Elucidating mechanisms by which Ca2+ signals are generated by monocytes is important for understanding monocyte function in health and disease. We have investigated mechanisms underlying Ca2+ signals generated following disruption of lysosomes by exposure to the cathepsin C substrate glycyl-L-phenylalanine-β-napthylamide (GPN). Exposure to 0.2 mM GPN resulted in robust increases in the intracellular Ca2+ concentration ([Ca2+]i) in the absence of extracellular Ca2+. The response was antagonised by thapsigargin and evoked capacitative Ca2+ entry. Dantrolene-sensitive Ca2+ responses were observed at higher concentrations of GPN (0.4 mM) but not at 0.2 mM. Strikingly, GPN-evoked Ca2+ responses and β-hexosaminidase secretion were inhibited by the ATPase/ADPase apyrase. Simultaneous measurement of [Ca2+]i and extracellular ATP revealed a concomitant secretion of ATP during GPN-evoked Ca2+ signalling. Furthermore, the ability of GPN to raise [Ca2+]i was inhibited by P2Y receptor antagonists or by inhibiting vesicular exocytosis with N-ethylmaleimide (NEM). NEM treatment was associated with an inability of GPN to trigger ATP secretion, a drop in baseline [Ca2+]i and reduction in extracellular ATP concentration. Antagonism of purinergic signalling also caused a reduction in baseline [Ca2+]i. In summary, these data suggest that P2Y receptor activation contributes significantly to GPN-evoked Ca2+ signalling, and that constitutive secretion of lysosomal ATP is a major determinant of Ca2+ homeostasis in monocytes. Lysosomal Ca2+ stores can communicate with ER Ca2+ stores either directly through activation of ryanodine receptors, or indirectly through release of ATP and engagement of P2Y receptors.

Monocytes play an important role in immune defence and the development of chronic inflammatory diseases including atherosclerosis. Changes in cytosolic calcium concentration ([Ca2+]i) constitute a critical requirement for monocyte function including response to pro-inflammatory molecules including chemokines (Gouwy et al., 2008), bacterial lipopolysaccharide (McLeish et al., 1989), chemotaxis (Badolato et al., 1995), phagocytosis (Hishikawa et al., 1991) and cytokine production (MacIntyre et al., 1991). Monocyte calcium dynamics are altered in pathological conditions including sepsis (Shin et al., 1997), Leishmania infection (Olivier et al., 1992) and foam cell formation in atherosclerosis (Deng et al., 2005). Understanding mechanisms by which calcium signals are generated and shaped is therefore important for understanding how monocytes function in health and disease.

In resting cells [Ca2+]i is tightly buffered (typically 100 nM), but it can rise dramatically in response to extracellular cues including hormones and stressors (Berridge et al., 2000). When a calcium signal is generated, calcium enters the cytoplasm through plasma membrane entry mechanisms including translocators (i.e. Na+/Ca2+ exchange) and capacitative Ca2+-entry mechanisms, and/or through release of calcium stored within intracellular compartments (Pozzan et al., 1994; Clapham, 2007). The archetypal calcium store in animal cells is the endoplasmic reticulum (ER). Calcium is pumped into the ER by the thapsigargin-sensitive Ca2+-ATPase SERCA and released through activation of two distinct receptor classes located on the store: (i) inositol 1,4,5-trisphosphate [Ins(1,4,5)P3] receptors and (ii) ryanodine receptors (RyR). Ins(1,4,5)P3 formation and mediated calcium release are linked to the activation of G-protein-coupled receptors (GPCRs; specifically Gq), conveying extracellular signals from stressors and hormones (Berridge et al., 2000). Ryanodine receptors are widely expressed in muscle and non-muscle cell types, including leukocytes (Hosoi et al., 2001), are activated by increased [Ca2+]i and facilitate calcium-induced calcium release of the ER store. Activation of ER ryanodine receptors can aid amplification of Ca2+ signals generated by the ER but also other potential stores.

It has long been recognised that calcium is stored in acidic organelles distinct from the classical pool of ER calcium. Acidic stores include lysosomes (Lloyd-Evans et al., 2008; Churchill et al., 2002), secretory lysosomes (Holmsen and Weiss, 1979), acidocalcisomes (Luo et al., 2004) and vacuoles (Patel and Docampo, 2010). Moreover, recent studies have demonstrated that such pools are also mobilisable (Churchill et al., 2002; Lange et al., 2009). Leukocyte lysosomes play a key role in immunity and disease (Holt et al., 2006) by inactivating pathogenic organisms, processing antigens for presentation (Mullins and Bonifacino, 2001) and secreting pro-inflammatory molecules (Andrei et al., 2004). Lysosomal calcium stores have been identified in diverse cell types including endothelium (Srinivas et al., 2002), epithelium (Haller et al., 1996), macrophage (Christensen et al., 2002), mesenchymal stem cells (Lee et al., 2010) and vascular smooth muscle (Zhang et al., 2010), though their existence in monocytes remains elusive. In this study we sought evidence for functional lysosomal calcium stores in human monocytic cells, their interaction with the ER calcium pool, and explore their contribution to calcium homeostasis and monocyte function. We employ the use of glycyl-L-phenylalanine-β-napthylamide (GPN), a cathepsin C substrate, used extensively to selectively disrupt lysosomes in various cell types (Berg et al., 1994; Haller et al., 1996; Srinivas et al., 2002; Dickinson et al., 2010).

Interaction between GPN-sensitive and thapsigargin-sensitive calcium stores in monocytes

The lysosomotropic agent glycyl-L-phenylalanine-β-napthylamide (GPN) is suggested to raise cytoplasmic [Ca2+]i by stimulating the osmotic lysis of lysosomes and subsequent release of their Ca2+ content. In THP-1 monocytes, application of GPN evoked a transient elevation in [Ca2+]i which decayed to baseline over several minutes in the absence of extracellular calcium (Fig. 1A). GPN treatment resulted in a loss of Lysotracker staining in pre-stained cells, suggesting GPN is capable of disrupting lysosomes in THP-1 monocytes as observed in other cell types (Fig. 1B,D). Application of the ER calcium store inhibitor thapsigargin also evoked an increase in [Ca2+]i which was more sustained and approximately 3-fold greater in magnitude to the GPN-evoked response (Fig. 1C,D). Despite the larger magnitude of change in cytosolic calcium evoked by thapsigargin the initial rate of change was greater with GPN, 0.5±0.2 and 1.8±0.3 Ca2+ F(%) (n = 4-15; P<0.05) for thapsigargin and GPN, respectively. Thapsigargin was also capable of discharging Lysotracker staining (Fig. 1B). GPN was unable to increase calcium following thapsigargin treatment (Fig. 1C,D), suggesting an interaction between ER and lysosomal compartments in monocytes. The interaction could be explained if a direct interaction existed whereby GPN required ER calcium stores to raise [Ca2+]i. To test this, we explored the ability of GPN to evoke a ‘calcium add-back’ response, characteristic of ER calcium store depletion and capacitive calcium entry at the plasma membrane, observed in other leukocytes. 0.2 mM GPN raised [Ca2+]i in the absence of extracellular calcium, addition of 1.5 mM CaCl2 induced a large Ca2+ influx (Fig. 1E) which was not observed in the absence of GPN (data not shown). This data suggests GPN does cause release of calcium stored within the ER. One possible explanation for this observation is that the osmotic lysis and subsequent release of lysosomal calcium induced by GPN, could trigger release of ER calcium via a calcium-induced calcium release mechanism. We employed the ryanodine receptor antagonist dantrolene to inhibit calcium-induced calcium release. At concentrations of GPN used throughout this study and by others to disrupt lysosomes i.e. 0.2 mM, dantrolene had no effect on the ability of GPN to raise [Ca2+]i. However, increasing GPN to 0.4 mM evoked a more substantial increase in [Ca2+]i (Fig. 1F,G) which was inhibited by dantrolene (Fig. 1F), suggesting the increase in [Ca2+]i evoked by 0.4 mM but not 0.2 mM GPN can trigger ryanodine receptor activation and release of ER calcium. Moreover, this data suggests 0.2 mM GPN releases ER calcium via a mechanism other than calcium-induced calcium release. This data suggests that depletion of the ER can disrupt lysosomes and therefore can explain why GPN-evoked increase in [Ca2+]i is abolished with thapsigargin pre-treatment (Fig. 1C).

Fig. 1.

Interaction between lysosomal and ER calcium stores in human monocytes. (A) The lysosomotropic agent GPN transiently mobilises [Ca2+]i in the absence of external Ca2+, measured in Fluo-4 loaded THP-1 cells (n = 15). (B) Lysotracker staining (100 nM, 20 mins) of lysosomes can be discharged following exposure to GPN or thapsigargin (Tg; 5 µM; n = 4). Scale bar, 20 µm. (C) Thapsigargin-evoked [Ca2+]i mobilisation abolishes subsequent Ca2+ response to GPN (n = 5). (D) Comparison of peak Ca2+ responses to GPN in the presence and absence of thapsigargin (5 µM), or to thapsigargin alone (n = 5). (E) GPN evokes a ‘calcium add-back response’ following addition of 1.5 mM external CaCl2 after GPN (n = 4). (F) A higher concentration of GPN (0.4 vs 0.2 mM) evokes an enhanced peak Ca2+ response through engagement of ryanodine receptors. Response to 0.2 and 0.4 mM GPN are shown in control cells (black line) and in cells treated with dantrolene (20 µM, 30 mins; grey line). (G) The effect of the different concentrations of dantrolene on GPN-evoked Ca2+ peak calcium response (n = 5; *P<0.01). All experiments are performed in the absence of external Ca2+ unless otherwise stated. GPN was used at 0.2 mM unless stated otherwise.

Fig. 1.

Interaction between lysosomal and ER calcium stores in human monocytes. (A) The lysosomotropic agent GPN transiently mobilises [Ca2+]i in the absence of external Ca2+, measured in Fluo-4 loaded THP-1 cells (n = 15). (B) Lysotracker staining (100 nM, 20 mins) of lysosomes can be discharged following exposure to GPN or thapsigargin (Tg; 5 µM; n = 4). Scale bar, 20 µm. (C) Thapsigargin-evoked [Ca2+]i mobilisation abolishes subsequent Ca2+ response to GPN (n = 5). (D) Comparison of peak Ca2+ responses to GPN in the presence and absence of thapsigargin (5 µM), or to thapsigargin alone (n = 5). (E) GPN evokes a ‘calcium add-back response’ following addition of 1.5 mM external CaCl2 after GPN (n = 4). (F) A higher concentration of GPN (0.4 vs 0.2 mM) evokes an enhanced peak Ca2+ response through engagement of ryanodine receptors. Response to 0.2 and 0.4 mM GPN are shown in control cells (black line) and in cells treated with dantrolene (20 µM, 30 mins; grey line). (G) The effect of the different concentrations of dantrolene on GPN-evoked Ca2+ peak calcium response (n = 5; *P<0.01). All experiments are performed in the absence of external Ca2+ unless otherwise stated. GPN was used at 0.2 mM unless stated otherwise.

GPN and thapsigargin triggers lysosomes exocytosis in monocytes

As exocytosis of lysosomes has been shown to be stimulated by an increase in cytoplasmic [Ca2+]i in other leukocytes (Colvin et al., 2010; Chen et al., 2010), we hypothesised that an interaction between the ER and lysosomes could exist in monocytes whereby ER derived calcium promoted exocytosis of lysosomes. Moreover, this could account for the loss of ability of GPN to raise [Ca2+]i following thapsigargin treatment (Fig. 1C), as intact lysosomes have already been discharged. To examine lysosome exocytosis, we assayed the extracellular release of the lysosomal marker enzyme β-hexosaminidase. Application of thapsigargin evoked a time-dependent increase in extracellular β-hexosaminidase (Fig. 2A), demonstrating that the release of ER calcium can promote lysosome secretion. Furthermore, GPN also promoted substantial β-hexosaminidase release (Fig. 2B). The rate of GPN triggered β-hexosaminidase secretion was much faster than that of thapsigargin (Fig. 2A,B), which may relate to the quicker calcium rise evoked by GPN with respect to thapsigargin (Fig. 1A,C). Interestingly, GPN-induced β-hexosaminidase secretion was abolished by pre-incubation with NEM (Fig. 2C), suggesting that the NEM-sensitive factor (NSF) and vesicle exocytosis is required for GPN and thapsigargin to trigger β-hexosaminidase secretion. Microscopic examination of monocytes revealed dramatic ruffling of the plasma membrane upon addition of GPN (Fig. 2D), characteristic of exocytosis described in other cell types (Benninger et al., 2009). We next tested the dependency of GPN-evoked calcium response on NEM. The increase in [Ca2+]i in response to thapsigargin was unaffected by NEM treatment (data not shown), though strikingly the Ca2+ response to GPN was abolished (Fig. 2E,F). This data demonstrate that the observed GPN-evoked increase in [Ca2+]i and β-hexosaminidase secretion are both dependent upon lysosome exocytosis. The calcium response evoked by 0.4 mM GPN was less sensitive to NEM (Fig. 2F). This partial sensitivity is consistent with a calcium rise independent of exocytosis and with the dantrolene sensitivity of the response to 0.4 mM GPN (Fig. 1G). Next we sought to test the dependency of GPN triggered β-hexosaminidase secretion on changes in calcium by buffering cytosolic calcium with BAPTA-AM (50 µM, 30 mins). Incubation of cells with BAPTA-AM abolished the ability of GPN to trigger β-hexosaminidase secretion (Fig. 2G).

Fig. 2.

GPN-evoked Ca2+ responses are dependent upon lysosome exocytosis. Thapsigargin (5 µM; A) and GPN (0.2 mM, B) stimulate release of lysosomal enzyme β-hexosaminidase in THP-1 monocytes (n = 5; *P<0.01). Control responses are with DMSO vehicle. (C) Inhibition of vesicular exocytosis by N-ethylmaleimide (1 mM, 15 mins; NEM) inhibits time-dependent GPN-evoked β-hexosaminidase release (n = 6; *P<0.05; **P<0.01). (D) GPN (0.2 mM, 5 mins) stimulates membrane protrusions and reorganisation (arrows). Scale bar: 20 µm. (E,F) NEM pre-treatment (1 mM, 15 mins) abolishes 0.2 mM GPN-evoked Ca2+ responses (n = 5; *P<0.01 vs control) but not response to 0.4 mM GPN (n = 3; #P<0.01 vs 0.2 mM GPN with NEM). White bars, control; black bars, NEM. (G) β-hexosaminidase release 2 mins after GPN (0.2 mM) application with and without cytosolic Ca2+ buffering with BAPTA-AM (50 mM, 30 mins; n = 4; *P<0.01).

Fig. 2.

GPN-evoked Ca2+ responses are dependent upon lysosome exocytosis. Thapsigargin (5 µM; A) and GPN (0.2 mM, B) stimulate release of lysosomal enzyme β-hexosaminidase in THP-1 monocytes (n = 5; *P<0.01). Control responses are with DMSO vehicle. (C) Inhibition of vesicular exocytosis by N-ethylmaleimide (1 mM, 15 mins; NEM) inhibits time-dependent GPN-evoked β-hexosaminidase release (n = 6; *P<0.05; **P<0.01). (D) GPN (0.2 mM, 5 mins) stimulates membrane protrusions and reorganisation (arrows). Scale bar: 20 µm. (E,F) NEM pre-treatment (1 mM, 15 mins) abolishes 0.2 mM GPN-evoked Ca2+ responses (n = 5; *P<0.01 vs control) but not response to 0.4 mM GPN (n = 3; #P<0.01 vs 0.2 mM GPN with NEM). White bars, control; black bars, NEM. (G) β-hexosaminidase release 2 mins after GPN (0.2 mM) application with and without cytosolic Ca2+ buffering with BAPTA-AM (50 mM, 30 mins; n = 4; *P<0.01).

GPN-evoked Ca2+ response is dependent upon secreted ATP

As exocytosis of lysosomes is associated with release of ATP in other cell types including neuronal astrocytes (Zhang et al., 2007), we sought to determine whether the Ca2+ response to GPN was dependent upon extracellular ATP. In paired experiments were [Ca2+]i and the extracellular ATP concentration ([ATP]ext) were sampled simultaneously, it is evident that a release of ATP is associated with the GPN-evoked Ca2+ response (Fig. 3A). At rest, a basal [ATP]ext of approximately 35 nM was detectable, which rose to about 100 nM at the peak of the GPN-evoked Ca2+ response (Fig. 3B). The GPN-evoked increase in [ATP]ext was abolished following treatment with BAPTA-AM or apyrase (Fig. 3B). BAPTA-AM treatment also reduced [ATP]ext in the absence of GPN (Fig. 3B). As observed for GPN-evoked β-hexosaminidase secretion and [Ca2+]i increase, ATP secretion was also inhibited by NEM (Fig. 3C). NEM had no direct effect itself on the luciferase-luciferin assay or β-hexosaminidase secretion assays used in this study (supplementary material Fig. S1). In further experiments we tested whether scavenging the GPN-evoked released ATP impacted on the Ca2+ response. Surprisingly treatment with apyrase inhibited the GPN-evoked Ca2+ response by over 80% (Fig. 3D). This effect was not observed following heat-inactivation of apyrase, suggesting ATP/ADP hydrolase activity is required for the inhibitory action of apyrase (Fig. 3D,E). These data suggest that the GPN-evoked release of ATP mediates the GPN-evoked Ca2+ increase. MTT cell viability assays where performed to exclude GPN-induced cell lysis as an alternative route of ATP release (Fig. 3F).

Fig. 3.

GPN-evoked Ca2+ responses couple to and are dependent upon vesicular secreted ATP. (A) Simultaneous measurement of GPN-evoked Ca2+ response and extracellular ATP concentration. Arrow denotes sampling points for ATP measurement. (B) Summarised extracellular ATP concentration in unstimulated and GPN-stimulated THP-1 monocytes. Addition of BAPTA-AM (50 µM, 30 mins) or apyrase (4 IU/ml) abolishes GPN-evoked external ATP increase (n = 4-6; *P<0.01 test vs control; #P<0.01 test vs GPN alone). (C) Inhibition of vesicular exocytosis by NEM (1 mM, 15 mins) abolishes GPN-evoked ATP secretion (n = 5; *P<0.01). (D) Scavenging extracellular ATP with apyrase (4 IU/ml) attenuates GPN-evoked Ca2+ responses. Heat-inactivated (HI) apyrase is ineffective (n = 5). (E) Average peak Ca2+ responses evoked by GPN with and without ATP scavenging (n = 5; *P<0.01). (F) MTT cell viability assay. MTT absorbance at 595 nm of control monocytes or monocytes exposed to 0.2 or 0.4 mM GPN for 30 mins (n = 3; P>0.05).

Fig. 3.

GPN-evoked Ca2+ responses couple to and are dependent upon vesicular secreted ATP. (A) Simultaneous measurement of GPN-evoked Ca2+ response and extracellular ATP concentration. Arrow denotes sampling points for ATP measurement. (B) Summarised extracellular ATP concentration in unstimulated and GPN-stimulated THP-1 monocytes. Addition of BAPTA-AM (50 µM, 30 mins) or apyrase (4 IU/ml) abolishes GPN-evoked external ATP increase (n = 4-6; *P<0.01 test vs control; #P<0.01 test vs GPN alone). (C) Inhibition of vesicular exocytosis by NEM (1 mM, 15 mins) abolishes GPN-evoked ATP secretion (n = 5; *P<0.01). (D) Scavenging extracellular ATP with apyrase (4 IU/ml) attenuates GPN-evoked Ca2+ responses. Heat-inactivated (HI) apyrase is ineffective (n = 5). (E) Average peak Ca2+ responses evoked by GPN with and without ATP scavenging (n = 5; *P<0.01). (F) MTT cell viability assay. MTT absorbance at 595 nm of control monocytes or monocytes exposed to 0.2 or 0.4 mM GPN for 30 mins (n = 3; P>0.05).

GPN requires engagement of P2Y receptors to elevate [Ca2+]i

Previous studies have demonstrated that monocytes abundantly express ATP-activated purinergic receptors (Moore et al., 2001). As the GPN-evoked rise in [ATP]ext is insufficient to activate P2X receptors (North, 2002; Fountain and North, 2006), we sought to test an interaction between GPN and P2Y receptor activation. ATP, ADP, UTP and UDP all stimulated significant increases in [Ca2+]i (Fig. 4A,B). All responses were inhibited by pre-incubation with thapsigargin or the phospholipase C inhibitor U-73122 (Fig. 4C). These data suggest that P2Y-receptor-mediated responses are completely dependent upon Ins(1,4,5)P3 generation and release of ER calcium. Despite this, GPN pre-treatment was able to inhibit nucleotide-evoked Ca2+ responses in THP-1 monocytes (Fig. 4A,B). This is unlikely to represent a dependency upon release of stored lysosomal calcium as the response is entirely dependent upon ER calcium release (Fig. 4C). Rather this is likely to be due to GPN-evoked ATP release (Fig. 3A) causing a desensitisation of subsequent nucleotide responses. Indeed, we observed that the response to GPN requires engagement of P2Y receptors, as antagonising P2Y receptors with suramin or MRS-2578 significantly impairs GPN-evoked [Ca2+]i (Fig. 5), and Ca2+ responses to exogenous ATP display marked desensitisation (Fig. 4D). Consistent with results given in Fig. 1G and Fig. 2F, the calcium response to 0.4 mM GPN is less sensitive to P2 receptor antagonists (Fig. 5C). This further demonstrates that the additional calcium responses evoked by 0.4 mM GPN is independent upon exocytosis and P2Y receptor engagement but dependent upon direct release of ER calcium via ryanodine receptors.

Fig. 4.

P2Y-receptor-mediated Ca2+ responses are attenuated by GPN treatment. (A) Ca2+ responses evoked by 30 µM nucleotides (ATP, ADP, UTP or UDP) with (grey line) or without (black line) GPN pre-incubation (0.2 mM, 15 mins). (B) Average peak Ca2+ response (n = 6; *P<0.05). (C) Inhibition of ATP-evoked Ca2+ response by pre-incubation with thapsigargin (5 µM, 15 mins) and or U-73122 (10 µM, 15 mins) demonstrates dependency of ATP response on phospholipase C activity and ER calcium mobilisation (n = 4). (D) Desensitisation of ATP-evoked Ca2+ responses by repeated ATP application. Response to 30 µM ATP with 10 min intervals between applications. All experiments are performed in the absence of external Ca2+.

Fig. 4.

P2Y-receptor-mediated Ca2+ responses are attenuated by GPN treatment. (A) Ca2+ responses evoked by 30 µM nucleotides (ATP, ADP, UTP or UDP) with (grey line) or without (black line) GPN pre-incubation (0.2 mM, 15 mins). (B) Average peak Ca2+ response (n = 6; *P<0.05). (C) Inhibition of ATP-evoked Ca2+ response by pre-incubation with thapsigargin (5 µM, 15 mins) and or U-73122 (10 µM, 15 mins) demonstrates dependency of ATP response on phospholipase C activity and ER calcium mobilisation (n = 4). (D) Desensitisation of ATP-evoked Ca2+ responses by repeated ATP application. Response to 30 µM ATP with 10 min intervals between applications. All experiments are performed in the absence of external Ca2+.

Fig. 5.

GPN-evoked Ca2+ responses require engagement of purinergic receptors. (A,B) Effect of P2Y receptor antagonists suramin (100 µM; A) and MRS2578 (1 µM; B) on GPN-evoked Ca2+ responses in THP-1 monocytes. (C) Sensitivity of calcium responses to P2 receptor antagonists when exposed to 0.2 mM and 0.4 mM GPN (n = 3-5; *P<0.05 vs control; #P<0.05 vs 0.2 mM GPN equivalent. Average peak Ca2+ responses in control conditions and in the presence of antagonist (n = 5; *P<0.05).

Fig. 5.

GPN-evoked Ca2+ responses require engagement of purinergic receptors. (A,B) Effect of P2Y receptor antagonists suramin (100 µM; A) and MRS2578 (1 µM; B) on GPN-evoked Ca2+ responses in THP-1 monocytes. (C) Sensitivity of calcium responses to P2 receptor antagonists when exposed to 0.2 mM and 0.4 mM GPN (n = 3-5; *P<0.05 vs control; #P<0.05 vs 0.2 mM GPN equivalent. Average peak Ca2+ responses in control conditions and in the presence of antagonist (n = 5; *P<0.05).

Constitutive release of ATP and P2Y receptor activation control Ca2+ homeostasis in monocytes

Our prior observations demonstrate that apyrase inhibits the GPN-induced increase in [ATP]ext (Fig. 3D). However, we also observe that apyrase treatment significantly reduces baseline [ATP]ext (Fig. 3B). In paired experiments where baseline [Ca2+]i and [ATP]ext are measured simultaneously, application of NEM reduced both [Ca2+]i and [ATP]ext (Fig. 6A). These data suggest inhibition of the exocytotic release of ATP is a determinant of baseline [Ca2+]i. To test this further, we examined the effect of apyrase and purinergic receptor antagonists on resting [Ca2+]i. Application of apyrase (Fig. 6C), but not heat-inactivated apyrase (Fig. 6D), caused a reduction in resting [Ca2+]i, as did the broad-spectrum P2 receptor antagonist suramin (Fig. 6E). The selective P2Y11 antagonist NF380 also reduced baseline [Ca2+]i (Fig. 6F), though the selective P2Y6 antagonist MRS2578 caused a small transient increase in calcium which returned to a pre-application baseline (data not shown). Interestingly, the P1 adenosine receptor antagonist CGS15943 elevated resting [Ca2+]i, suggesting tonic P1 receptor activation negatively regulates resting [Ca2+]i in monocytes (Fig. 6G). In a concluding series of experiments we sought to examine whether disrupting lysosomes with GPN could impact on monocyte function in vitro. In Transwell migration assays, THP-1 monocytes migrated toward 1 µM ATP (Fig. 7). Migration was inhibited by suramin (Fig. 7), suggesting engagement of P2 receptors is required for ATP-evoked migration. Importantly, GPN abolished migration (Fig. 7), suggesting disruption of lysosomes impairs the ability of monocytes to migrate towards ATP. GPN, apyrase or P2 receptor antagonists had no effect on basal cell migration (Fig. 7C). Monocytes also migrated towards serum yet this was not inhibited by GPN or apyrase (Fig. 7D).

Fig. 6.

Constitutive vesicular secretion of ATP controls Ca2+ homeostasis in human monocytes. (A,B) Simultaneous measurement of basal intracellular Ca2+ concentration (A) and extracellular ATP concentration (B) following inhibition of vesicular exocytosis by NEM. Arrows denote sampling points for extracellular ATP. Bar chart represents extracellular ATP concentration at sampling points 1 and 6 as given in A (n = 5; *P<0.01). (C-G) Effect of antagonising purinergic receptors on baseline intracellular Ca2+ concentration. (C,D) Effect of scavenging extracellular ATP with apyrase (4 IU/ml; C) or heat-inactivated (HI) apyrase (D). (E-G) Effect of non-selective P2 receptor antagonist suramin (100 µM), selective P2Y11 antagonist NF380 or P1 receptor antagonist CGS15943 (1 µM) on basal intracellular Ca2+ concentration. (H) Average effects of pharmacological interventions on basal intracellular Ca2+ concentration (n = 5; *P<0.05).

Fig. 6.

Constitutive vesicular secretion of ATP controls Ca2+ homeostasis in human monocytes. (A,B) Simultaneous measurement of basal intracellular Ca2+ concentration (A) and extracellular ATP concentration (B) following inhibition of vesicular exocytosis by NEM. Arrows denote sampling points for extracellular ATP. Bar chart represents extracellular ATP concentration at sampling points 1 and 6 as given in A (n = 5; *P<0.01). (C-G) Effect of antagonising purinergic receptors on baseline intracellular Ca2+ concentration. (C,D) Effect of scavenging extracellular ATP with apyrase (4 IU/ml; C) or heat-inactivated (HI) apyrase (D). (E-G) Effect of non-selective P2 receptor antagonist suramin (100 µM), selective P2Y11 antagonist NF380 or P1 receptor antagonist CGS15943 (1 µM) on basal intracellular Ca2+ concentration. (H) Average effects of pharmacological interventions on basal intracellular Ca2+ concentration (n = 5; *P<0.05).

Fig. 7.

GPN inhibits chemotactic migration of monocytes toward ATP. (A) Transwell migration assays using 1 µM ATP as the chemoattractant in the lower chamber. Representative Transwell membranes showing THP-1 monocytes stained with Crystal Violet following migration (1 hour) with ATP, in the absence of ATP, and with ATP in the presence of GPN (0.2 mM) or suramin (100 µM). Scale bar: 200 μm. (B) Average chemotactic index of monocytes for ATP in different experimental conditions (n = 5; *P<0.05). (C,D) Effect of GPN, apyrase and P2 receptor antagonists on basal cell migration (C) and with the addition of 1% serum (D) (n = 3 or 4; P>0.05).

Fig. 7.

GPN inhibits chemotactic migration of monocytes toward ATP. (A) Transwell migration assays using 1 µM ATP as the chemoattractant in the lower chamber. Representative Transwell membranes showing THP-1 monocytes stained with Crystal Violet following migration (1 hour) with ATP, in the absence of ATP, and with ATP in the presence of GPN (0.2 mM) or suramin (100 µM). Scale bar: 200 μm. (B) Average chemotactic index of monocytes for ATP in different experimental conditions (n = 5; *P<0.05). (C,D) Effect of GPN, apyrase and P2 receptor antagonists on basal cell migration (C) and with the addition of 1% serum (D) (n = 3 or 4; P>0.05).

GPN-induced Ca2+ mobilisation

Exposure of human monocytes to the cathepsin C (dipeptidyl peptidase I) (Muno et al., 1993) substrate GPN at low millimolar concentrations caused robust elevation in [Ca2+]i in the absence of extracellular Ca2+. GPN is a useful pharmacological tool for the selective disruption of lysosomes, capable of distinguishing lysosomes from endosomes and autophagosomes (Berg et al., 1994). The proposed mechanism of action of GPN is osmotic lysis of lysosomes attributed to increased luminal tonicity following GPN hydrolysis by cathepsin C (Srinivas et al., 2002), consequently released stored Ca2+ into the cytoplasm. Our data are constituent with the notion that GPN disrupts lysosomes, as Lysotracker staining in monocytes is discharged following only short exposures to GPN (Fig. 1B). However, it is clear from our current study that GPN also disrupts lysosomal Ca2+ pools by triggering their exocytosis as GPN-evoked β-hexosaminidase secretion is attenuated following inhibition of vesicular exocytosis by N-ethylmaleimide (Fig. 2C). Experiments with BAPTA-AM demonstrate that exocytosis is dependent upon a rise in intracellular calcium (Fig. 2G, Fig. 3B). Ca2+-dependent exocytosis of lysosomes has also been described in fibroblasts and epithelia (Rodríguez et al., 1999; Martinez et al., 2000), with raised [Ca2+]i promoting exocytosis through activation of various regulated proteins associated with lysosomes (Holt et al., 2006; Higashio et al., 2008). Inhibition of GPN-evoked β-hexosaminidase secretion in monocytes implicates a NEM-sensitive factor (NSF) regulates lysosome exocytosis following raised [Ca2+]i, suggesting regulation by SNAREs (Rao et al., 2004). Exocytosis of monocytic lysosomes can occur following activation by inflammatory stimulants including bacterial lipopolysaccharide and contributes to the processing and secretion of interleukin-1β (IL-1β) (Andrei et al., 2004).

Interaction between lysosomal and ER Ca2+ stores in monocytes

The data presented here suggests that communication between lysosome and ER Ca2+ pools is bidirectional and both direct, through activation of ER ryanodine receptors, and indirect through release of ATP and activation of cell surface PLC-coupled purinergic receptors. Exposure to thapsigargin was sufficient to discharge Lysotracker staining, stimulate β-hexosaminidase secretion and ablate GPN-evoked Ca2+ responses. This data suggests ER calcium release is capable of promoting lysosome exocytosis. This can explain why GPN fails to elicit a Ca2+ response following exposure to thapsigargin. Studies in epithelial cells demonstrate that lysosomal Ca2+ pools can be mobilised by Ins(I,4,5)P3 (Haller et al., 1996); however, similar studies in endothelium reveal lysosomal Ca2+ is not linked to thapsigargin-sensitive stores (Srinivas et al., 2002). It is evident that release of ER Ca2+ can be triggered directly by GPN-evoked release of lysosomal Ca2+ through engagement of ER ryanodine receptors, as the enhanced Ca2+ response to higher concentrations (0.4 mM) of GPN (presumably working by disrupting greater numbers of lysosomes) is antagonised by dantrolene (Fig. 1F). These findings are consistent with the identification of functional ryanodine receptors in monocytes by others (Bracci et al., 2007; Cardaba and Mueller, 2009; Guidarelli et al., 2009). The Ca2+ influx observed following GPN-evoked Ca2+ responses after Ca2+ add-back is reminiscent of capacitative calcium entry (CCE) observed following depletion of ER. Our data is consistent with activation of CCE pathways in corneal endothelium following disruption of lysosomal Ca2+ pools (Srinivas et al., 2002).

ATP secretion couples lysosomal and ER Ca2+ stores via P2Y receptor activation

Scavenging extracellular ATP/ADP using apyrase severely attenuates Ca2+ responses evoked by GPN (Fig. 3). Heat-inactivated apyrase had no effect suggesting the ATP diphosphohydrolase is necessary requirement for inhibition. An apyrase-resistant component to the GPN-evoked Ca2+ response is apparent (Fig. 3D), which may represent raised [Ca2+]i due to lytic activity of GPN on lysosomes. In experiments where extracellular ATP and [Ca2+]i are simultaneously measured, it is clear that the GPN-evoked Ca2+ response is associated with ATP secretion (Fig. 3). As for GPN-evoked β-hexosaminidase secretion, the GPN-evoked Ca2+ response is also ablated following inhibition of vesicular exocytosis by N-ethylmaleimide. This data strongly suggest the source of extracellular ATP is exocytosis of lysosomes. Astrocytes also respond to elevated [Ca2+]i by vesicular release of ATP (Zhang et al., 2004a; Zhang et al., 2004b). Some elegant studies have demonstrated that the vesicles responsible for exocytotic release of ATP are lysosomes (Zhang et al., 2007). Granular release of ATP/ADP is important for platelet function (Daniel et al., 1998). The peak extracellular ATP concentration following GPN exposure was approximately 100 nM, which is sufficient to activate P2Y receptors but not P2X receptors (North, 2002; von Kügelgen, 2006). In addition, apyrase attenuates GPN-evoked Ca2+ responses in the absence of extracellular Ca2+ (Fig. 3D), suggesting a dependency on release of intracellular calcium rather than calcium influx. P2Y receptors differ in their activation profile by adenine (ATP, ADP) and uracil nucleotides (UTP, UDP) (von Kügelgen, 2006). We observed Ca2+ responses to micromolar ATP, ADP, UTP and UDP in THP-1 monocytic cells (Fig. 4). Human primary monocytes and monocytic cells express various P2X and P2Y receptor subtypes, of which P2Y2, P2Y6 and P2Y11 are expressed by unstimulated THP-1 cells (Moore et al., 2001; Li and Fountain, 2012). P2Y2, P2Y6 and P2Y11 are all PLC-coupled receptors and hence have the capacity to mobilise ER Ca2+ through Ins(I,4,5)P3 synthesis when activated. Antagonism by suramin is not sufficient to discriminate between P2Y2, P2Y6 or P2Y11 (von Kügelgen, 2006), though antagonism by MRS2578 demonstrates activation of P2Y6 is concomitant with the GPN-evoked Ca2+ response. Though UDP is the preferred agonist at P2Y6 receptors, ADP also activates the receptor (Robaye et al., 1997). The effect of apyrase, a specific adenosine diphosphatase, on the GPN-evoked Ca2+ response implicates extracellular ATP or ADP as mediators, though co-secretion of uridine nucleotides cannot be excluded.

Effect of GPN on P2Y-receptor-mediated Ca2+ responses

Pre-treatment with GPN attenuated P2Y-receptor-mediated Ca2+ responses in THP-1 monocytes (Fig. 4). This may suggest that P2Y receptors couple to lysosomal Ca2+ stores and that depleting the store with GPN subsequently decreases the magnitude of P2Y-receptor-mediated responses. However, we suggest that the effect of GPN on P2Y-receptor-mediated Ca2+ responses is due to desensitisation of P2Y receptors following GPN-evoked ATP release. This notion is supported by several observations. Firstly, ATP-evoked Ca2+ responses are completely inhibited by thapsigargin and U-73122 (Fig. 4C). The effect of thapsigargin could possibly be explained by disruption of lysosomal Ca2+ stores by lysosome exocytosis (Fig. 2A); however, inhibition by U-73122 suggests a requirement for phospholipase C activity and therefore Ins(I,4,5)P3-dependent release of ER Ca2+ is a more plausible explanation. Secondly, ATP-evoked Ca2+ responses are highly sensitive to desensitisation by repeated agonist application (Fig. 4D). P2Y receptor activation could also explain why GPN triggers CCE (Fig. 1E). Our data also raises the possibility that agonists which release ER Ca2+ could also induce lysosomal ATP secretion. Indeed, the ATP release is associated with reception of chemoattractant stimulates such as N-formyl-methionyl-leucly-phenylalanine in neutrophils (Chen et al., 2006) though the signalling coupling to ATP secretion is not clear. We suggest the chemotactic response to ATP is impaired due to GPN-evoked ATP secretion and P2Y receptor desensitisation (Fig. 4).

Constitutive vesicular ATP release contributes to resting [Ca2+]i

Application of apyrase (but not heat-inactivated apyrase) or suramin caused a robust decrease in baseline [Ca2+]i which slowly recovered over several minutes post exposure (Fig. 6). In luminometer experiments, the amount of extracellular ATP measured in resting cells was around 30 nM (Fig. 6). Basal secretion of ATP has been observed in other cell types including renal epithelium (Hovater et al., 2008), osteoblasts (Genetos et al., 2005), pituitary cells (Li et al., 2011) and astrocytes (Coco et al., 2003). The basal level of ATP secreted by THP-1 cells breaches the activation threshold for many P2Y receptors (Parr et al., 1994; von Kügelgen et al., 2006). Though the level of pericellular ATP is likely to be much higher, the absence of extracellular calcium in these experiments precludes any involvement of P2X receptors. Of the Gq-coupled P2Y receptors activated by ATP or ADP, P2Y2, P2Y6 and P2Y11 are expressed by THP-1 monocytes (Moore et al., 2001). P2Y2 and P2Y11 are both antagonised by suramin (Communi et al., 1999; von Kügelgen, 2005) and P2Y11 selectively antagonised by NF340 (Meis et al., 2010). P2Y2 and P2Y11 mediated response are completely blocked at 100 µM suramin (Communi et al., 1999; von Kügelgen, 2005) as used in Fig. 6, and therefore the smaller effect of NF380 on baseline [Ca2+]i suggests a major contribution of P2Y2, for which no selective antagonists are available. The effect of suramin, apyrase and NF380 are therefore best explained if constitutive activation of P2Y receptors contributes to resting [Ca2+]i in these experiments, though the failure of MRS2578 to reduce baseline calcium suggest P2Y6 is not constitutively active. Conversely, antagonism of P1 adenosine receptors with CGS15943 raises baseline [Ca2+]i. These data suggest tonic P1 receptor activation negatively regulates resting [Ca2+]i, though the precise mechanism requires further investigation. The basal concentration of extracellular ATP is achieved through vesicular release of ATP, as N-ethylmaleimide caused a concomitant reduction in extracellular ATP and [Ca2+]i. The likely source of vesicular ATP is lysosomes as N-ethylmaleimide suppresses β-hexosaminidase secretion (Fig. 2). Furthermore, it is evident that monocytes caused a time-dependent conditioning of buffer with β-hexosaminidase in the absence of stimulant (Fig. 2), strongly suggesting that lysosomes undergo constitutive exocytosis. Experiments with BAPTA-AM demonstrate that buffering cytosolic calcium reduces the amount of constitutively secreted ATP (Fig. 3B), suggesting that constitutive as well as stimulated lysosome exocytosis is calcium dependent in monocytes.

Implications for GPN as a pharmacological tool

Investigation into the function and regulation of acidic Ca2+ stores has increased greatly in the past 10 years with many landmark discoveries (Calcraft et al., 2009; Patel and Docampo, 2010). In many such studies GPN is a central pharmacological tool used to destroy lysosomes and their subsequent contribution to cellular Ca2+ signals. The data presented here suggests that secreted ATP may be a common feature when cells are exposed to GPN and that consequent second messenger signalling through P2Y receptor activation may have to be taken into consideration. However, it remains to be demonstrated whether GPN-evoked ATP secretion is observed outside cells of hematopoietic lineage that possess Ca2+-sensitive secretory lysosomes (Holt et al., 2006). Nucleotide-evoked Ca2+ responses appear unaffected in corneal endothelium following exposure to GPN (Srinivas et al., 2002).

Conclusions

The data presented here suggests that constitutive vesicular secretion of ATP regulates Ca2+ homeostasis in THP-1 human monocytic cells via tonic activation of P2Y receptors. ATP secretion and subsequent P2Y receptor activation contributes significantly to Ca2+ signals generated by lysosome disruption with the cathepsin C substrate GPN. Bidirectional communication exists between lysosomal and ER Ca2+ stores via several routes (i) ER to lysosome: release of ER Ca2+ triggers lysosome exocytosis; (ii) lysosome to ER: activation of ryanodine receptors; and (iii) lysosome to ER: release of ATP and mobilisation of ER calcium through activation of PLC-coupled P2Y receptors (Fig. 8).

Fig. 8.

Bidirectional communication between lysosomal and ER Ca2+ in monocytes. Lysosome to ER: release of lysosomal Ca2+ by the cathepsin C substrate GPN stimulates lysosome exocytosis by a NEM-sensitive-factor (NSF)-dependent pathway causing extracellular release of ATP. ATP activates cell surface phospholipase C (PLC)-coupled P2Y receptors to generate Ins(1,4,5)P3 (IP3), activate Ins(1,4,5)P3 receptor (IP3R) and release ER Ca2+. Release of lysosomal Ca2+ can also directly release ER Ca2+ through activation of ryanodine receptors (RyR). ER to lysosome: release of ER Ca2+ stimulates exocytosis of lysosomal Ca2+ stores.

Fig. 8.

Bidirectional communication between lysosomal and ER Ca2+ in monocytes. Lysosome to ER: release of lysosomal Ca2+ by the cathepsin C substrate GPN stimulates lysosome exocytosis by a NEM-sensitive-factor (NSF)-dependent pathway causing extracellular release of ATP. ATP activates cell surface phospholipase C (PLC)-coupled P2Y receptors to generate Ins(1,4,5)P3 (IP3), activate Ins(1,4,5)P3 receptor (IP3R) and release ER Ca2+. Release of lysosomal Ca2+ can also directly release ER Ca2+ through activation of ryanodine receptors (RyR). ER to lysosome: release of ER Ca2+ stimulates exocytosis of lysosomal Ca2+ stores.

Intracellular calcium measurements

THP-1 monocytes were cultured in RPMI 1640 medium with L-glutamine and 10% (v/v) serum at 37°C and 5% CO2. Cultures contained 50 IU/ml penicillin and 50 µg/ml streptomycin. Cultures were maintained between 1×105 and 8×105 cells/ml. For intracellular calcium measurements, 1×106 cells/ml were loaded with Fluo-4 in SBS buffer containing (mM): NaCl, 130; KCl, 5; MgCl2, 1.2; CaCl2, 1.5; D-glucose, 8; HEPES, 10; pH 7.4) plus 0.01% (w/v) pluronic acid and 2 µM Fluo-4-AM. Cells were loaded for 1 hour at 37°C. Cells were resuspended at 1×106 cells/ml in SBS buffer supplemented with 1 mM EGTA and without CaCl2. Fluo-4 fluorescence (494 nm excitation; 516 nm emission) was sampled at 1 s intervals using a Hitachi F-2000 fluorescence spectrophotometer. Cells in a quartz cuvette were continuously agitated by means of a magnetic stirrer. Maximum fluorescence (Fmax) signals were generated by addition of 40 µM digitonin in the presence of 1.5 mM CaCl2. Calcium responses to drugs were expressed at a percentage of Fmax.

β-hexosaminidase secretion assay

THP-1 monocytes were resuspended at 2×106 cells/ml in SBS buffer (no calcium+1 mM EGTA) in 24-well plates. Cells were equilibrated in SBS 3 hours prior to experimentation. β-hexosaminidase activity in 200 µl supernatant samples was measured by incubation with 200 µl of 1 mM p-nitrophenyl N-acetyl-β-D-glucosaminide in 0.1 M citrate buffer (0.05 M citric acid, 0.05 M sodium citrate, pH 4.5) for 1 hour at 37°C. Reactions were quenched by addition of 400 µl 0.1 M sodium carbonate buffer. Absorbance was read at 405 nm. Absorbance values were background subtracted using zero time-point values and normalised to maximal signals generated following cell lysis with 1% (v/v) Triton X-100.

ATP secretion assay

Sample ATP was assayed by means of an ATP bioluminescence assay kit (CLSII kit, Roche). Extracellular ATP was simultaneously measured with intracellular calcium by sampling supernatants from Fluo-4-loaded cells undergoing intracellular calcium measurements as described above. 150 µl samples were withdrawn and cells sedimented at 4°C. Clarified samples were mixed 1∶4 with luciferase reagent before luminescence measurements were made using a Modulus Luminometer (Turner BioSystems) and 7 s integration time.

Transwell migration assay

Migration assays were performed in 24-well plates using polyethylene terephthalate (PET) membrane Transwell inserts with 3 µm pores (BD Biosciences). 1×106 monocytes in RMPI without serum were added to upper chambers and 1 µM ATP chemoattractant to lower chambers. GPN or suramin was added to upper wells where appropriate. Cell migration was allowed to progress for 1 hour at 37°C with 5% CO2 in a humidified incubator. Inserts were fixed with −20°C methanol, upper chambers were swabbed and cells stained with 0.5% (w/v) Crystal Violet. Migrated cells were scored using an Olympus CKX41 inverted microscope equipped with a Leica digital camera. Basal migration was assessed in the absence of chemoattractant in the lower chamber.

Microscopy

Differential interference contrast and fluorescent images were obtained using an Olympus IX71 inverted microscope equipped with a Hamatasu digital camera. For Lysotracker staining, monocytes were incubated with 100 nM Lysotracker DND-99 in Ca2+-free SBS buffer for 20 mins prior to exposure to drugs. Drugs were delivered by bath perfusion.

Cell viability assays

THP-1 monocytes (1×106) were exposed to GPN at 0.2 and 0.3 mM for 30 mins at 37°C. Cell viability for assessed by the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay. Cells were incubated with 12.5 mg/ml MTT for 1 hour at 37°C followed by solubilisation in DMSO and measurement of absorbance at 595 nm.

Chemicals and reagents

All chemicals were purchased from Sigma with the exception of GPN (Insight Biotechnology Ltd), Fluo-4-AM and Lysotracker (Invitrogen), Suramin, MRS2578 and CGS15943 (Tocris). THP-1 cells where procured from ATCC.

Temperature conditions

All experiments were performed at room temperature (22°C) unless otherwise stated.

Statistical analysis

Data is expressed as means ± s.e.m. Hypothesis testing was by means of paired Student t-tests.

Funding

This work was supported by the BBSRC [grant number BB/F023588 to S.J.F.]; and The Physiological Society studentship award to S.B.

Andrei
C.
,
Margiocco
P.
,
Poggi
A.
,
Lotti
L. V.
,
Torrisi
M. R.
,
Rubartelli
A.
(
2004
).
Phospholipases C and A2 control lysosome-mediated IL-1 β secretion: Implications for inflammatory processes.
Proc. Natl. Acad. Sci. USA
101
,
9745
9750
.
Badolato
R.
,
Johnston
J. A.
,
Wang
J. M.
,
McVicar
D.
,
Xu
L. L.
,
Oppenheim
J. J.
,
Kelvin
D. J.
(
1995
).
Serum amyloid A induces calcium mobilization and chemotaxis of human monocytes by activating a pertussis toxin-sensitive signaling pathway.
J. Immunol.
155
,
4004
4010
.
Benninger
R. K. P.
,
Vanherberghen
B.
,
Young
S.
,
Taner
S. B.
,
Culley
F. J.
,
Schnyder
T.
,
Neil
M. A. A.
,
Wüstner
D.
,
French
P. M. W.
,
Davis
D. M.
et al.  (
2009
).
Live cell linear dichroism imaging reveals extensive membrane ruffling within the docking structure of natural killer cell immune synapses.
Biophys. J.
96
,
L13
L15
.
Berg
T. O.
,
Strømhaug
E.
,
Løvdal
T.
,
Seglen
O.
,
Berg
T.
(
1994
).
Use of glycyl-L-phenylalanine 2-naphthylamide, a lysosome-disrupting cathepsin C substrate, to distinguish between lysosomes and prelysosomal endocytic vacuoles.
Biochem. J.
300
,
229
236
.
Berridge
M. J.
,
Lipp
P.
,
Bootman
M. D.
(
2000
).
The versatility and universality of calcium signalling.
Nat. Rev. Mol. Cell Biol.
1
,
11
21
.
Bracci
L.
,
Vukcevic
M.
,
Spagnoli
G.
,
Ducreux
S.
,
Zorzato
F.
,
Treves
S.
(
2007
).
Ca2+ signaling through ryanodine receptor 1 enhances maturation and activation of human dendritic cells.
J. Cell Sci.
120
,
2232
2240
.
Calcraft
P. J.
,
Ruas
M.
,
Pan
Z.
,
Cheng
X.
,
Arredouani
A.
,
Hao
X.
,
Tang
J.
,
Rietdorf
K.
,
Teboul
L.
,
Chuang
K-T.
et al.  (
2009
).
NAADP mobilizes calcium from acidic organelles through two-pore channels.
Nature
459
,
596
600
.
Cardaba
C. M.
,
Mueller
A.
(
2009
).
Distinct modes of molecular regulation of CCL3 induced calcium flux in monocytic cells.
Biochem. Pharmacol.
78
,
974
982
.
Chen
F. W.
,
Li
C.
,
Ioannou
Y. A.
(
2010
).
Cyclodextrin induces calcium-dependent lysosomal exocytosis.
PLoS ONE
5
,
e15054
.
Chen
Y.
,
Corriden
R.
,
Inoue
Y.
,
Yip
L.
,
Hashiguchi
N.
,
Zinkernagel
A.
,
Nizet
V.
,
Insel
P. A.
,
Junger
W. G.
(
2006
).
ATP release guides neutrophil chemotaxis via P2Y2 and A3 receptors.
Science
314
,
1792
1795
.
Christensen
K. A.
,
Myers
J. T.
,
Swanson
J. A.
(
2002
).
pH-dependent regulation of lysosomal calcium in macrophages.
J. Cell Sci.
115
,
599
607
.
Churchill
G. C.
,
Okada
Y.
,
Thomas
J. M.
,
Genazzani
A. A.
,
Patel
S.
,
Galione
A.
(
2002
).
NAADP mobilizes Ca(2+) from reserve granules, lysosome-related organelles, in sea urchin eggs.
Cell
111
,
703
708
.
Clapham
D. E.
(
2007
).
Calcium signaling.
Cell
131
,
1047
1058
.
Coco
S.
,
Calegari
F.
,
Pravettoni
E.
,
Pozzi
D.
,
Taverna
E.
,
Rosa
P.
,
Matteoli
M.
,
Verderio
C.
(
2003
).
Storage and release of ATP from astrocytes in culture.
J. Biol. Chem.
278
,
1354
1362
.
Colvin
R. A.
,
Means
T. K.
,
Diefenbach
T. J.
,
Moita
L. F.
,
Friday
R. P.
,
Sever
S.
,
Campanella
G. S. V.
,
Abrazinski
T.
,
Manice
L. A.
,
Moita
C.
et al.  (
2010
).
Synaptotagmin-mediated vesicle fusion regulates cell migration.
Nat. Immunol.
11
,
495
502
.
Communi
D.
,
Robaye
B.
,
Boeynaems
J. M.
(
1999
).
Pharmacological characterization of the human P2Y11 receptor.
Br. J. Pharmacol.
128
,
1199
1206
.
Daniel
J. L.
,
Dangelmaier
C.
,
Jin
J.
,
Ashby
B.
,
Smith
J. B.
,
Kunapuli
S. P.
(
1998
).
Molecular basis for ADP-induced platelet activation. I. Evidence for three distinct ADP receptors on human platelets.
J. Biol. Chem.
273
,
2024
2029
.
Deng
T. L.
,
Yu
L.
,
Ge
Y. K.
,
Zhang
L.
,
Zheng
X. X.
(
2005
).
Intracellular-free calcium dynamics and F-actin alteration in the formation of macrophage foam cells.
Biochem. Biophys. Res. Commun.
338
,
748
756
.
Dickinson
G. D.
,
Churchill
G. C.
,
Brailoiu
E.
,
Patel
S.
(
2010
).
Deviant nicotinic acid adenine dinucleotide phosphate (NAADP)-mediated Ca2+ signaling upon lysosome proliferation.
J. Biol. Chem.
285
,
13321
13325
.
Fountain
S. J.
,
North
R. A.
(
2006
).
A C-terminal lysine that controls human P2X4 receptor desensitization.
J. Biol. Chem.
281
,
15044
15049
.
Genetos
D. C.
,
Geist
D. J.
,
Liu
D.
,
Donahue
H. J.
,
Duncan
R. L.
(
2005
).
Fluid shear-induced ATP secretion mediates prostaglandin release in MC3T3-E1 osteoblasts.
J. Bone Miner. Res.
20
,
41
49
.
Gouwy
M.
,
Struyf
S.
,
Noppen
S.
,
Schutyser
E.
,
Springael
J-Y.
,
Parmentier
M.
,
Proost
P.
,
Van Damme
J.
(
2008
).
Synergy between coproduced CC and CXC chemokines in monocyte chemotaxis through receptor-mediated events.
Mol. Pharmacol.
74
,
485
495
.
Guidarelli
A.
,
Cerioni
L.
,
Fiorani
M.
,
Cantoni
O.
(
2009
).
Differentiation-associated loss of ryanodine receptors: a strategy adopted by monocytes/macrophages to prevent the DNA single-strand breakage induced by peroxynitrite.
J. Immunol.
183
,
4449
4457
.
Haller
T.
,
Dietl
P.
,
Deetjen
P.
,
Völkl
H.
(
1996
).
The lysosomal compartment as intracellular calcium store in MDCK cells: a possible involvement in InsP3-mediated Ca2+ release.
Cell Calcium
19
,
157
165
.
Higashio
H.
,
Nishimura
N.
,
Ishizaki
H.
,
Miyoshi
J.
,
Orita
S.
,
Sakane
A.
,
Sasaki
T.
(
2008
).
Doc2 α and Munc13-4 regulate Ca(2+)-dependent secretory lysosome exocytosis in mast cells.
J. Immunol.
180
,
4774
4784
.
Hishikawa
T.
,
Cheung
J. Y.
,
Yelamarty
R. V.
,
Knutson
D. W.
(
1991
).
Calcium transients during Fc receptor-mediated and nonspecific phagocytosis by murine peritoneal macrophages.
J. Cell Biol.
115
,
59
66
.
Holmsen
H.
,
Weiss
H. J.
(
1979
).
Secretable storage pools in platelets.
Annu. Rev. Med.
30
,
119
134
.
Holt
O. J.
,
Gallo
F.
,
Griffiths
G. M.
(
2006
).
Regulating secretory lysosomes.
J. Biochem.
140
,
7
12
.
Hosoi
E.
,
Nishizaki
C.
,
Gallagher
K. L.
,
Wyre
H. W.
,
Matsuo
Y.
,
Sei
Y.
(
2001
).
Expression of the ryanodine receptor isoforms in immune cells.
J. Immunol.
167
,
4887
4894
.
Hovater
M. B.
,
Olteanu
D.
,
Hanson
E. L.
,
Cheng
N-L.
,
Siroky
B.
,
Fintha
A.
,
Komlosi
P.
,
Liu
W.
,
Satlin
L. M.
,
Bell
P. D.
et al.  (
2008
).
Loss of apical monocilia on collecting duct principal cells impairs ATP secretion across the apical cell surface and ATP-dependent and flow-induced calcium signals.
Purinergic Signal.
4
,
155
170
.
Lange
I.
,
Yamamoto
S.
,
Partida–Sanchez
S.
,
Mori
Y.
,
Fleig
A.
,
Penner
R.
(
2009
).
TRPM2 functions as a lysosomal Ca2+-release channel in beta cells.
Sci. Signal.
2
,
ra23
.
Lee
H.
,
Lee
J. K.
,
Min
W-K.
,
Bae
J-H.
,
He
X.
,
Schuchman
E. H.
,
Bae
J. S.
,
Jin
H. K.
(
2010
).
Bone marrow-derived mesenchymal stem cells prevent the loss of Niemann-Pick type C mouse Purkinje neurons by correcting sphingolipid metabolism and increasing sphingosine-1-phosphate.
Stem Cells
28
,
821
831
.
Li
J.
,
Fountain
S. J.
(
2012
).
Fluvastatin suppresses native and recombinant human P2X4 receptor function.
Purinergic Signal.
8
,
311
316
.
Li
S.
,
Tomić
M.
,
Stojilkovic
S. S.
(
2011
).
Characterization of novel Pannexin 1 isoforms from rat pituitary cells and their association with ATP-gated P2X channels.
Gen. Comp. Endocrinol.
174
,
202
210
.
Lloyd–Evans
E.
,
Morgan
A. J.
,
He
X.
,
Smith
D. A.
,
Elliot–Smith
E.
,
Sillence
D. J.
,
Churchill
G. C.
,
Schuchman
E. H.
,
Galione
A.
,
Platt
F. M.
(
2008
).
Niemann-Pick disease type C1 is a sphingosine storage disease that causes deregulation of lysosomal calcium.
Nat. Med.
14
,
1247
1255
.
Luo
S.
,
Rohloff
P.
,
Cox
J.
,
Uyemura
S. A.
,
Docampo
R.
(
2004
).
Trypanosoma brucei plasma membrane-type Ca(2+)-ATPase 1 (TbPMC1) and 2 (TbPMC2) genes encode functional Ca(2+)-ATPases localized to the acidocalcisomes and plasma membrane, and essential for Ca(2+) homeostasis and growth.
J. Biol. Chem.
279
,
14427
14439
.
MacIntyre
J. P.
,
Pope
B. L.
(
1991
).
The involvement of protein kinase C, calcium, and 5-lipoxygenase in the production of tumor necrosis factor by a cloned interleukin-3 dependent cell line with natural cytotoxic activity.
Int. J. Immunopharmacol.
13
,
175
184
.
Martinez
I.
,
Chakrabarti
S.
,
Hellevik
T.
,
Morehead
J.
,
Fowler
K.
,
Andrews
N. W.
(
2000
).
Synaptotagmin VII regulates Ca(2+)-dependent exocytosis of lysosomes in fibroblasts.
J. Cell Biol.
148
,
1141
1150
.
McLeish
K. R.
,
Dean
W. L.
,
Wellhausen
S. R.
,
Stelzer
G. T.
(
1989
).
Role of intracellular calcium in priming of human peripheral blood monocytes by bacterial lipopolysaccharide.
Inflammation
13
,
681
692
.
Meis
S.
,
Hamacher
A.
,
Hongwiset
D.
,
Marzian
C.
,
Wiese
M.
,
Eckstein
N.
,
Royer
H. D.
,
Communi
D.
,
Boeynaems
J. M.
,
Hausmann
R.
et al.  (
2010
).
NF546 [4,4′-(carbonylbis(imino-3,1-phenylene-carbonylimino-3,1-(4-methyl-phenylene)-carbonylimino))-bis(1,3-xylene-alpha,alpha’-diphosphonic acid) tetrasodium salt] is a non-nucleotide P2Y11 agonist and stimulates release of interleukin-8 from human monocyte-derived dendritic cells.
J. Pharmacol. Exp. Ther.
332
,
238
247
.
Moore
D. J.
,
Chambers
J. K.
,
Wahlin
J-P.
,
Tan
K. B.
,
Moore
G. B.
,
Jenkins
O.
,
Emson
P. C.
,
Murdock
P. R.
(
2001
).
Expression pattern of human P2Y receptor subtypes: a quantitative reverse transcription-polymerase chain reaction study.
Biochim. Biophys. Acta
1521
,
107
119
.
Mullins
C.
,
Bonifacino
J. S.
(
2001
).
The molecular machinery for lysosome biogenesis.
BioEssays
23
,
333
343
.
Muno
D.
,
Ishidoh
K.
,
Ueno
T.
,
Kominami
E.
(
1993
).
Processing and transport of the precursor of cathepsin C during its transfer into lysosomes.
Arch. Biochem. Biophys.
306
,
103
110
.
North
R. A.
(
2002
).
Molecular physiology of P2X receptors.
Physiol. Rev.
82
,
1013
1067
.
Olivier
M.
,
Baimbridge
K. G.
,
Reiner
N. E.
(
1992
).
Stimulus-response coupling in monocytes infected with Leishmania. Attenuation of calcium transients is related to defective agonist-induced accumulation of inositol phosphates.
J. Immunol.
148
,
1188
1196
.
Parr
C. E.
,
Sullivan
D. M.
,
Paradiso
A. M.
,
Lazarowski
E. R.
,
Burch
L. H.
,
Olsen
J. C.
,
Erb
L.
,
Weisman
G. A.
,
Boucher
R. C.
,
Turner
J. T.
(
1994
).
Cloning and expression of a human P2U nucleotide receptor, a target for cystic fibrosis pharmacotherapy.
Proc. Natl. Acad. Sci. USA
91
,
3275
3279
.
Patel
S.
,
Docampo
R.
(
2010
).
Acidic calcium stores open for business: expanding the potential for intracellular Ca2+ signaling.
Trends Cell Biol.
20
,
277
286
.
Pozzan
T.
,
Rizzuto
R.
,
Volpe
P.
,
Meldolesi
J.
(
1994
).
Molecular and cellular physiology of intracellular calcium stores.
Physiol. Rev.
74
,
595
636
.
Rao
S. K.
,
Huynh
C.
,
Proux–Gillardeaux
V.
,
Galli
T.
,
Andrews
N. W.
(
2004
).
Identification of SNAREs involved in synaptotagmin VII-regulated lysosomal exocytosis.
J. Biol. Chem.
279
,
20471
20479
.
Robaye
B.
,
Boeynaems
J-M.
,
Communi
D.
(
1997
).
Slow desensitization of the human P2Y6 receptor.
Eur. J. Pharmacol.
329
,
231
236
.
Rodríguez
A.
,
Martinez
I.
,
Chung
A.
,
Berlot
C. H.
,
Andrews
N. W.
(
1999
).
cAMP regulates Ca2+-dependent exocytosis of lysosomes and lysosome-mediated cell invasion by trypanosomes.
J. Biol. Chem.
274
,
16754
16759
.
Shin
H.
,
Charlton
R.
,
Mollitt
D. L.
(
1997
).
Altered monocyte calcium dynamics in sepsis.
J. Trauma
42
,
889
893
,
discussion 893–894
.
Srinivas
S. P.
,
Ong
A.
,
Goon
L.
,
Goon
L.
,
Bonanno
J. A.
(
2002
).
Lysosomal Ca(2+) stores in bovine corneal endothelium.
Invest. Ophthalmol. Vis. Sci.
43
,
2341
2350
.
von Kügelgen
I.
(
2006
).
Pharmacological profiles of cloned mammalian P2Y-receptor subtypes.
Pharmacol. Ther.
110
,
415
432
.
Zhang
F.
,
Xia
M.
,
Li
P-L.
(
2010
).
Lysosome-dependent Ca2+ release response to Fas activation in coronary arterial myocytes through NAADP: evidence from CD38 gene knockouts.
Am. J. Physiol. Cell Physiol.
298
,
C1209
C1216
.
Zhang
Q.
,
Fukuda
M.
,
Van Bockstaele
E.
,
Pascual
O.
,
Haydon
P. G.
2004a
).
Synaptotagmin IV regulates glial glutamate release.
Proc. Natl. Acad. Sci. USA
101
,
9441
9446
.
Zhang
Q.
,
Pangršič
T.
,
Kreft
M.
,
Kržan
M.
,
Li
N.
,
Sul
J-Y.
,
Halassa
M.
,
Van Bockstaele
E.
,
Zorec
R.
,
Haydon
P. G.
2004b
).
Fusion-related release of glutamate from astrocytes.
J. Biol. Chem.
279
,
12724
12733
.
Zhang
Z.
,
Chen
G.
,
Zhou
W.
,
Song
A.
,
Xu
T.
,
Luo
Q.
,
Wang
W.
,
Gu
X. S.
,
Duan
S.
(
2007
).
Regulated ATP release from astrocytes through lysosome exocytosis.
Nat. Cell Biol.
9
,
945
953
.

Supplementary information