMCA2wb (red), as indicated. Values represent mean±s.e.m. of 10–30 cells from two or three independent experiments. (C) Areas of Ca2+ (GCaMP2) signals under the ATP peaks (C1) and Ca2+ entry peaks (C2) (arbitrary units). Data are mean±s.e.m. of 15–60 cells analyzed from three independent experiments. Asterisks represent significant difference between bars indicated with brackets. **P<0.01; ***P<0.0001, Student's t-test.

Next, we examined how PMCAs affected InsP3 formation and the Ca2+ signal in the absence of external Ca2+. In both HeLa and sh-HeLa cells, addition of ATP resulted in a sustained increase in the cytosolic intensity of the PtdIns(4,5)P2 sensor indicating a substantial release of InsP3 (Fig. 4A1,B1, black curves). In good correlation with the elevated InsP3 concentration, the corresponding Ca2+ signaling experiments in Fig. 4A2,B2 (black curves) showed a rapid Ca2+ release from the internal store followed by a slowly descending phase. The subsequent histamine stimulus resulted in only minor peaks of both InsP3 and Ca2+ superimposed on the first one suggesting that the freely available PtdIns(4,5)P2 and/or ER resources were largely depleted. The shape of the Ca2+ signals was similar in both wild-type HeLa and sh-HeLa cells (Fig. 4A2,B2) and the area under the ATP peaks was not significantly different between the two types of cells (Fig. 4C1).

In contrast, in HeLa cells overexpressing PMCA4b-LA, ATP caused a much shorter Ca2+ peak followed by a short histamine-induced secondary response (Fig. 4B2, blue curve). Consistent with this, the translocation of the PtdIns(4,5)P2 sensor was also less pronounced (Fig. 4B1), and the signal decayed after the ATP and histamine stimuli, indicating quick relocation of the sensor to the plasma membrane in the PMCA4b-LA-expressing cells. Strikingly, the Ca2+ signals in cells expressing PMCA4b-LA or the inactive dPMCA4b-LA were almost identical during the early period of agonist stimulation in the absence of extracellular Ca2+ (Fig. 4B2,C1). The fact that Ca2+ returned to the baseline level in case of the inactive dPMCA4b-LA underlines the importance of other mechanisms (the SERCA pump and/or mitochondria) in eliminating Ca2+ from the cytosol. The signals of the PtdIns(4,5)P2 sensor were also similar, although the cells with the inactive dPMCA4b-LA showed a slightly higher peak after the first stimulus than the cells with the active pump (Fig. 4B1). These experiments suggest that during the early period of agonist stimulation, the slow PMCA4 variant did not act through its Ca2+-removing ability but rather through its ability to bind PtdIns(4,5)P2 making it less accessible to PLC. To fulfill this role it is likely that a relatively large amount of the pump is required, which is accomplished by the transient transfection in our experiments (supplementary material Fig. S1B).

The PtdIns(4,5)P2 sensor signal was similar in cells expressing either the ‘fast’ PMCA2wb or the ‘slow’ PMCA4b-LA pumps (Fig. 4A1,B1), suggesting that the PtdIns(4,5)P2-binding capacity of both pumps was similar. The Ca2+ transients, however, returned to the baseline level more quickly in cells expressing the ‘fast’ PMCA2wb (supplementary material Fig. S3A), indicating that in the ‘slow’ PMCA4b-LA-expressing cells it was mostly the PtdIns(4,5)P2-binding ability that contributed to the attenuation of the InsP3-mediated Ca2+ signal, whereas in the case of the ‘fast’ PMCA2wb-expressing cells it was both the PtdIns(4,5)P2-binding and pumping function that contributed. Control experiments shown in supplementary material Fig. S3B, show that expression of the PtdIns(4,5)P2 sensor alone had no effect on the Ca2+ signal, suggesting that the sensor does not interfere with PtdIns(4,5)P2 hydrolysis, as expected (Szentpetery et al., 2009; van der Wal et al., 2001).

Fig. 4 also shows that the Ca2+-pumping activity of PMCA4b-LA was essential in removing Ca2+ after Ca2+ entry through the plasma membrane Ca2+ channels. Adding Ca2+ back to the PMCA4b-LA-expressing cells after ER store depletion resulted in a Ca2+ peak with smaller amplitude and shorter period than that observed in wild type HeLa cells. Here, we found a pronounced difference between cells expressing the active or the inactive dPMCA4b-LA; in the dPMCA4b-LA-expressing cells the Ca2+ curve had a long lasting decay phase similarly to the control cells (Fig. 4B2,C2). These results are in accordance with previous findings on the role of the PMCA in shaping Ca2+-entry-mediated responses (Bautista et al., 2002; Bautista and Lewis, 2004; Ritchie et al., 2012; Snitsarev and Taylor, 1999).

Molecular dynamics simulation revealed four PtdIns(4,5)P2-binding pockets around the neck of the PMCAs forming a ‘blue collar’

Next, we wanted to understand how PMCAs provide such a protecting environment for the PtdIns(4,5)P2 molecules. The model of PMCA in the E1 conformation (see Materials and Methods) shows a collar of positively charged residues (Arg and Lys) at the cytoplasmic face of the membrane (Fig. 5A). Fig. 5B–D shows the surface electrostatic potential representations of B, the same molecule; C, a hypothetical molecule missing the PMCA-specific inserts (these include the AL region and the complete C-terminal tail); and D, the E1 conformation of SERCA (PDB 1T5S). These maps show a nearly continuous positive surface potential around the neck (the ‘blue collar’) of the PMCA that must arise from a cluster of basic residues. The previously identified lipid-binding (AL) domain was a stretch of 25 amino acids, which were contiguous in the primary sequence of PMCA. In contrast, the ‘blue collar’ is composed of residues from all parts of the stalk regions of PMCA, which is also present in the hypothetical PMCA molecule missing the AL region (Fig. 5C). The blue collar residues are assembled into a coherent structure when the secondary and tertiary structures of PMCA are formed. Fig. 5D shows SERCA, which lacks nearly all basic residues in the stalk region (supplementary material Figs S4, S5) and therefore, also the blue collar.

Fig. 5.

The blue collar is present in PMCA but lacking in SERCA. (A) Ribbon diagram of the model of PMCA4b, made as described in the Materials and Methods. Arginine and lysine residues in the lower half of the stalk, near the cytoplasmic surface of the membrane comprise the blue collar; they are shown in space-filling blue. (B) The electrostatic potential at the surface of the same model as in A, with red representing negatively charged surface, white neutral surface and blue positively charged surface. (C) A molecules as in B, except that the inserts that are not homologous to SERCA are omitted. As that part of the figure shows, those inserts contributed mainly to the negatively charged surface regions; their omission did not affect the blue collar. (D) The E1 conformation of SERCA (PDB 1T5S) which was used to make the PMCA model. This structure lacks the blue collar.

Fig. 5.

The blue collar is present in PMCA but lacking in SERCA. (A) Ribbon diagram of the model of PMCA4b, made as described in the Materials and Methods. Arginine and lysine residues in the lower half of the stalk, near the cytoplasmic surface of the membrane comprise the blue collar; they are shown in space-filling blue. (B) The electrostatic potential at the surface of the same model as in A, with red representing negatively charged surface, white neutral surface and blue positively charged surface. (C) A molecules as in B, except that the inserts that are not homologous to SERCA are omitted. As that part of the figure shows, those inserts contributed mainly to the negatively charged surface regions; their omission did not affect the blue collar. (D) The E1 conformation of SERCA (PDB 1T5S) which was used to make the PMCA model. This structure lacks the blue collar.

In order to observe if and where the PtdIns(4,5)P2 molecules bind to the blue collar, 15 molecular dynamics runs were carried out; each run lasted 10 nanoseconds and contained five PtdIns(4,5)P2 molecules, for a total of 75 PtdIns(4,5)P2 molecules. Of these 75 molecules, 20 bound to one residue, 24 bound to more than one amino acid residue and 31 remained unbound. A PtdIns(4,5)P2 molecule was considered bound when it remained within 4.0 Å of a PMCA residue for many nanoseconds. Each run showed a different pattern of binding because of the randomness of the original PtdIns(4,5)P2 positions, but these runs showed four pockets of positively charged residues that bound PtdIns(4,5)P2.

The images in Fig. 6A,B show the four binding pockets; left to right, they are K173 (red), R993 (green), K980 (purple) and R914 (blue). Of the 24 multiply bound PtdIns(4,5)P2 molecules, eight bound to pocket R993, seven to R914, five to K980, three to K173 and one in an unusual way to residues K365 and K909. The interaction energies of the PtdIns(4,5)P2 molecules with their binding pockets are summarized in Fig. 6D. Given that it was only possible to do 10-nanosecond runs, it is probable that these PtdIns(4,5)P2 molecules are not yet in their most favorable positions relative to the binding pocket. This point of view is reinforced by the observation that the positions of PtdIns(4,5)P2 molecules in the different runs are different. For comparison, the interaction energy was calculated for a PtdIns(4,5)P2-binding site from a K+ channel (PDB 3SPI) (Hansen et al., 2011). Given that this structure was that of a crystal, we would expect the PtdIns(4,5)P2 and the binding pocket to be closer to their most favorable position. This is probably why the interaction energy is stronger for the 3SPI channel than the strongest interaction in PMCA.

Fig. 6.

The four pockets that bind PtdIns(4,5)P2. (A) PMCA4b with the four PtdIns(4,5)P2-binding pockets embedded in the bilayer. On the left, for comparison, is the surface potential of PMCA4b. Pocket R993 is outlined by a rectangle, and the area of the rectangle is shown in a stick model (C). (B) A slice through the model showing the pockets from above. The residues of pocket R993 are highlighted in green, pocket R914 in blue, pocket K980 in purple and pocket K173 in red. (D) The relative energies of the PtdIns(4,5)P2-pocket complexes are shown, the 3SPI bar is the energy of the complex in a known PtdIns(4,5)P2-binding protein. (E) A summary of the residues involved in each pocket.

Fig. 6.

The four pockets that bind PtdIns(4,5)P2. (A) PMCA4b with the four PtdIns(4,5)P2-binding pockets embedded in the bilayer. On the left, for comparison, is the surface potential of PMCA4b. Pocket R993 is outlined by a rectangle, and the area of the rectangle is shown in a stick model (C). (B) A slice through the model showing the pockets from above. The residues of pocket R993 are highlighted in green, pocket R914 in blue, pocket K980 in purple and pocket K173 in red. (D) The relative energies of the PtdIns(4,5)P2-pocket complexes are shown, the 3SPI bar is the energy of the complex in a known PtdIns(4,5)P2-binding protein. (E) A summary of the residues involved in each pocket.

The strongest interaction energy for a binding of PtdIns(4,5)P2 to R993 was −2550 kJ/mol, about 62% of the −4091 kJ/mol found for binding of PtdIns(4,5)P2 to its site in the K+ channel (Hansen et al., 2011). The strength of the interaction of PtdIns(4,5)P2 with R993 shows that its binding is probably biologically significant.

Table 1 shows that PtdIns(4,5)P2 binds to amino acid residues more strongly than other phospholipids do. PtdIns(4,5)P2 also binds more frequently to multiple residues. Not only does PtdIns(4,5)P2 have a higher negative charge than the other lipids, but its negative charge is in two locations, the P1 phosphorus atom and the P4 and P5 phosphorus atoms. The P1 atom is closer to the surface of the membrane, where it can bind to the lower part of the blue collar, whereas the P4 and P5 atoms are further from the membrane surface and can bind to the upper parts of the blue collar.

Table 1.
The frequency with which each lipid binds to multiple amino acids
graphic
graphic

Phosphatidylcholine and phosphatidylethanolamine bind to two (doubles) or three (triples) amino acids ∼12% of the total interactions. PtdIns and phosphatidylserine bind to multiple residues at an even lower frequency, ∼2%. PtdIns(4,5)P2 binds to multiple residues much more frequently (48.3%), with many triples and quadupless. Eav is the average energy of binding per lipid. In the case of multiple binding, all the interactions are included. Eav is much higher for PtdIns(4,5)P2 because of the multiple bindings, many of which are quite strong.

To compile Table 1, all of the saved coordinates from the 15 molecular dynamics runs were analyzed. The coordinates were saved every 0.2 nanoseconds except that the first few runs were saved more frequently.

This charge distribution of PtdIns(4,5)P2 contributes especially strongly to its binding to pocket R993. The alignments of supplementary material Figs S4 and S5 show the alpha carbon of residue R993 in the middle of transmembrane helix M9. In the three-dimensional model, residue R993 reaches towards the cytoplasm, so that the charged guanido group is in the amphiphilic boundary layer between the hydrophobic core of the membrane and the cytoplasm. Here, it is positioned so that it will interact with the P1 charged group of PtdIns(4,5)P2 and other anionic lipids, as shown in Fig. 6C.

All of the studies reported here were performed with overexpressed PMCA and with models. Nevertheless, they have provided useful information about the binding of PtdIns(4,5)P2 to PMCA and suggest new roles for PMCA in managing the Ca2+ signal. It has long been known that PMCA pumps Ca2+ out of cells and that PtdIns(4,5)P2 stimulates that activity. By using a PtdIns(4,5)P2 sensor we found that expression of PMCA protects PtdIns(4,5)P2 molecules from hydrolysis by PLC and that the protection operates by two mechanisms. One requires only an intact PMCA (no activity is required); the second requires a fully active PMCA. The presence of a buffering mechanism by which inactive PMCA acts suggests that PMCAs must bind PtdIns(4,5)P2 and create a PtdIns(4,5)P2 pool, which is probably in equilibrium with free PtdIns(4,5)P2 molecules in the plasma membrane. Hence, the retention of the PtdIns(4,5)P2 sensor in the plasma membrane upon expression of the inactive PMCA is due to the binding of the sensor to the free PtdIns(4,5)P2 molecules, which are maintained in equilibrium with the PtdIns(4,5)P2 bound by the PMCA. The role of such a PtdIns(4,5)P2-buffering mechanism has been described recently, as the MARCKS protein can regulate the activity of TRPC1 channel through buffering PtdIns(4,5)P2 molecules in the vicinity of the channel (Shi et al., 2013).

In general, cells express small amounts of PMCAs; red blood cells, for example, express only 35 copies per square micron so is much less abundant than plasma membrane PtdIns(4,5)P2 molecules [about 5000–10,000 per square micron (Suh and Hille, 2008)]. We suggest that buffering of PtdIns(4,5)P2 by the PMCAs might have a substantial impact on PtdIns(4,5)P2 signaling when a large amount of pump is being expressed at discrete locations, like the dendritic spines or the stereocilia of hair cells (Burette et al., 2010; Zhao et al., 2012). We must emphasize, however, that even a small amount of active PMCA could be protective if it is located near the PtdIns(4,5)P2 pool. The PMCA–PtdIns(4,5)P2 pool could have a great importance in PtdIns(4,5)P2-related processes by (1) preventing complete depletion of PtdIns(4,5)P2 and (2) encouraging the formation of extensive PtdIns(4,5)P2-rich areas in specific membrane locales where PMCA is abundant.

Calculation of the electrostatic potential at the surface of a PMCA4b model disclosed a positively charged electrostatic patch around the stalk region of the molecule that we called the blue collar. Molecular dynamics simulation experiments revealed four PtdIns(4,5)P2-binding pockets formed from the blue collar residues of PMCA4b. On the basis of these findings we propose that PtdIns(4,5)P2 interacts not only through non-specific electrostatic interactions with the previously described lipid-binding AL sequence (Brodin et al., 1992; Filoteo et al., 1992) but also through specific interactions by fitting its phosphatidylinositol head-group into binding pockets. Specific pocket binding of the PtdIns(4,5)P2 head is also used by other molecules. X-ray crystal structures of the Kir2.2 (Hansen et al., 2011) and GIRK2 K+ (Whorton and MacKinnon, 2011) channels have identified highly specific phosphatidylinositol-binding regions at an interface between the transmembrane domain and the cytoplasmic domains of these molecules. In addition, several phosphoinositide-binding signal molecules use structured domains to achieve specific recognition of different phosphoinositide head groups (Kutateladze, 2010).

Supplementary material Figs S4–S6 show the relationship of the human PMCA4b to other PMCAs and SERCA. Supplementary material Fig. S5 shows that the four genes that encode for human PMCA retain almost all of the residues that make up the blue collar. Of the residues that comprise the binding sites (highlighted in cyan), the only exceptions are R993 (missing in human PMCA3 and human PMCA1), R1052 (present only in human PMCA4) and K89 (missing in human PMCA3). This figure also shows that SERCA1a does not retain any corresponding residues, except for R438, R439 and K442. In many of the corresponding positions, SERCA1a has acidic residues instead of basic ones. These relationships indicate that the PtdIns(4,5)P2-binding sites of PMCA have special functions not shared by SERCA.

Supplementary material Fig. S6 shows that the PtdIns(4,5)P2-binding sites are conserved in PMCAs throughout the animal kingdom. From humans to fish and insects, the binding residues are almost always retained. However, in other organisms (plants and prokaryotes) PMCA-type molecules have very different structures not only in the blue collar region, but also in most other parts of the molecule. Supplementary material Fig. S4 shows the alignment of the whole SERCA and PMCA sequences, which were used for constructing the model.

The properties of PMCA reported here suggest that it might function as a unique PIPmodulin that mediates PtdIns(4,5)P2 clustering at specific locations. Given that PMCA binds PtdIns(4,5)P2 and provides a low Ca2+ milieu; it might form a ‘Ca2+-less PMCAPtdIns(4,5)P2 pool’ (CPPP) in the cell membrane. The known PIPmodulins, such as the myristoylated alanine-rich C-kinase substrate (MARCKS), growth-associated protein-43 (GAP43) and cytoskeleton-associated protein (CAP23), also sequester PtdIns(4,5)P2 molecules (Laux et al., 2000), but when they bind Ca2+-calmodulin these PIPmodulins release PtdIns(4,5)P2. In contrast, as is seen in Figs 1, 2, PMCA keeps PtdIns(4,5)P2 in the plasma membrane even in the presence of elevated Ca2+. PtdIns(4,5)P2 also enhances the activity of the PMCAs; this would be expected to reduce the Ca2+ concentration in the vicinity of the CPPP, reducing the activity of PLC and stabilizing the PMCA-bound PtdIns(4,5)P2 compartment.

Moreover, there are certain plasma membrane compartments with high PtdIns(4,5)P2 content where the members of the MARCKS family are not expressed and the putative PIPmodulin is likely to be PMCA. Three-dimensional simulations suggest that local sequestration of PtdIns(4,5)P2 in the spine heads of Purkinje neurons is responsible for the observed InsP3 signaling (Brown et al., 2008). MARCKS, GAP43 or CAP23 are not found in Purkinje cells (ConsoleBram et al., 1996; Ouimet et al., 1990) but PMCA2 is concentrated in the spine heads of Purkinje neurons (Burette et al., 2010). Another example where both PtdIns(4,5)P2 and PMCA are abundant in the same specific compartment is the stereocilia of hair cells in the inner ear. In these regions of the hair cells, ∼2000 PMCA2 molecules are gathered in a square micron (Yamoah et al., 1998). If we take the assumption that about 5000–10,000 PtdIns(4,5)P2 molecules are in a square micron of the plasma membrane we might assume that most of them are sequestered by the large amount of PMCA in the stereocilia. In good accordance with this hypothesis, recent studies have demonstrated that PtdIns(4,5)P2 is concentrated at the tips of stereocilia together with the PMCA2 protein (Zhao et al., 2012). Further experiments have demonstrated that a specific asymmetric distribution of PtdIns(4,5)P2 (Hirono et al., 2004) and PMCA2 (Kozel et al., 1998) is essential for the appropriate function of hair cells, underlining the importance of a putative CPPP in hearing.

In summary, we suggest that by protecting PtdIns(4,5)P2 molecules, PMCA can mediate the cellular concentrations of two messenger molecules, InsP3 and Ca2+, and, hence, might play a substantial role in the complex interplay between local PtdIns(4,5)P2 and Ca2+ signaling. In addition, by binding PtdIns(4,5)P2, PMCAs might affect many cellular functions regulated by PtdIns(4,5)P2, such as the activity of channels and pumps, remodeling of the actin cytoskeleton, endocytosis and migration. Finally, imbalances of the PtdIns(4,5)P2 and PtdIns(3,4,5)P3 phospholipids might cause defects in cell functions leading to cancer and other diseases. Therefore, controlling the level of these molecules by use of PMCAs might protect cells against such diseases.

Reagents

Fugene HD Transfection Reagent was obtained from Roche Applied Science (Mannheim, Germany). All other chemicals used were of reagent grade.

DNA constructs

The mammalian expression construct for PHPLCδ1–RFP was a kind gift from Péter Várnai (Semmelweis University, Hungary) and is as described previously (Várnai and Balla, 2008). pN1-GCaMP2 plasmid was a kind gift from Junichi Nakai, RIKEN Brain Science Institute, Saitama, Japan (Nakai and Ohkura, 2003). Generation of the pEGFP-PMCA2wb plasmid was described previously (Chicka and Strehler, 2003). The mCherry–PMCA2wb construct was created by replacing the EGFP fragment of the pEGFP-PMCA2wb plasmid with the mCherry coding sequence from the pmCherry-C1 vector (Clontech) using the AgeI-KpnI restriction sites. The pEGFP-PMCA4b-L1167–1169A plasmid was constructed as described previously (Antalffy et al., 2013). To create the mCherry-PMCA4b-L1167–1169A construct, site-directed mutagenesis, introducing the L1167–1169A triple mutation was performed on the mCherry-PMCA4b template plasmid (Antalffy et al., 2013) using the QuikChange II Site-Directed Mutagenesis Kit (Stratagene) with the following primers: forward, 5′-CTAAGTTTGGGACTAGGGTGGCAGCGGCGGATGGTGAGGTCACTCCATATGCC-3′; reverse, 5′-GGCATATGGAGTGACCTCACCATCCGCCGCTGCCACCCTAGTCCCAAACTTAG-3′. pEGFP-PMCA4b-D672E-L1167–1169A and mCherry-PMCA4b-D672E-L1167–1169A constructs were created by introducing the D672E point mutation to the PMCA4b-LA plasmids using the QuikChange II Site-Directed Mutagenesis Kit (Stratagene). PCR primers were as follows: forward, 5′-GGTGGGCATTGAGGAGCCTGTGCGCCCAGAG-3′; reverse, 5′-CTCTGGGCGCACAGGCTCCTCAATGCCCACC-3′.

sh-HeLa cell line

The sh-HeLa cell line was generated by using the PMCA4 shRNA plasmid (Santa Cruz Biotechnology, sc-42602-SH). HeLa cells were transfected with PMCA4 shRNA plasmid or with control shRNA plasmid-A (sc-108060) in a six-well plate by using shRNA plasmid transfection medium (sc-108062) and shRNA plasmid transfection reagent according to the manufacturer's protocol. At 48 hours after transfection cells were selected by puromycin dihydrochloride (2 µg/ml) (sc-108071) in DMEM for 2 weeks and medium was changed to fresh medium with puromycin every 2–3 days. After selection, cell clones were prepared using cloning rings. PMCA4b expression was tested by western blot analysis. From the most efficient clone, the sh-HeLa cell line was generated.

Cell culture and transfection

HeLa cells (obtained from ECACC) or sh-HeLa cells were grown in DMEM supplemented with 10% FBS, 100 U/ml penicillin, 100 µg/ml streptomycin and 2 mM glutamine under 5% CO2 at 37°C. At 1 day prior to transfection, cells were seeded into a Lab-TekTM II Chambered Slide (Nalge Nunc Int.) at a density of 4×104–6×104 cells. Transfection using the appropriate combination of plasmids, described in supplementary material Fig. S1, was carried out with FuGene HD (Roche) according to the protocol of the manufacturer. This procedure yielded about 60–70% transfected cells in the cell culture. Cells were inspected 24 hours after transfection.

Analysis of PtdIns(4,5)P2 translocation and Ca2+ signaling by confocal imaging

HeLa or sh-HeLa cells were seeded into a Lab-TekTM II Chambered Slide (Nalge Nunc Int.) and transfected with PHPLCδ1–RFP alone or together with GFP-tagged PMCA constructs for the PtdIns(4,5)P2 translocation experiments; or with GCaMP2 alone or together with mCherry-tagged PMCA constructs for Ca2+ imaging (supplementary material Fig. S1). Prior to the measurement of sensitivity of cells to ionomycin, the culture medium was replaced with Hank's buffered salt solution (HBSS) supplemented with 0.9 mM MgCl2, 2 mM CaCl2 and 20 mM Hepes (pH 7.4). Cells were sequentially exposed to 2 µM, 5 µM and 10 µM ionomycin at 0, 6 and 12 minutes. In order to study activation of the PLC pathway by G-protein-receptor-coupled agonists the culture medium was replaced with nominally Ca2+-free HBSS supplemented with 0.9 mM MgCl2, 100 µM EGTA, 100 µM CaCl2 and 20 mM Hepes pH 7.4. Cells were stimulated by the consecutive additions of 100 µM ATP and 100 µM histamine (at 0 and 2 minutes) in the absence of Ca2+ (InsP3-mediated signal). After 5 minutes incubation, the extracellular medium was replenished by 2 mM Ca2+ and the store-operated Ca2+ entry (SOCE signal) was monitored for 5 more minutes. Finally, the maximal response was induced by the addition of 10 µM ionomycin.

Images were taken with an Olympus IX-81 laser scanning confocal microscope and Fluoview FV500 software using an Olympus PLAPO 60× (1.4) oil immersion objective. For PHPLCδ1–RFP imaging, cells were excited at 543 nm and emission was collected above 560 nm. For GCaMP2 imaging, cells were excited at 488 nm and emission was collected between 505 and 535 nm. Regions of interest were selected in the cytosol, nuclei were omitted. GFP-tagged PMCAs were illuminated at 488 nm and emission between 505 and 535 nm was recorded. mCherry-tagged PMCAs were illuminated at 543 nm and emission above 560 nm was recorded. Images were acquired every 0.3–1.2 seconds, the z-resolution was 0.5 µm. Time-lapse sequences were recorded with the Fluoview Tiempo (v4.3) time course software at room temperature. The relative fluorescence was calculated as F/F0 (where F0 was the average initial fluorescence). GraphPad Prism4 software was used to analyze the experimental data.

Data analysis and statistics

Data presented in Fig. 4 are means of at least three independent experiments (±s.e.m.) and statistical significance was determined by Student's t-test. For statistical analysis of data in Fig. 2, cells were scored according to the ionomycin concentration at which full translocation of the PtdIns(4,5)P2 sensor occurred. Statistical significance of image scores was determined by the χ2 test. Statistical analyses were performed using Excel software (Microsoft) and GraphPad Prism 4 software (GraphPad Software Inc.).

Construction of a model of PMCA4b

The Ca2+ pump of rabbit sarcoplasmic reticulum SERCA1a has been crystallized at several stages of its catalytic cycle (Toyoshima et al., 2004; Moller et al., 2010) and has considerable resemblance to PMCA4b. Supplementary material Fig. S4 shows an alignment of these two pumps. Study of this figure shows that PMCA is longer than SERCA, the extra protein in PMCA being at the N- and C-terminal domains and in a long insert marked as I. Not including these three regions, the PMCA sequence contained 31% residues identical to SERCA, as well as numerous similar residues. In addition to sequence identities, proper positioning of the hydrophobic transmembrane regions assures an accurate alignment (PMCA and SERCA have ten transmembrane regions each).

The structures of the SERCA AMPPCP-Ca2 (PDB 1T5S and 1VFP) and of the N-domains of the Na+/K+ ATPase (PDB 1MO7 and 1MO8) were aligned with PMCA4b and a model of PMCAE1 was made. The alignment of PMCA4b with SERCA1a was the same as is shown in supplementary material Fig. S4. Modeller 8v2 was then used to create the PMCA model covering residues 1–1058. It has been reported that F1094 crosslinks to the peptide C537-ALLGFVT in the N domain (Carafoli et al., 1992; Falchetto et al., 1991; Falchetto et al., 1992). Accordingly, residues 1059–1094 (part of the C-terminus) were added next, with F1094 placed near C537. This caused the C-terminus to go away from the membrane interface, up over the cytoplasmic top of PMCA. The remainder of the C-terminus (residues 1095–1205) was modeled using Tasser (Roy et al., 2010) and attached to the end of PMCA.

In general, the subsequent work was performed using CHARMM (Brooks et al., 1983; http://www.charmm.org/) and CHARMM-GUI (Jo et al., 2008; http://www.charmm-gui.org/). The PMCA was put into a hexagonal box with a lipid bilayer, TIP3 water and enough K+ and Cl ions to make a neutral system with 0.15 M salt. The lipid composition was chosen to approximate that of the plasma membrane of 3T3 fibroblasts (Pankov et al., 2006). The outer leaflet of the membrane contained 17 cholesterol molecules (CHL-13%), 82 of palmitoyloleoylphosphatidylcholine (POPC, 63%) and 31 of stearoylarachidonylphosphatidylcholine (SAPC, 24% representing sphingomyelin). The inner (cytoplasmic) leaflet contained 17 cholesterol molecules (CHL, 13%), 31 of palmitoyloleoylphosphatidylcholine (POPC, 24%), 4 of palmitoyloleoylphoshatidic acid (PA, 3%), 42 of palmitoyloleoylphosphatidylethanolamine (POPE-32%), 16 of palmitoyloleylphosphatidylserine (POPS, 13%) 15 of stearoylarachidonylphosphatidylinositol (PI, 12%) and 5 of stearoylarachidonylphosphatidylinositolbisphosphate [PtdIns(4,5)P2, 3%].

After minimization and heating, the molecular dynamics studies were performed on this hexagonal box containing the 1205 residues of PMCA, 250 molecules of membrane lipid, 32074 TIP3 water molecules, 132 K+ ions and 63 Cl ions at 310° K. The box was 104 Å on each side and its initial length was 155 Å. The early stages of the dynamics run were performed at constant pressure and temperature; the dimensions of the box were allowed to vary to keep the pressure constant. After the initial fluctuations in the dimensions had diminished, the dimensions of the box were fixed and the run carried on at constant volume and temperature.

The electrostatic and van der Waals interaction energies between various atoms were calculated by CHARMM. These energies are calculated directly from the physical formulas, and so are the total energies rather than the energies with respect to a standard reference state. They also lack a PV term, but this does not affect the validity of the results, because the electrostatic energy is clearly the dominant term, and the one that changes the most.

In order to keep the molecular dynamics run times to reasonable values, a large portion of the atoms in the model were fixed in space. After the initial phase was running smoothly, all atoms in the cytoplasmic half of the prism were fixed, as well as the backbone of the protein, the side chains and waters in the extracellular part of the prism. This left the side chains of the collar, the lipids of the membrane and nearby waters free to move.

Also because of time limitations, it was necessary to place the PtdIns(4,5)P2 molecules near PMCA in order to observe the interactions within the 10 nanoseconds of simulation. The five PtdIns(4,5)P2 molecules were placed at random starting positions near the PMCA in order to sample all the possible binding sites. 15 runs of 10 nanoseconds each were performed, and the interactions of lipid with the protein observed in each.

Some variations from these conditions occurred. In the first two runs, the PtdIns(4,5)P2 was not placed near PMCA, and only one interaction was observed. In some of the early runs, the side chains of the blue collar were not allowed to move.

We are grateful to Krisztina Lór for excellent technical assistance, to Junichi Nakai (RIKEN Brain Science Institute, Saitama, Japan) for providing the pN1-GCaMP2 plasmid, to Péter Várnai (Semmelweis University, Hungary) for the PHPLCδ1–RFP plasmid and to Robert Katona (Institute of Genetics, Biological Research Center of the Hungarian Academy of Sciences) for the pcDNA3-mCherry plasmid. The SB-CAG-Amaxa GFP vector was a gift of Tamás Orbán (Institute of Molecular Pharmacology, Research Centre for Natural Sciences, Hungarian Academy of Sciences).

Author contributions

J.T.P., R.P. and A.E. conceived and designed the experiments, and wrote the manuscript; R.P. performed the majority of the experiments, and analyzed the data; K.P. did the Ca2+ signaling experiments; J.T.P. developed the model and ran the molecular dynamics simulations; L.H. and K.V. generated and validated the PMCA plasmid constructs; A.E. supervised and coordinated the project.

Funding

This work was supported by grants from the Hungarian Scientific Research Fund (OTKA) [grant numbers CK 80283 and K 101064 to A.E.]; and Hungarian National Development Agency [grant number TRANSRAT KMR_12-1-2012-0112 to K.P.].

Adamo
H. P.
,
Filoteo
A. G.
,
Enyedi
A.
,
Penniston
J. T.
(
1995
).
Mutants in the putative nucleotide-binding region of the plasma membrane Ca2+-pump. A reduction in activity due to slow dephosphorylation.
J. Biol. Chem.
270
,
30111
30114
.
Antalffy
G.
,
Pászty
K.
,
Varga
K.
,
Hegedűs
L.
,
Enyedi
A.
,
Padányi
R.
(
2013
).
A C-terminal di-leucine motif controls plasma membrane expression of PMCA4b.
Biochim. Biophys. Acta
1833
,
2561
2572
.
Balla
T.
(
2005
).
Inositol-lipid binding motifs: signal integrators through protein-lipid and protein-protein interactions.
J. Cell Sci.
118
,
2093
2104
.
Bautista
D. M.
,
Lewis
R. S.
(
2004
).
Modulation of plasma membrane calcium-ATPase activity by local calcium microdomains near CRAC channels in human T cells.
J. Physiol.
556
,
805
817
.
Bautista
D. M.
,
Hoth
M.
,
Lewis
R. S.
(
2002
).
Enhancement of calcium signalling dynamics and stability by delayed modulation of the plasma-membrane calcium-ATPase in human T cells.
J. Physiol.
541
,
877
894
.
Berridge
M. J.
(
2009
).
Inositol trisphosphate and calcium signalling mechanisms.
Biochim. Biophys. Acta
1793
,
933
940
.
Bird
G. S.
,
DeHaven
W. I.
,
Smyth
J. T.
,
Putney
J. W.
 Jr
(
2008
).
Methods for studying store-operated calcium entry.
Methods
46
,
204
212
.
Brini
M.
,
Carafoli
E.
(
2009
).
Calcium pumps in health and disease.
Physiol. Rev.
89
,
1341
1378
.
Brini
M.
,
Di Leva
F.
,
Ortega
C. K.
,
Domi
T.
,
Ottolini
D.
,
Leonardi
E.
,
Tosatto
S. C. E.
,
Carafoli
E.
(
2010
).
Deletions and mutations in the acidic lipid-binding region of the plasma membrane Ca2+ pump: a study on different splicing variants of isoform 2.
J. Biol. Chem.
285
,
30779
30791
.
Brodin
P.
,
Falchetto
R.
,
Vorherr
T.
,
Carafoli
E.
(
1992
).
Identification of two domains which mediate the binding of activating phospholipids to the plasma-membrane Ca2+ pump.
Eur. J. Biochem.
204
,
939
946
.
Brooks
B. R.
,
Bruccoleri
R. E.
,
Olafson
B. D.
,
States
D. J.
,
Swaminathan
S.
,
Karplus
M.
(
1983
).
Charmm - a Program for Macromolecular Energy, Minimization, and Dynamics Calculations.
J. Comput. Chem.
4
,
187
217
.
Brown
S. A.
,
Morgan
F.
,
Watras
J.
,
Loew
L. M.
(
2008
).
Analysis of phosphatidylinositol-4,5-bisphosphate signaling in cerebellar Purkinje spines.
Biophys. J.
95
,
1795
1812
.
Bublitz
M.
,
Poulsen
H.
,
Morth
J. P.
,
Nissen
P.
(
2010
).
In and out of the cation pumps: P-type ATPase structure revisited.
Curr. Opin. Struct. Biol.
20
,
431
439
.
Bublitz
M.
,
Morth
J. P.
,
Nissen
P.
(
2011
).
P-type ATPases at a glance.
J. Cell Sci.
124
,
2515
2519
.
Burette
A. C.
,
Strehler
E. E.
,
Weinberg
R. J.
(
2010
).
A plasma membrane Ca2+ ATPase isoform at the postsynaptic density.
Neuroscience
169
,
987
993
.
Carafoli
E.
,
Kessler
F.
,
Falchetto
R.
,
Heim
R.
,
Quadroni
M.
,
Krebs
J.
,
Strehler
E. E.
,
Vorherr
T.
(
1992
).
The molecular basis of the modulation of the plasma membrane calcium pump by calmodulin.
Ann. N. Y. Acad. Sci.
671
,
58
68, discussion 68-69
.
Caride
A. J.
,
Filoteo
A. G.
,
Penheiter
A. R.
,
Pászty
K.
,
Enyedi
A.
,
Penniston
J. T.
(
2001
).
Delayed activation of the plasma membrane calcium pump by a sudden increase in Ca2+: fast pumps reside in fast cells.
Cell Calcium
30
,
49
57
.
Chicka
M. C.
,
Strehler
E. E.
(
2003
).
Alternative splicing of the first intracellular loop of plasma membrane Ca2+-ATPase isoform 2 alters its membrane targeting.
J. Biol. Chem.
278
,
18464
18470
.
Choquette
D.
,
Hakim
G.
,
Filoteo
A. G.
,
Plishker
G. A.
,
Bostwick
J. R.
,
Penniston
J. T.
(
1984
).
Regulation of plasma membrane Ca2+ ATPases by lipids of the phosphatidylinositol cycle.
Biochem. Biophys. Res. Commun.
125
,
908
915
.
Console-Bram
L. M.
,
Fitzpatrick-McElligott
S. G.
,
McElligott
J. G.
(
1996
).
Distribution of GAP-43 mRNA in the immature and adult cerebellum: a role for GAP-43 in cerebellar development and neuroplasticity.
Brain Res. Dev. Brain Res.
95
,
97
106
.
Dickson
E. J.
,
Falkenburger
B. H.
,
Hille
B.
(
2013
).
Quantitative properties and receptor reserve of the IP(3) and calcium branch of G(q)-coupled receptor signaling.
J. Gen. Physiol.
141
,
521
535
.
Enyedi
A.
,
Flura
M.
,
Sarkadi
B.
,
Gardos
G.
,
Carafoli
E.
(
1987
).
The maximal velocity and the calcium affinity of the red cell calcium pump may be regulated independently.
J. Biol. Chem.
262
,
6425
6430
.
Falchetto
R.
,
Vorherr
T.
,
Brunner
J.
,
Carafoli
E.
(
1991
).
The plasma membrane Ca2+ pump contains a site that interacts with its calmodulin-binding domain.
J. Biol. Chem.
266
,
2930
2936
.
Falchetto
R.
,
Vorherr
T.
,
Carafoli
E.
(
1992
).
The calmodulin-binding site of the plasma membrane Ca2+ pump interacts with the transduction domain of the enzyme.
Protein Sci.
1
,
1613
1621
.
Filoteo
A. G.
,
Enyedi
A.
,
Penniston
J. T.
(
1992
).
The lipid-binding peptide from the plasma membrane Ca2+ pump binds calmodulin, and the primary calmodulin-binding domain interacts with lipid.
J. Biol. Chem.
267
,
11800
11805
.
Gamper
N.
,
Shapiro
M. S.
(
2007
).
Target-specific PIP(2) signalling: how might it work?
J. Physiol.
582
,
967
975
.
Hansen
S. B.
,
Tao
X.
,
MacKinnon
R.
(
2011
).
Structural basis of PIP2 activation of the classical inward rectifier K+ channel Kir2.2.
Nature
477
,
495
498
.
Hirono
M.
,
Denis
C. S.
,
Richardson
G. P.
,
Gillespie
P. G.
(
2004
).
Hair cells require phosphatidylinositol 4,5-bisphosphate for mechanical transduction and adaptation.
Neuron
44
,
309
320
.
Hirose
K.
,
Kadowaki
S.
,
Tanabe
M.
,
Takeshima
H.
,
Iino
M.
(
1999
).
Spatiotemporal dynamics of inositol 1,4,5-trisphosphate that underlies complex Ca2+ mobilization patterns.
Science
284
,
1527
1530
.
Jo
S.
,
Kim
T.
,
Iyer
V. G.
,
Im
W.
(
2008
).
Software news and updates - CHARNIM-GUI: A web-based grraphical user interface for CHARMM.
J. Comput. Chem.
29
,
1859
1865
.
Koch
M.
,
Holt
M.
(
2012
).
Coupling exo- and endocytosis: An essential role for PIP2 at the synapse.
Biochim. Biophys. Acta.
1821
,
1114
1132
.
Kozel
P. J.
,
Friedman
R. A.
,
Erway
L. C.
,
Yamoah
E. N.
,
Liu
L. H.
,
Riddle
T.
,
Duffy
J. J.
,
Doetschman
T.
,
Miller
M. L.
,
Cardell
E. L.
 et al. (
1998
).
Balance and hearing deficits in mice with a null mutation in the gene encoding plasma membrane Ca2+-ATPase isoform 2.
J. Biol. Chem.
273
,
18693
18696
.
Kutateladze
T. G.
(
2010
).
Translation of the phosphoinositide code by PI effectors.
Nat. Chem. Biol.
6
,
507
513
.
Kwiatkowska
K.
(
2010
).
One lipid, multiple functions: how various pools of PI(4,5)P(2) are created in the plasma membrane.
Cell. Mol. Life Sci.
67
,
3927
3946
.
Lanier
L. M.
,
Gertler
F. B.
(
2000
).
Actin cytoskeleton: thinking globally, actin' locally.
Curr. Biol.
10
,
R655
R657
.
Laux
T.
,
Fukami
K.
,
Thelen
M.
,
Golub
T.
,
Frey
D.
,
Caroni
P.
(
2000
).
GAP43, MARCKS, and CAP23 modulate PI(4,5)P(2) at plasmalemmal rafts, and regulate cell cortex actin dynamics through a common mechanism.
J. Cell Biol.
149
,
1455
1472
.
Lehotsky
J.
,
Raeymaekers
L.
,
Missiaen
L.
,
Wuytack
F.
,
De Smedt
H.
,
Casteels
R.
(
1992
).
Stimulation of the catalytic cycle of the Ca2+ pump of porcine plasma-membranes by negatively charged phospholipids.
Biochim. Biophys. Acta
1105
,
118
124
.
Lemonnier
L.
,
Trebak
M.
,
Putney
J. W.
 Jr
(
2008
).
Complex regulation of the TRPC3, 6 and 7 channel subfamily by diacylglycerol and phosphatidylinositol-4,5-bisphosphate.
Cell Calcium
43
,
506
514
.
McLaughlin
S.
,
Murray
D.
(
2005
).
Plasma membrane phosphoinositide organization by protein electrostatics.
Nature
438
,
605
611
.
McLaughlin
S.
,
Wang
J. Y.
,
Gambhir
A.
,
Murray
D.
(
2002
).
PIP(2) and proteins: interactions, organization, and information flow.
Annu. Rev. Biophys. Biomol. Struct.
31
,
151
175
.
Michailidis
I. E.
,
Helton
T. D.
,
Petrou
V. I.
,
Mirshahi
T.
,
Ehlers
M. D.
,
Logothetis
D. E.
(
2007
).
Phosphatidylinositol-4,5-bisphosphate regulates NMDA receptor activity through alpha-actinin.
J. Neurosci.
27
,
5523
5532
.
Michailidis
I. E.
,
Rusinova
R.
,
Georgakopoulos
A.
,
Chen
Y. B.
,
Iyengar
R.
,
Robakis
N. K.
,
Logothetis
D. E.
,
Baki
L.
(
2011
).
Phosphatidylinositol-4,5-bisphosphate regulates epidermal growth factor receptor activation.
Pflugers Archiv-European Journal of Physiology
461
,
387
397
.
Missiaen
L.
,
Raeymaekers
L.
,
Wuytack
F.
,
Vrolix
M.
,
de Smedt
H.
,
Casteels
R.
(
1989a
).
Phospholipid-protein interactions of the plasma-membrane Ca2+-transporting ATPase. Evidence for a tissue-dependent functional difference.
Biochem. J.
263
,
687
694
.
Missiaen
L.
,
Wuytack
F.
,
Raeymaekers
L.
,
De Smedt
H.
,
Casteels
R.
(
1989b
).
Polyamines and neomycin inhibit the purified plasma-membrane Ca2+ pump by interacting with associated polyphosphoinositides.
Biochem. J.
261
,
1055
1058
.
Moller
J. V.
,
Olesen
C.
,
Winther
A. M.
,
Nissen
P.
(
2010
).
The sarcoplasmic Ca2+-ATPase: design of a perfect chemi-osmotic pump.
Q. Rev. Biophys.
43
,
501
566
.
Moss
S. E.
(
2012
).
How actin gets the PIP.
Sci. Signal.
5
,
pe7
.
Mundell
S.
,
Kelly
E.
(
2011
).
Adenosine receptor desensitization and trafficking.
Biochim. Biophys. Acta
1808
,
1319
1328
.
Nakai
J.
,
Ohkura
M.
(
2003
).
Probing calcium ions with biosensors.
Biotechnol. Genet. Eng. Rev.
20
,
3
21
.
Ouimet
C. C.
,
Wang
J. K. T.
,
Walaas
S. I.
,
Albert
K. A.
,
Greengard
P.
(
1990
).
Localization of the MARCKS (87 kDa) protein, a major specific substrate for protein kinase C, in rat brain.
J. Neurosci.
10
,
1683
1698
.
Pankov
R.
,
Markovska
T.
,
Antonov
P.
,
Ivanova
L.
,
Momchilova
A.
(
2006
).
The plasma membrane lipid composition affects fusion between cells and model membranes.
Chem. Biol. Interact.
164
,
167
173
.
Pinto
F. T.
,
Adamo
H. P.
(
2002
).
Deletions in the acidic lipid-binding region of the plasma membrane Ca2+ pump. A mutant with high affinity for Ca2+ resembling the acidic lipid-activated enzyme.
J. Biol. Chem.
277
,
12784
12789
.
Ritchie
M. F.
,
Samakai
E.
,
Soboloff
J.
(
2012
).
STIM1 is required for attenuation of PMCA-mediated Ca2+ clearance during T-cell activation.
EMBO J.
31
,
1123
1133
.
Roy
A.
,
Kucukural
A.
,
Zhang
Y.
(
2010
).
I-TASSER: a unified platform for automated protein structure and function prediction.
Nat. Protoc.
5
,
725
738
.
Schill
N. J.
,
Anderson
R. A.
(
2009
).
Out, in and back again: PtdIns(4,5)P(2) regulates cadherin trafficking in epithelial morphogenesis.
Biochem. J.
418
,
247
260
.
Shi
J.
,
Birnbaumer
L.
,
Large
W. A.
,
Albert
A. P.
(
2013
).
Myristoylated alanine-rich C kinase substrate coordinates native TRPC1 channel activation by phosphatidylinositol 4,5-bisphosphate and protein kinase C in vascular smooth muscle.
FASEB J
Snitsarev
V. A.
,
Taylor
C. W.
(
1999
).
Overshooting cytosolic Ca2+ signals evoked by capacitative Ca2+ entry result from delayed stimulation of a plasma membrane Ca2+ pump.
Cell Calcium
25
,
409
417
.
Strehler
E. E.
,
Zacharias
D. A.
(
2001
).
Role of alternative splicing in generating isoform diversity among plasma membrane calcium pumps.
Physiol. Rev.
81
,
21
50
.
Strehler
E. E.
,
Caride
A. J.
,
Filoteo
A. G.
,
Xiong
Y.
,
Penniston
J. T.
,
Enyedi
A.
(
2007
).
Plasma membrane Ca2+ ATPases as dynamic regulators of cellular calcium handling.
Ann. N. Y. Acad. Sci.
1099
,
226
236
.
Suh
B. C.
,
Hille
B.
(
2008
).
PIP2 is a necessary cofactor for ion channel function: how and why?
Annual Review of Biophysics
37
,
175
195
.
Szentpetery
Z.
,
Balla
A.
,
Kim
Y. J.
,
Lemmon
M. A.
,
Balla
T.
(
2009
).
Live cell imaging with protein domains capable of recognizing phosphatidylinositol 4,5-bisphosphate; a comparative study.
BMC Cell Biol.
10
,
67
.
Toyoshima
C.
,
Nakasako
M.
,
Nomura
H.
,
Ogawa
H.
(
2000
).
Crystal structure of the calcium pump of sarcoplasmic reticulum at 2.6 A resolution.
Nature
405
,
647
655
.
Toyoshima
C.
,
Nomura
H.
,
Tsuda
T.
(
2004
).
Lumenal gating mechanism revealed in calcium pump crystal structures with phosphate analogues.
Nature
432
,
361
368
.
Trebak
M.
,
Lemonnier
L.
,
DeHaven
W. I.
,
Wedel
B. J.
,
Bird
G. S.
,
Putney
J. W.
 Jr
(
2009
).
Complex functions of phosphatidylinositol 4,5-bisphosphate in regulation of TRPC5 cation channels.
Pflugers Arch.
457
,
757
769
.
van den Bogaart
G.
,
Meyenberg
K.
,
Risselada
H. J.
,
Amin
H.
,
Willig
K. I.
,
Hubrich
B. E.
,
Dier
M.
,
Hell
S. W.
,
Grubmüller
H.
,
Diederichsen
U.
 et al. (
2011
).
Membrane protein sequestering by ionic protein-lipid interactions.
Nature
479
,
552
555
.
van der Wal
J.
,
Habets
R.
,
Várnai
P.
,
Balla
T.
,
Jalink
K.
(
2001
).
Monitoring agonist-induced phospholipase C activation in live cells by fluorescence resonance energy transfer.
J. Biol. Chem.
276
,
15337
15344
.
Várnai
P.
,
Balla
T.
(
2008
).
Live cell imaging of phosphoinositides with expressed inositide binding protein domains.
Methods
46
,
167
176
.
Verma
A. K.
,
Filoteo
A. G.
,
Stanford
D. R.
,
Wieben
E. D.
,
Penniston
J. T.
,
Strehler
E. E.
,
Fischer
R.
,
Heim
R.
,
Vogel
G.
,
Mathews
S.
 et al. (
1988
).
Complete primary structure of a human plasma membrane Ca2+ pump.
J. Biol. Chem.
263
,
14152
14159
.
Whorton
M. R.
,
MacKinnon
R.
(
2011
).
Crystal structure of the mammalian GIRK2 K+ channel and gating regulation by G proteins, PIP2, and sodium.
Cell
147
,
199
208
.
Yamoah
E. N.
,
Lumpkin
E. A.
,
Dumont
R. A.
,
Smith
P. J. S.
,
Hudspeth
A. J.
,
Gillespie
P. G.
(
1998
).
Plasma membrane Ca2+-ATPase extrudes Ca2+ from hair cell stereocilia.
J. Neurosci.
18
,
610
624
.
Zhao
Q.
,
Logothetis
D. E.
,
Séguéla
P.
(
2007
).
Regulation of ATP-gated P2X receptors by phosphoinositides.
Pflugers Archiv-European Journal of Physiology
455
,
181
185
.
Zhao
H. Y.
,
Williams
D. E.
,
Shin
J. B.
,
Brügger
B.
,
Gillespie
P. G.
(
2012
).
Large membrane domains in hair bundles specify spatially constricted radixin activation.
J. Neurosci.
32
,
4600
4609
.
Zvaritch
E.
,
James
P.
,
Vorherr
T.
,
Falchetto
R.
,
Modyanov
N.
,
Carafoli
E.
(
1990
).
Mapping of functional domains in the plasma membrane Ca2+ pump using trypsin proteolysis.
Biochemistry
29
,
8070
8076
.

Competing interests

The authors declare no competing interests.

Supplementary information