Abstract
Calcium (Ca2+) signaling is an essential process in all cells that is maintained by a plethora of channels, pumps, transporters, receptors, and intracellular Ca2+ sequestering stores. Changes in cytosolic Ca2+ concentration govern processes as far reaching as fertilization, cell growth, and motility through to cell death. In recent years, lysosomes have emerged as a major intracellular Ca2+ storage organelle with an increasing involvement in triggering or regulating cellular functions such as endocytosis, autophagy, and Ca2+ release from the endoplasmic reticulum. This review will summarize recent work in the area of lysosomal Ca2+ signaling and homeostasis, including newly identified functions, and the involvement of lysosome-derived Ca2+ signals in human disease. In addition, we explore recent controversies in the techniques used for measurement of lysosomal Ca2+ content.
Calcium (Ca2+) is a ubiquitous primary signaling messenger that is essential for the function of all cells and the regulation of a diverse range of key cellular events including fertilization, cellular metabolism, growth, division, motility, and apoptosis (Berridge et al. 2000). Because of the ability of Ca2+ to readily precipitate with anions (e.g., carbonates, phosphates) and form insoluble salts, high Ca2+ concentrations are toxic to cells and Ca2+ homeostasis requires tight control and regulation, which has been honed during evolution (Williams 2006). The origin of the cellular use of Ca2+ as a signaling ion is likely to have come from its flexible chemistry and prevalence in ancient seawater (∼10 mm) (Williams 2006; Jozef et al. 2013). First, Ca2+ can form longer bonds than other divalent cations prevalent in seawater (e.g., Mg2+, 1–3 mm); for example, Ca2+-oxygen bonds range from 2.3 to 2.5 Å depending on whether they are monodentate or bidentate, whereas Mg2+-oxygen bonds are almost always 2.05 Å as Mg2+ will be surrounded by an octahedron of oxygen atoms (Bertini 2007). Furthermore, Ca2+ can form six to eight bonds, which have a more rapid rate of substitution of water molecules by 103 compared with Mg2+ (Katz et al. 1996; Bertini 2007). It is therefore clear why early unicellular organisms would use Ca2+ over Mg2+ as a signaling ion. The presence of high extracellular free Ca2+ concentration, which is maintained in the bodily fluids of higher order organisms (e.g., ∼1–1.3 mm in human plasma; Harris et al. 2014), versus low intracellular Ca2+ concentration (∼100 nm in the cytosol; Berridge et al. 2000) necessitated the emergence of mechanisms to maintain this concentration gradient and avoid cellular toxicity, for example, the development of Ca2+ pumps such as plasma membrane (PM) Ca2+ ATPases (PMCAs) and Na+/Ca2+ exchangers (Williams 2006).
INTRACELLULAR AND ORGANELLAR Ca2+ HOMEOSTASIS
In addition to extrusion of Ca2+ out of cells to maintain cytosolic Ca2+ in the range of 100 nm, intracellular Ca2+ stores function to buffer and induce orders of magnitude changes in cytosolic-free Ca2+ concentration. Ca2+ levels within the nuclear and mitochondrial matrix are similar to the cytosol (Bagur and Hajnóczky 2017), at least at times when cells are not stressed (Celsi et al. 2009). The endoplasmic reticulum (ER), however, is a major intracellular Ca2+ store with estimations of intraorganellar Ca2+ ranging from 12 µm to 2000 µm (reviewed in Bygrave and Benedetti 1996). This well-defined Ca2+ store is maintained by a plethora of Ca2+ transport mechanisms, chief among these being the inositol 1,4,5-trisphosphate receptors (IP3 receptors) and the ryanodine receptors (RyRs) (Takeshima et al. 2015), Ca2+ leak channels (e.g., Bax inhibitor 1, presenilin 1; Bultynck et al. 2014; Takeshima et al. 2015), and one major Ca2+ uptake pump, the sarcoplasmic reticulum (SR)/ER Ca2+ ATPase (SERCA) (Takeshima et al. 2015). Ca2+ release from the ER is mediated mainly by the action of second messengers including IP3 and cyclic ADP-ribose (cADPr), which are typically generated from extracellular signals binding to G-protein-coupled receptors that activate enzymes, including phospholipase C or adenylyl cyclase (Galione and Churchill 2002). Ca2+ release from the ER can be amplified by processes including direct coupling of the SR Ca2+ channels to PM Ca2+ channels, or by Ca2+-induced Ca2+ release (CICR) (Galione and Churchill 2002). The estimated cellular volume of the ER is in excess of 10% (Alberts 2002), which allows for substantial cellular coverage and Ca2+ release within the cell. The importance of controlling and maintaining the Ca2+ content of organelles and the cytosol is most clearly shown via a plethora of human diseases. In certain lysosomal diseases (discussed in more detail below), the normally very tightly controlled ER Ca2+ release has been reported to go awry (Pelled et al. 2003, 2005). This leads to elevated concentrations of Ca2+ in the cytosol, which is buffered by mitochondria (Duchen 2000; Sano et al. 2009). Ultimately, the combined ER Ca2+ dysfunction coupled to prolonged elevation in mitochondrial Ca2+ triggers neuronal dysfunction and death (Duchen 2000; Tessitore et al. 2004).
In addition to the ER, a substantial amount of free Ca2+ (existing in an unbound state) has been measured in the endocytic system (Lloyd-Evans et al. 2010). Although endocytosis engulfs extracellular fluid, which has an estimated 1–2 mm free Ca2+, the initialization of the process of acidification of the endocytic system organelles via H+ transport by the vacuolar ATPase (vATPase) leads to the rapid extrusion of Ca2+. This process is thought to be mediated, at least in part, by the action of the transient receptor potential (TRP) mucolipin 3 (TRPML3) ion channel (Lelouvier and Puertollano 2011). This results in an estimated 5–40 µm free Ca2+ within the early endocytic system (Gerasimenko et al. 1998). In contrast to early endosomes, the intravesicular Ca2+ content inside lysosomes, which are the terminal point of the endocytic system, is considerably higher (see below) (Christensen et al. 2002; Lloyd-Evans et al. 2008). Indeed, over the last 10 to 15 years, lysosomes, or the so-called “acidic stores,” which encompass late endosomes, fused intermediates and lysosome-related organelles, have emerged as a new and important intracellular Ca2+ store (Patel and Cai 2015).
LYSOSOMAL Ca2+ AND ION HOMEOSTASIS
Lysosomes, and vacuoles in simpler organisms, are essential acidic organelles found in all nucleated cells, serving as centers of macromolecular degradation and recycling, endocytic recycling, cellular nutrient regulation, and as a critical Ca2+ signaling organelle for the cell (Saftig and Klumperman 2009; Lloyd-Evans and Platt 2011; Settembre et al. 2013). The lysosome is the second-largest store of Ca2+ within the mammalian cell with a free Ca2+ concentration in the region of 500 µm–600 µm in multiple mammalian cells (Christensen et al. 2002; Lloyd-Evans et al. 2008). The Ca2+ concentration within the lysosome lumen has been measured using endocytosed luminal Ca2+-binding fluorophores conjugated to high molecular weight dextran. In mammals, lysosomes typically comprise 2%–3% of the volume of the cell, which suggests that, although lysosomes have a high intraluminal Ca2+ concentration, their overall contribution to global cellular Ca2+ signaling is relatively low in comparison to the ER (Lloyd-Evans 2016). This likely also explains the difficulty in measuring lysosomal Ca2+ indirectly using Ca2+-sensitive fluorophores (Lloyd-Evans 2016), which is expanded on in detail below. However, lysosomes have been shown to initiate global changes in cellular Ca2+ signaling, which may also explain discrepancies in measuring lysosomal Ca2+ levels (Shen et al. 2012; Vienken et al. 2017; Atakpa et al. 2019), also discussed in more detail below. In comparison, simpler organisms with single vacuoles (lysosome-like organelles), such as yeast and plants, have had their vacuolar free Ca2+ concentration measured using electrodes, resulting in recordings ranging between 0.2 µm and 2.3 mm (Schönknecht 2013). These measurements are in agreement with other recordings in Saccharomyces cerevisiae using the Ca2+-sensitive fluorophore Indo-1 (1.3 mm) (Halachmi and Eilam 1989). In contrast, total Ca2+ levels have been estimated in plant vacuoles, mainly using X-ray elemental microanalysis, as being considerably higher, in the range of 60–70 mm in Eudicot mesophyll cells (Conn and Gilliham 2010). The high values for vacuolar free Ca2+ concentration support the measurements in mammalian cells, and clearly show that these organelles can maintain high concentrations of free Ca2+ within this acidic environment. Unsurprisingly, the Ca2+ concentration is higher in vacuoles of simple organisms compared with mammalian lysosomes as the vacuole is one of the largest organelles in these organisms (e.g., yeast) comprising 10%–20% of total cellular volume (Armstrong 2010). Critical to intravesicular lysosomal Ca2+ quantitation is the ability to also measure pH in the same experiment (Christensen et al. 2002; Lloyd-Evans et al. 2008). pH has a critical effect on the level of free ionized Ca2+; in alkaline conditions, Ca2+ precipitates more freely with proteins, whereas acidic pH decreases protein binding leading to increased free Ca2+ levels (Wang et al. 2002). This supports the observations that within lysosomes there is ∼5000–6000 times the amount of free Ca2+ compared with the cytosol (Christensen et al. 2002; Lloyd-Evans et al. 2008). In comparison, lysosomes have ∼1000-fold more H+ ions, in the region of 2 times more Na+ (20 mm compared with 8–12 mm in the cytosol; Lodish 2000; Steinberg et al. 2010) and up to 3 times less K+ (50 mm compared with 140–150 mm in the cytosol; Lodish 2000; Steinberg et al. 2010) than the equivalent cytosolic concentrations of these ions. The vastly greater concentration gradient for Ca2+ across the lysosomal membrane is a further indication of the importance of this organelle in localized and global intracellular signaling.
Lysosomal Ca2+ Store Filling
For lysosomes to have a high concentration of Ca2+, it is likely that they have mechanisms for transporting Ca2+ from the cytosol, or, as recently suggested, from the ER into the lysosome lumen (Garrity et al. 2016). A variety of mechanisms mediating this process, detailed below, have been suggested, but no actual candidate transporters or pumps have yet been identified in placental mammals (Melchionda et al. 2016). In lower order organisms with vacuoles, Ca2+ uptake is an energy-dependent process with both Ca2+/H+ exchangers (CAX) and Ca2+ ATPases present on the vacuolar membrane (Pittman 2011). P-type Ca2+ ATPases, present across all life, are high-affinity, low-turnover transporters that sequester Ca2+ from the cytosol, whereas Ca2+ exchangers are low-affinity, high-capacity transporters (Pittman 2011). In yeast and plants, it is the acidification of vacuoles by vATPase proton pumps that generates a sufficient proton gradient to drive an exchange of Ca2+ ions for H+ (Pittman 2011). CAX genes have been identified in all species up to nonplacental mammals (Melchionda et al. 2016), indicating that further mechanisms exist for lysosomal Ca2+ store filling in mammals. In mammalian cells, the transport of Ca2+ into lysosomes requires ATP, and is inhibited by heavy metals and potentially by the long-chain base sphingosine (Lemons and Thoene 1991; Adachi et al. 1996; Colina et al. 2002; Lloyd-Evans et al. 2008). Furthermore, a recent study has suggested that acidification may not be necessary to allow Ca2+ entry into lysosomes and that instead the ER is the most critical cellular source of Ca2+ to ensure normal lysosomal Ca2+ store filling (Garrity et al. 2016). Despite this, the majority of studies have indicated that maintenance of resting Ca2+ levels within lysosomes, and indeed vacuoles, is dependent on maintaining vATPase activity (Christensen et al. 2002; Pittman 2011). Potential candidates for lysosomal transporters mediating Ca2+ entry include the TBHQ-inhibitable SERCA3, identified as a lysosomal Ca2+ transporter in platelet lysosomes (López et al. 2008), and SLC24A5, a Na+/K+/Ca2+ exchanger found on melanosomes, which are lysosome-related organelles responsible for pigmentation (Lamason et al. 2005). These findings could suggest that there are multiple mechanisms for lysosomal Ca2+ uptake that are cell-type and specialized-lysosome dependent. It should also be noted that extracellular Ca2+ contributes minimally to late endosomal/lysosomal Ca2+ content as chelation of extracellular Ca2+ had no significant long-term impact on intralumenal lysosomal Ca2+ levels (Christensen et al. 2002).
Local Requirement for Lysosomal Ca2+ Signaling
The process of endocytosis is critical to all cells and is regulated by a host of machinery and signals, one of which is Ca2+. Indeed, in yeast, it has been shown that before membrane tethering by SNARE proteins there are three key requirements to allow membrane fusion: (1) release of Ca2+ from the vacuole, (2) calmodulin, and (3) protein phosphatase 1 (Peters and Mayer 1998; Peters et al. 1999). Yeast mutants deficient in the ability of calmodulin to bind Ca2+, but are otherwise functional, can release Ca2+ from vacuolar stores but cannot trigger vesicular fusion, illustrating the critical requirement for Ca2+-bound calmodulin (Ca2+/calmodulin) as a sensor in mediating fusion events (Peters and Mayer 1998). In mammalian cells and cell-free systems, it has been shown that an approximate elevation of 0.5 µm cytosolic Ca2+ in the vicinity of endosomes, lysosomes, and autophagosomes is an absolute essential component mediating trafficking and fusion between these compartments (Gordon et al. 1993; Pryor et al. 2000; Lloyd-Evans et al. 2008). This is in contrast to the effect of high Ca2+ concentrations (0.1–1 mm), which inhibits endocytosis (Pryor et al. 2000). Furthermore, fusion between purified late endosomes is inhibited by the action of the fast Ca2+ chelator BAPTA, but not the slow Ca2+ chelator EGTA (Pryor et al. 2000). Taken together, these data indicate that only local changes in Ca2+ in the vicinity of lysosomes governs lysosomal transport and fusion events. As with yeast, in mammalian cells, one of the key effectors of vesicular fusion that uses this local elevation in Ca2+ to complete vesicular docking and promote fusion is calmodulin (Colombo et al. 1997; Peters and Mayer 1998; Pryor et al. 2000). Calmodulin inhibitors (e.g., by W7, calmidazolium, and chlorpromazine; Masson et al. 1992) have been shown to retard endosome fusion and alter cellular lipid metabolism in a manner that resembles the lysosomal storage disease (LSD), Niemann-Pick C1 (NPC1) (Masson et al. 1992), in which lysosomal Ca2+ levels and release are reduced (discussed below) (Lloyd-Evans et al. 2008). It has been suggested that the mechanisms by which Ca2+/calmodulin mediates vesicular fusion likely involves SNARE proteins that promote vesicle association through tethering (Burgoyne and Clague 2003; Luzio et al. 2005). Of these, syntaxin 13 is a likely candidate as a direct interaction with Ca2+/calmodulin has been shown and it has previously been implicated in early endosome fusion (Mills et al. 2001).
Further evidence that lysosomal transport and fusion events require the release of Ca2+ from the lysosome lumen have come from studies into lysosomal diseases (reviewed below) and lysosomal ion channel function, particularly the two-pore channels (TPCs). Overexpression of TPC1, TPC2, and TPC3 in mammalian cells leads to potentiated (TPC1 and TPC2) and inhibited (TPC3) endolysosomal Ca2+ release in response to a membrane permeant version of the lysosomal Ca2+ releasing second messenger nicotinic acid adenine dinucleotide phosphate (NAADP-AM) (Ruas et al. 2010). In all three cases, the endocytosis of lipids and their transport between the late endosomal system and the Golgi was impaired with endocytosed ganglioside GM1 bound to the B subunit of cholera toxin (a specific GM1 ganglioside marker) accumulating in the lysosomes of the TPC-overexpressing cells compared with Golgi localization in the controls (Ruas et al. 2010). This was accompanied by lysosomal expansion and intra-lysosomal accumulation of multiple lipids in the TPC1- and TPC2-overexpressing cells (Ruas et al. 2010), mimicking the phenotypes observed in Niemann-Pick C (NPC) disease cells, presumably as a result of lowered intralysosomal Ca2+ content (Lloyd-Evans et al. 2008). Further evidence of the important role of maintaining normal lysosomal Ca2+ signaling and lysosomal ion channel function comes via the TRPML1 and CACNA1A lysosomal Ca2+ channels. Alterations in the functions of these channels has been shown to impair endolysosomal–autolysosomal transport and fusion events, and are discussed in more detail below. It is therefore clear that maintaining physiological Ca2+ homeostasis and release from lysosomal/vacuolar Ca2+ stores is critical for the normal function of the endolysosomal system.
Global Requirement for Lysosomal Ca2+ Signaling
Membrane contact sites between organelles, for example, the ER and mitochondria (Patergnani et al. 2011) and the ER and the PM (Elbaz and Schuldiner 2011), have been well studied in terms of Ca2+ signaling and the potential for amplification of Ca2+ signals (Penny et al. 2015). Contact sites between lysosomes and the ER exist but are not yet particularly well characterized. However, there appears to be a key role for ER–lysosome contact sites in potentiating local lysosomal Ca2+ release events into global Ca2+ release events (Fig. 1; Kilpatrick et al. 2013; Morgan et al. 2013) and in providing a source of Ca2+ to refill the depleted lysosomal Ca2+ store (Garrity et al. 2016; Atakpa et al. 2018). (For a detailed review of ER–lysosome contact points, see Penny et al. 2015.)
Figure 1.
Interplay between lysosomal and endoplasmic reticulum (ER) Ca2+ release. Lysosomes and the ER are now well known to have contact sites that bring them together in physical space. This ensures that Ca2+ uptake and release from lysosomes is further modulated by the ER. Indicated are different examples of endogenous (1,2) and pharmacological (3,4) release of Ca2+ from lysosomal stores and the subsequent impact that this may have on Ca2+ release from the ER. Movement of Ca2+ ions (blue for lysosomal Ca2+, red for ER Ca2+) is indicated by the arrows either through channels (rectangles and triangles) or via lysosomal membrane perforation induced by lysosomal cathepsin-mediated hydrolysis of Gly-Phe-β-naphthylamide (GPN).
LYSOSOMAL Ca2+ CHANNELS
In recent years, a plethora of ion channels have been identified as functioning to release Ca2+ from lysosomes, chief among these, and the most well characterized, are the TPCs and TRPML1. TPCs have been extensively reviewed elsewhere, but it is worth reiterating that a recent controversy surrounding the ion selectivity of TPC2 has been addressed with this ion channel clearly releasing Ca2+ in response to the second messenger NAADP (Ruas et al. 2015). Other Ca2+ permeable channels found in lysosomes include the neuronal voltage-gated Ca2+ channel CACNA1A (Tian et al. 2015), and the ATP-gated multi-ion channel P2X4 (Huang et al. 2014; Murrell-Lagnado 2018). The mechanisms by which these channels function in the lysosome is not well understood. CACNA1A, a voltage-gated Ca2+ channel, has been identified to reside on lysosomes where it is thought to mediate neuron-specific lysosomal fusion with endosomes and autophagic vacuoles (Tian et al. 2015). P2X4 on the other hand is inhibited at low pH and is therefore not active in lysosomes, but is thought to become active on induction of lysosomal exocytosis, for example, to facilitate secretion of surfactant by alveolar type II cells (Murrell-Lagnado 2018), or phagocytosis, in which it cooperates with PM P2X7 to facilitate killing of mycobacteria (Fairbairn et al. 2001; Qureshi et al. 2007). In this review, we are going to predominantly focus on another lysosomal ion channel, namely TRPML1.
TRPML1
TRPML1, or mucolipin-1, is a six-transmembrane domain channel belonging to the TRP superfamily and located on late endosomes and lysosomes (Waller-Evans and Lloyd- Evans 2015). It is evolutionarily conserved in Drosophila and Caenorhabditis elegans (Treusch et al. 2004; Wong et al. 2012).
TRPML1 Ion Selectivity
Despite being referred to as the major Ca2+ efflux channel of the lysosome, TRPML1 has actually been reported to be permeable to several additional cations including H+, K+, Na+, Fe2+, Mn2+, and Zn2+ (Waller-Evans and Lloyd- Evans 2015). TRPML1 was first identified as an inwardly rectifying (i.e., ions move from the lysosome to the cytoplasm) cation channel permeable to Ca2+, K+, and Na+ by LaPlante et al. (2002) using Xenopus oocytes overexpressing human TRPML1. K+ permeability has also been observed by Raychowdhury et al. (2004), using TRPML1 in planar lipid bilayers, and Kiselyov et al. (2005), using whole-cell recordings of HEK293T cells overexpressing TRPML1. It has also been suggested that TRPML1 acts as a lysosomal H+ leak channel (Soyombo et al. 2006).
Following generation of a constitutively active TRPML1, based on the activating varitint-waddler TRPML3 mutation, by Xu et al. (2007) and Grimm et al. (2007), Ca2+ permeability has been confirmed several times (Dong et al. 2010; Yamaguchi et al. 2011; Shen et al. 2012; Chen et al. 2017; Schmiege et al. 2017). This variant has also been used to detect Fe2+ (but not Fe3+), Zn2+, and Mn2+ permeability via patch-clamping of lysosomes swollen using vacuolin (Dong et al. 2008). However, as the effects of vacuolin on endosomal membranes and ion content have yet to be determined, these experiments may not reflect TRPML1 ion selectivity under physiological conditions. There is some supporting evidence in that anemia is common among patients suffering from mucolipidosis type IV (caused by loss of TRPML1 function), although this could be caused by reduced absorption through the gut (Chen et al. 2014). Additionally, Zn2+ homeostasis is altered in the absence of TRPML1 (Eichelsdoerfer et al. 2010; Kukic et al. 2013; Chen et al. 2014). However, these experiments were performed with nonphysiologically high concentrations of extracellular Zn2+ so, again, may not reflect normal physiology. In summary, there is substantial evidence that TRPML1 is permeable to Ca2+ and K+, but it may also be permeable to other cations.
TRPML1 Regulation
Phosphatidylinositols, a group of signaling lipids, have been identified as the native agonists and antagonists of TRPML1. Phosphatidyl-(3,5)-bisphosphate (PI(3,5)P2), an endolysosome-specific phosphatidylinositol generated from phosphatidyl-(3)-phosphate (PI3P) by PIK-fyve, activates TRPML1, while PI(4,5)P2, which is abundant at the PM, inhibits TRPML1, as do PI(3,4)P2 and PI(3,4,5)P3 (Dong et al. 2010). A motif rich in basic amino acids in the amino terminus of TRPML1 is necessary for phosphatidylinositol-mediated regulation (Zhang et al. 2012). TRPML, the Drosophila TRPML1 ortholog, is also activated by PI(3,5)P2 and inhibited by PI(4,5)P2 (but not by other phosphatidylinositols) when expressed in HEK293T cells, suggesting an evolutionarily ancient regulatory mechanism (Feng et al. 2014a). It is very easy to see how this activation and inhibition by different phosphatidylinositols found in different cellular compartments could prevent aberrant triggering of TRPML1 at the PM, where it is occasionally found following lysosomal exocytosis (Medina and Ballabio 2015).
Several synthetic agonists and antagonists have been developed for TRPML1, with MLSA1 (mucolipin synthetic agonist 1) the best known. This was identified by Shen et al. (2012), who screened analogs of SF-51, a TRPML3 agonist identified in an earlier high-throughput screen (Grimm et al. 2010), using changes to whole-cell and whole-endosome currents in HEK293T cells overexpressing TRPML1. MLSA1 interacts directly with the pore region of TRPML1 and forces it into an open conformation (Fine et al. 2018). This is in contrast to the endogenous ligand, PI(3,5)P2, which acts as an allosteric activator. There is significant synergy between MLSA1 and PI(3,5)P2, with TRPML1 more sensitive to MLSA1 activation in the presence of PI(3,5)P2. There are also differences in the way MLSA1 affects mammalian and Drosophila TRPML1. Although MLSA1 can directly activate mammalian TRPML1, even in the absence of PI(3,5)P2, it can only act to increase the sensitivity of Drosophila TRPML to PI(3,5)P2 (Feng et al. 2014b). Since the discovery of MLSA1, several other TRPML1 agonists have been discovered (Wang et al. 2015; Zhang et al. 2016), as have antagonists (Samie et al. 2013; Wang et al. 2015), although the mechanism of action of these compounds is unknown.
In addition to activation and inhibition by phosphatidylinositols, TRPML1 is subject to several other layers of regulation. It is regulated by pH, although there is contradictory evidence as to whether TRPML1 is more active in an acidic environment, or is inhibited by it. Earlier single-channel studies by LaPlante et al. (2002) and Raychowdhury et al. (2004) found that TRPML1 conductance was inhibited at low pH. However, later studies using the constitutively active varitint TRPML1 mutant have reported increased activation of TRPML1 at a more acidic pH (Dong et al. 2008; Feng et al. 2014a), with optimal TRPML1 activity at pH4.5 followed by inhibition with increasing acidity. Interestingly, the same effect has been seen with Drosophila TRPML, but with a slightly higher pH optimum of 5.2 (Feng et al. 2014a). These pH optima correspond with the lysosomal pH of mammals and Drosophila, respectively, suggesting that TRPML1 is most active in lysosomes (Venkatachalam et al. 2015). Recent structural studies identified aspartate residues in the luminal pore that are important for pH regulation. These are thought to be protonated at low pH, which would prevent the inhibition of TRPML1 by high luminal Ca2+ concentrations that occurs at neutral pH (Li et al. 2017). These structural studies therefore support the idea that TRPML1 is more likely to be active in an acidic environment such as the lysosome.
As well as direct action of protons on the channel, TRPML1 is indirectly regulated by pH as it is inactivated through cathepsin B-dependent cleavage (Kiselyov et al. 2005). Addition of purified cathepsin B to TRPML1-overexpressing HEK293T cells drastically reduced TRPML1-mediated current and the amount of full-length TRPML1 present in cells. As cathepsin B is a lysosomal enzyme with a pH optimum of 4.5–5.5 (Linebaugh et al. 1999; Lloyd-Evans and Haslett 2016), it is therefore unlikely to be involved in inactivation of TRPML1 at the PM, but rather to regulate TRPML1 activity within lysosomes, possibly by restricting the time during which TRPML1 is active (Kiselyov et al. 2005). Finally, TRPML1 has been suggested to be inhibited by high lysosomal concentrations of sphingomyelin (Shen et al. 2012) and adenosine (Zhong et al. 2017), although the relevance of this inhibition in normal physiological function is not clear. What is clear from the multiple layers of regulation is that it is very important for the cell to tightly control TRPML1 activity.
Cellular Function of TRPML1
One of the major cellular roles for TRPML1 is as a regulator of autophagy (Di Paola et al. 2018). Autophagy is the process by which defective organelles and macromolecules are engulfed by specialist membrane-bound compartments called autophagosomes. These fuse with late endosomes to form amphisomes, which then fuse with lysosomes where their contents can be degraded. Autophagy is a constitutive process needed to maintain organelle quality control, but is a vital component in the cellular response to stress, allowing the cell to use itself as a nutrient source (Onyenwoke et al. 2015). Defects in autophagy have been observed in C. elegans, Drosophila, and mammalian models following loss of TRPML1 function, as well as in mucolipidosis type IV patients (Venkatachalam et al. 2008; Vergarajauregui et al. 2008; Curcio-Morelli et al. 2010; Sun et al. 2011). Amphisome-like structures are observed accumulating within cells, suggesting that TRPML1 is needed for amphisome–lysosome fusion.
TRPML1 interacts with the major regulators of autophagy, mammalian target of rapamycin complex 1 (mTORC1) and transcription factor EB (TFEB) (Di Paola et al. 2018). The lysosomal membrane amino acid sensor mTORC1 suppresses autophagy under basal conditions. In the absence of TRPML1, mTORC1 activity is diminished, causing increased formation of autophagosomes and build-up of amphisomes. Reactivation of mTORC1 can alleviate this amphisome accumulation, even in the absence of TRPML1 (Wong et al. 2012). As well as regulating mTORC1 activity, TRPML1 is regulated by mTORC1, and can be directly phosphorylated by mTOR in vivo, which is thought to result in inhibition of TRPML1 activity under basal conditions (Onyenwoke et al. 2015). This is supported by evidence that TRPML1 is up-regulated following starvation, or initiation of autophagy by Torin treatment, which inhibits mTORC1 (Wang et al. 2015). TRPML1 activity is also needed for the translocation of TFEB to the nucleus that occurs on induction of autophagy (Medina et al. 2015). This process is mediated by calcineurin. Ca2+ released via TRPML1 activates calcineurin, which dephosphorylates TFEB. This dephosphorylation promotes nuclear translocation of TFEB. TFEB is a master regulator of lysosomal and autophagy genes, and movement of TFEB to the nucleus up-regulates lysosome biogenesis and autophagy genes, including TRPML1 (Palmieri et al. 2011). It is suggested that nuclear translocation of TFEB during autophagy, dependent on TRPML1 activity, is responsible for the up-regulation of TRPML1 during autophagy, in another example of reciprocal regulation (Wang et al. 2015). It is clear that TRPML1 is intimately involved in autophagy via a number of complex regulatory mechanisms. (For a more detailed review on TRPML1 in autophagy, see Di Paola et al. 2018.)
Another major cellular role for TRPML1 is in lysosomal exocytosis, which is the fusion of lysosomes with the PM, important for processes including PM repair and phagocytosis (Samie and Xu 2014). Lysosomal exocytosis is controlled by TFEB. Overexpression of TFEB causes lysosomes to move close to the PM along microtubules, and also increases lysosomal Ca2+ release, needed for lysosome–PM fusion, via up-regulation of TRPML1 (Medina et al. 2011; Samie and Xu 2014). TFEB cannot induce lysosomal exocytosis in the absence of TRPML1. This has knock on consequences for phagocytosis, which requires the extra membrane derived from lysosomal exocytosis to engulf material (Samie et al. 2013). Potentially related to its role in lysosomal exocytosis, TRPML1 has also been shown to be required for gastric acid secretion by parietal cells in the stomach (Chandra et al. 2011; Sahoo et al. 2017). TRPML1 is located on tubulovesicles, specialized organelles found in these cells that are rich in the K+/H+ ATPase that pumps acid into the stomach following stimulation. On stimulation of G-protein-coupled receptors on the PM of parietal cells, the tubulovesicles fuse with the extensive secretory network, canaliculi, to secrete acid. This process, in a similar manner to lysosomal exocytosis, is dependent on TRPML1-mediated Ca2+ release, and can be induced by directly activating TRPML1 (Sahoo et al. 2017). It is likely that this failure to fuse tubulovesicles with canaliculi underlies the achlorydia observed in mucolipidosis type IV patients (Schiffmann et al. 1998).
LYSOSOMAL Ca2+ SIGNALING IN DISEASE
Lysosomal Storage Diseases
Lysosomal storage disorders are a group of individually rare inherited metabolic disorders that together are the most common cause of childhood neurodegeneration with a combined frequency of 1:5,000 live births (Cox and Cachón-González 2012). These diseases are predominantly caused by mutations in genes encoding lysosomal enzymes, or their accessory proteins, leading to primary intralysosomal accumulation of the enzyme substrate, and a plethora of downstream alterations in cellular function that ultimately result in cell death and neuronal loss (Cox and Cachón-González 2012). In addition, a second group of lysosomal diseases is caused by mutations in lysosomal transmembrane proteins that include transporters, ion channels, and proteins of unknown function (Cox and Cachón-González 2012). Although the precise function of many of these proteins remains to be elucidated, almost all associated diseases are characterized by similar changes in cell biology. These include intralysosomal accumulation of various substrates including lipids and proteins, and defects in endocytosis, autophagy, and mitochondrial function (Platt and Walkley 2004; Cox and Cachón-González 2012). In addition, changes in Ca2+ signaling has emerged as a contributing factor in the pathophysiology of these diseases (Vitner et al. 2010; Lloyd-Evans 2016). Earlier work indicated a role for abnormal ER Ca2+ signaling in the pathophysiology of Gaucher disease, Sandhoff disease, and GM1 gangliosidosis (Pelled et al. 2003, 2005; Tessitore et al. 2004). In these diseases, the accumulating lysosomal lipid (e.g., glucosylceramide in Gaucher disease) is presumed to exit the lysosome and is delivered to the ER, where they interfere with the function of ER Ca2+ channels (Vitner et al. 2010). Although some evidence exists for this mechanism, another potential explanation may be the potential for lysosomal storage lipids to move between lysosomes and other organelles including peroxisomes and the ER via the contact sites discussed above. For example, syntaxin 7 has recently been shown to assist cholesterol transport between lysosomes and peroxisomes (Chu et al. 2015). Perhaps these transport pathways could be hijacked by other lipids as well.
Niemann–Pick Disease Type C
It is only in the last decade that lysosomal Ca2+ signaling has emerged as a contributor in the pathogenic mechanisms underlying human disease, with the lysosomal disease NPC1 providing the first key evidence for a role of altered lysosomal Ca2+ levels and signaling in contributing to pathogenesis (Lloyd-Evans et al. 2008). NPC disease is predominantly a childhood disease in which patients present with a plethora of symptoms including ataxia, supranuclear gaze palsy, hallmarks of Alzheimer's pathology, and speech and swallowing impairment (Lloyd-Evans and Platt 2010). At the cellular level, almost all cells and tissues present with characteristic intralysosomal accumulation of lipids including free unesterified cholesterol, sphingolipids, and lyso-(bis)phosphatidic acid (LBPA), as well as defects in endocytic trafficking (Lloyd-Evans and Platt 2010). In 2008, Lloyd-Evans and colleagues reported reduced lysosomal Ca2+ levels and altered lysosomal Ca2+ signaling mediated by NAADP in mouse, human, and hamster cells either null for or carrying mutations in the NPC1 protein, as well as cells in which NPC1 function had been inhibited using the chemical U18666a. These defects in lysosomal Ca2+ in NPC disease were measured by both a direct in situ approach using Ca2+-sensitive fluorophores conjugated to dextran that had been calibrated for the approximate lysosomal pH as well as an indirect method of perforating the lysosomes with the small molecule cathepsin C substrate Gly-Phe-β-naphthylamide (GPN) and measuring the subsequent cytosolic Ca2+ elevation. Both methods supported one another and were further strengthened by the presence, in the NPC disease cells, of (1) reduced NAADP-mediated lysosomal Ca2+ release, (2) an observed reduction in lysosomal Ca2+ uptake following emptying of the ER store with thapsigargin, and (3) a complete phenocopy of NPC disease-like lysosomal storage in healthy cells following chelation of intraluminal Ca2+. The reduced lysosomal Ca2+ content and release in NPC disease cells was identified as the cause of the documented retardation in endocytic trafficking, and contributed to the resulting lipid storage characteristic of this disease. The lysosomal Ca2+-signaling defect could be compensated for following treatment with the weak SERCA antagonist curcumin, which elevated cytosolic Ca2+, reduced lipid storage, and improved function in Npc1−/− mice and astroglial cells. These data indicate that the lysosomal Ca2+ defect contributes directly to NPC disease pathogenesis. Subsequent reports have shown that the NPC disease lysosomal Ca2+ transport and signaling defects contribute to abnormalities in immune system function, neuronal function, can be used as a drug screening phenotype and can be ameliorated using an adenosine A2A receptor agonist that elevates cytosolic Ca2+ (Lee et al. 2010; Xu et al. 2012; Visentin et al. 2013; Speak et al. 2014).
Juvenile Neuronal Ceroid Lipofuscinosis/CLN3 Disease
Since the discovery of altered lysosomal Ca2+ transport and storage in NPC disease other lysosomal diseases have also been associated with aberrant lysosomal Ca2+ homeostasis (Lloyd-Evans et al. 2010; Lloyd-Evans 2016). However, only one lysosomal disease has been associated with elevated lysosomal Ca2+, namely, juvenile neuronal ceroid lipofuscinosis (CLN3 disease) (Chandrachud et al. 2015). CLN3 disease is caused by mutations in the CLN3 gene encoding the CLN3 protein, a six-transmembrane protein of the lysosome-limiting membrane of currently unknown function (Mole et al. 2011). Patients present with blindness, seizures, walking abnormalities, and neurodegeneration with death usually occurring in the second to third decades of life (Mole et al. 2011). At the cellular level, CLN3 patient cells present with intralysosomal accumulation of autofluorescent lipofuscin-like aggregating material that is characterized by the presence of subunit C of the mitochondrial ATP synthase (Mole et al. 2011). Defects in endocytic transport, autophagy, mitochondrial, and ER function have all been described, although the precise mechanisms leading from cellular dysfunction to neuronal loss remain poorly understood. Recent work from our research group has shown the presence of a number of Ca2+ signaling defects in Cln3 mutant mouse cerebellar cells carrying the common human disease-causing 1 kb deletion in exon 7–8 (Chandrachud et al. 2015). Most interesting is that these cells have elevated resting lysosomal Ca2+ measured using GPN, which we have confirmed by in situ Ca2+-sensitive dextran-conjugated probes (M Walker and E Lloyd-Evans, unpubl.), a phenotype that we have also observed in human CLN3 disease patient fibroblasts (H Waller-Evans, AS Alshehri, and E Lloyd-Evans, unpubl.). Although the cause of this elevated lysosomal Ca2+ remains to be elucidated, it may contribute to the abnormally enhanced ER Ca2+ release we observed in the Cln3 disease cerebellar cells in which either (1) low concentrations of thapsigargin elicited greater Ca2+ leak from the ER, or (2) caffeine induced considerably greater Ca2+ release from the ER RyR. These abnormalities in Ca2+ signaling in CLN3 disease had a clear effect on stimulating autophagy, leading to considerable accumulation of autophagosomes within the cells. How elevated lysosomal Ca2+ in CLN3 disease affects lysosomal Ca2+ release and global Ca2+ remains to be elucidated, but recent reports indicate alterations in both glial and neuronal Ca2+ signaling in CLN3 disease (Parviainen et al. 2017) and other NCLs, including CLN1 (Lange et al. 2018). Furthermore, a number of studies have suggested that Ca2+-channel modulators could be effective treatments for CLN3 disease (An Haack et al. 2011; Warnock et al. 2013). It is clear that further work is required to fully elucidate the role of lysosomal, ER, and global Ca2+ signaling on CLN3 disease pathogenesis, but it would appear that Ca2+ plays an important role in the pathogenesis of this disease in some way.
Lysosomal Ca2+ Homeostasis Defects in Diseases of Aging
Lysosomal Ca2+-signaling abnormalities have now been reported in a number of diseases of aging, including Parkinson's disease (PD), Alzheimer's disease (AD), and frontotemporal dementia (FTD) (Bae et al. 2014; Schondorf et al. 2014; Lee et al. 2015; Zou et al. 2015), and are described in detail elsewhere (Lloyd-Evans and Haslett 2016). Briefly, alterations in lysosomal Ca2+ homeostasis have been described in various forms of PD, including reduced lysosomal Ca2+ levels caused by mutations in the GBA1 gene (Sidransky et al. 2009; Kilpatrick et al. 2016a) (whose complete loss-of-function leads to Gaucher disease, the most common LSD; Cox and Cachón-González 2012) and also an involvement of TPC2 channels in PD caused by mutations in the LRRK2 gene (Hockey et al. 2015). Regarding AD, reduced lysosomal Ca2+ levels (Coen et al. 2012; Lee et al. 2015) and altered Ca2+ signaling (Lee et al. 2015) have been observed in familial AD in which mutations in PSEN1 lead to lysosomal deacidification (Lee et al. 2015) and in dementia caused by HIV in which TRPML1 activation is beneficial (Bae et al. 2014). Finally, mutations in FIG4 associated with FTD have been shown to disrupt lysosomal TRPML1 signaling, which can be corrected by activating Ca2+ release from TRPML1 with the synthetic agonist MLSA1 (Zou et al. 2015).
METHODS FOR ACCURATELY MEASURING LYSOSOMAL Ca2+
Accurate measurement of lysosomal Ca2+ content is complicated by the acidic pH and signaling cross talk between lysosomes and other intracellular Ca2+ stores, particularly the ER. The most accurate quantitation is undertaken by direct intravesicular measurements using probes endocytosed into lysosomes over a long timecourse of 24–48 h, confirmed to predominantly load lysosomes using antibody markers. These probes have a relatively low affinity for Ca2+, such as Rhod dextran, fura-dextran, or Oregon Green BAPTA 5N (Christensen et al. 2002; Lloyd-Evans et al. 2008; Lelouvier and Puertollano 2011). However, the Ca2+-binding affinity of all these probes are affected by pH. Therefore, to accurately estimate lysosomal Ca2+ content, it is also necessary to measure lysosomal pH, and to calibrate the Ca2+-binding probe appropriately. This should be performed with a pH-sensitive and -insensitive dextran combination, such as lysotracker yellow-blue dextran, and not a non-dextran pH-sensitive probe whose emissions do not always overlap with lysosomes. Using these methods, lysosomal free Ca2+ concentration has been measured as ∼500–600 µm in macrophages and human skin fibroblasts (Christensen et al. 2002; Lloyd-Evans et al. 2008).
It is also possible to undertake indirect measurements of lysosomal Ca2+. Perilysosomal Ca2+ can be measured using genetically encoded Ca2+ reporters such as cameleons or GCaMPs, conjugated to a lysosomal membrane protein. Cameleons and GCaMPs both use a conformational change brought about by Ca2+ binding to elicit changes in fluorescence (Kotlikoff 2007). When tethered to a lysosomal membrane protein, these probes can detect changes in cytosolic Ca2+ in the area immediately surrounding the lysosome, which should reduce interference by Ca2+ released from other intracellular stores. So far, the lysosomal membrane proteins LAMP-1 and TRPML1 have been used to create lysosome-specific Ca2+ reporters (Shen et al. 2012; McCue et al. 2013; Cheng et al. 2014). These can detect changes in perilysosomal Ca2+ concentration following addition of agents known to release lysosomal Ca2+, such as the cathepsin C substrate GPN, whose product causes osmotic lysis of lysosomes (Srinivas et al. 2002). Note that a recent study using cathepsin C inhibitors and null cells has raised questions about the ability of GPN to induce lysosomal Ca2+ release (Atakpa et al. 2019). However, other lysosomal proteases such as DPP7 can also hydrolyze Gly-Phe peptides (Rouf et al. 2013), suggesting that GPN is not a specific cathepsin C substrate and therefore remains a viable mechanism to induce lysosomal rupture. The most important consideration of the genetically encoded lysosomal near membrane Ca2+ sensors is that their use can potentially lead to overexpression of the protein. There is the potential that in expressing exaggerated levels of a protein, for example, TRPML1, above the normal range could in fact interfere with resting lysosomal Ca2+ levels. This could well be the case with the curious results obtained in NPC cells of no lysosomal Ca2+ defect when using the TRPML1-GCaMP, particularly when TRPML1 activation is supposed to improve NPC cellular function (Shen et al. 2012).
Finally, lysosomal Ca2+ can be measured indirectly using cytosolic Ca2+ probes such as Fura2-AM or Fluo dyes. Changes in cytosolic Ca2+ can be recorded following release of lysosomal Ca2+. This can also be achieved by osmotic lysis of lysosomes using the cathepsin C substrate GPN, by inducing lysosomal deacidification using bafilomycin A1, nigericin, or monensin, which exposes lysosomal Ca2+ leak, or by using agonists of lysosomal Ca2+ channels such as MLSA1 (TRPML1 agonist) or NAADP-AM (TPC2 agonist) (Shen et al. 2012; Morgan et al. 2015). However, as lysosomal Ca2+ release can trigger Ca2+ release from the ER, it is important to prevent ER Ca2+ release (Figs. 1 and 2). This can be achieved via pretreatment with ionomycin, a Ca2+ ionophore that releases Ca2+ from all intracellular stores apart from lysosomes, or inhibition of IP3 receptor-mediated Ca2+ release using xestospongin C (Davis et al. 2012; Kilpatrick et al. 2016b). It is easy to misinterpret lysosomal Ca2+ measurements if Ca2+ release from other intracellular stores is not clamped. This is best shown by measurements of lysosomal Ca2+ content in NPC disease cells, which was directly measured as 60% reduced compared with control cells (Lloyd-Evans et al. 2008). This reduced lysosomal Ca2+ content finding has been replicated by several other groups (Lee et al. 2010; Xu et al. 2012; Visentin et al. 2013; Speak et al. 2014; Höglinger et al. 2015), but in a few studies has not been replicated (Shen et al. 2012; Vienken et al. 2017). The two aspects that these studies have in common is (1) a lack of clamping of ER Ca2+ stores, which does not prevent a much larger ER Ca2+ release (Kilpatrick et al. 2013) that masks the NPC Ca2+ defect (Fig. 2), and (2) no direct intravesicular measurement, which should always be the gold standard when attempting to replicate others’ findings, as was the case with Coen et al. (2012). The cause of the NPC lysosomal Ca2+ defect has been attributed to the accumulation of sphingosine (Lloyd-Evans et al. 2008), a simple amino alcohol sphingolipid that is produced in the lysosome from the degradation of ceramide by the enzyme acid ceramidase. A recent study using sphingosine 1-phosphate lyase null cells to elevate intracellular sphingosine levels via S1P phosphatase has suggested that elevating intracellular/lysosomal sphingosine levels, as observed in NPC disease, does not cause defects in lysosomal Ca2+ (Vienken et al. 2017). However, it is important to note that in this study no direct measurements of lysosomal Ca2+ were performed, and with the indirect measurements no clamping of the ER Ca2+ store was performed; as such, the measurement of lysosomal Ca2+ content using GPN is most likely “drowned out” by a much greater associated ER Ca2+ release (Fig. 2). It is clear, therefore, that in a manner similar to the autophagy arena (Klionsky et al. 2016), the lysosomal Ca2+ field also requires a consensus on the best methodologies used to measure lysosomal Ca2+, otherwise this area of research will struggle to move forward.
Figure 2.
Accurate lysosomal Ca2+ measurements are impaired by failure to clamp intracellular stores and by whichever Ca2+-sensitive probe is used. (A–E) Comparison of quantification of release of lysosomal Ca2+ into the cytoplasm using 5 µm Fura-2, AM on a Zeiss Colibri LED widefield fluorescence system with AxioCam MRm charge-coupled device (CCD) camera (A–C) or 5 µm Calcium Green-1, AM in combination with Fura Red, AM on a Zeiss LSM510 confocal (D,E) all in Ca2+ free buffer. A comparison is made between the use of Gly-Phe-β-naphthylamide (GPN) to perforate lysosomes in the absence of intracellular store clamping (A,D) or following an initial addition of ionomycin (B,E) to clamp intracellular stores other than lysosomes (H Waller-Evans and AS Alshehri, unpubl.). As can be seen, in the absence of clamping (A,D) GPN releases considerably more Ca2+ than even the subsequent addition of ionomycin, which is not in keeping with the estimation of lysosomal volume (3%) in comparison with the endoplasmic reticulum (ER) (>10%). However, when the other intracellular stores are clamped (B,E), the amount of Ca2+ that is then released using GPN is considerably smaller. In the Fura 2 recordings (A–C), the Ca2+ released using GPN in clamped cells (B, inset) amounts to only ∼7% of the total Ca2+ released (C), which is in keeping with the estimation of the small cellular volume of lysosomes. In contrast, in the absence of clamping (A), GPN releases ∼82% of the total Ca2+ (C), which is clearly an overestimation and presumably reflects the induction of Ca2+ release from both lysosomes and other stores. Furthermore, a failure to clamp intracellular stores results in an underestimation of the lysosomal Ca2+ defect (D,E) reported in Niemann–Pick C (NPC) disease (red traces). In human fibroblast cells that are not clamped (D), the reduction in Ca2+ released from NPC cells on addition of GPN is only 21% compared with control. However, when intracellular stores are initially clamped with ionomycin the reduction in lysosomal Ca2+ content in the NPC human fibroblasts is now ∼58%, which is in keeping with the ∼55% reduction in intravesicular Ca2+ concentration in NPC cells measured directly with Ca2+-sensitive dextrans.
CONCLUDING REMARKS
The lysosomal Ca2+ field has expanded dramatically in the last decade, particularly in the area of human disease. However, some major questions remain, most importantly perhaps are issues concerning the best methods for measuring lysosomal Ca2+ content accurately, and what proteins mediate lysosomal Ca2+ uptake. Future directions in this field may focus on those areas, but also it is likely that advancement in our understanding of the proteins and mechanisms involved in maintaining ER–lysosome contact sites are going to have a big impact on our understanding of lysosomal Ca2+ homeostasis and how lysosomal Ca2+ regulates local and global Ca2+ signaling.
ACKNOWLEDGMENTS
Work in the Lloyd-Evans laboratory is generously supported by research grants from Action Medical Research (2337, 2069), Alzheimer Research UK (ARUK-IRG2015-7), European Research Council (ERC) Horizon 2020 Batcure, Sport Aiding Medical Research for Kids (SPARKS), and the UK Niemann–Pick Disease Group.
Footnotes
Editors: Geert Bultynck, Martin D. Bootman, Michael J. Berridge, and Grace E. Stutzmann
Additional Perspectives on Calcium Signaling available at www.cshperspectives.org
REFERENCES
- Adachi T, Arai K, Ohkuma S. 1996. A comparative study of (Ca2+–Mg2+)-ATPase on the lysosomal membrane and ecto-ATPase on the plasma membrane from rat liver. Biol Pharm Bull 19: 1291–1297. 10.1248/bpb.19.1291 [DOI] [PubMed] [Google Scholar]
- Alberts B. 2002. Molecular biology of the cell. Garland Science, New York. [Google Scholar]
- An Haack K, Narayan SB, Li H, Warnock A, Tan L, Bennett MJ. 2011. Screening for calcium channel modulators in CLN3 siRNA knock down SH-SY5Y neuroblastoma cells reveals a significant decrease of intracellular calcium levels by selected L-type calcium channel blockers. Biochim Biophys Acta 1810: 186–191. 10.1016/j.bbagen.2010.09.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Armstrong J. 2010. Yeast vacuoles: More than a model lysosome. Trends Cell Biol 20: 580–585. 10.1016/j.tcb.2010.06.010 [DOI] [PubMed] [Google Scholar]
- Atakpa P, Thillaiappan NB, Mataragka S, Prole DL, Taylor CW. 2018. IP3 receptors preferentially associate with ER–lysosome contact sites and selectively deliver Ca2+ to lysosomes. Cell Rep 25: 3180–3193.e7. 10.1016/j.celrep.2018.11.064 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Atakpa P, van Marrewijk LM, Apta-Smith M, Chakraborty S, Taylor CW. 2019. GPN does not release lysosomal Ca2+ but evokes Ca2+ release from the ER by increasing the cytosolic pH independently of cathepsin C. J Cell Sci 132: jcs223883 10.1242/jcs.223883 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bae M, Patel N, Xu H, Lee M, Tominaga-Yamanaka K, Nath A, Geiger J, Gorospe M, Mattson MP, Haughey NJ. 2014. Activation of TRPML1 clears intraneuronal Aα in preclinical models of HIV infection. J Neurosci 34: 11485–11503. 10.1523/jneurosci.0210-14.2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bagur R, Hajnóczky G. 2017. Intracellular Ca2+ sensing: Its role in calcium homeostasis and signaling. Mol Cell 66: 780–788. 10.1016/j.molcel.2017.05.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berridge MJ, Lipp P, Bootman MD. 2000. The versatility and universality of calcium signalling. Nat Rev Mol Cell Biol 1: 11–21. 10.1038/35036035 [DOI] [PubMed] [Google Scholar]
- Bertini I. 2007. Biological inorganic chemistry: Structure and reactivity. University Science Books, Sausalito, CA. [Google Scholar]
- Bultynck G, Kiviluoto S, Methner A. 2014. Bax inhibitor-1 is likely a pH-sensitive calcium leak channel, not a H+/Ca2+ exchanger. Sci Signal 7: pe22 10.1126/scisignal.2005764 [DOI] [PubMed] [Google Scholar]
- Burgoyne RD, Clague MJ. 2003. Calcium and calmodulin in membrane fusion. Biochim Biophys Acta 1641: 137–143. 10.1016/S0167-4889(03)00089-2 [DOI] [PubMed] [Google Scholar]
- Bygrave FL, Benedetti A. 1996. What is the concentration of calcium ions in the endoplasmic reticulum? Cell Calcium 19: 547–551. 10.1016/S0143-4160(96)90064-0 [DOI] [PubMed] [Google Scholar]
- Celsi F, Pizzo P, Brini M, Leo S, Fotino C, Pinton P, Rizzuto R. 2009. Mitochondria, calcium and cell death: A deadly triad in neurodegeneration. Biochim Biophys Acta 1787: 335–344. 10.1016/j.bbabio.2009.02.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chandra M, Zhou H, Li Q, Muallem S, Hofmann SL, Soyombo AA. 2011. A role for the Ca2+ channel TRPML1 in gastric acid secretion, based on analysis of knockout mice. Gastroenterology 140: 857–867. 10.1053/j.gastro.2010.11.040 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chandrachud U, Walker MW, Simas AM, Heetveld S, Petcherski A, Klein M, Oh H, Wolf P, Zhao WN, Norton S, et al. 2015. Unbiased cell-based screening in a neuronal cell model of batten disease highlights an interaction between Ca2+ homeostasis, autophagy, and CLN3 protein function. J Biol Chem 290: 14361–14380. 10.1074/jbc.M114.621706 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen CC, Keller M, Hess M, Schiffmann R, Urban N, Wolfgardt A, Schaefer M, Bracher F, Biel M, Wahl-Schott C, et al. 2014. A small molecule restores function to TRPML1 mutant isoforms responsible for mucolipidosis type IV. Nat Commun 5: 4681 10.1038/ncomms5681 [DOI] [PubMed] [Google Scholar]
- Chen Q, She J, Zeng W, Guo J, Xu H, Bai XC, Jiang Y. 2017. Structure of mammalian endolysosomal TRPML1 channel in nanodiscs. Nature 550: 415–418. 10.1038/nature24035 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cheng X, Zhang X, Gao Q, Ali Samie M, Azar M, Tsang WL, Dong L, Sahoo N, Li X, Zhuo Y, et al. 2014. The intracellular Ca2+ channel MCOLN1 is required for sarcolemma repair to prevent muscular dystrophy. Nat Med 20: 1187–1192. 10.1038/nm.3611 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Christensen KA, Myers JT, Swanson JA. 2002. pH-dependent regulation of lysosomal calcium in macrophages. J Cell Sci 115: 599–607. [DOI] [PubMed] [Google Scholar]
- Chu BB, Liao YC, Qi W, Xie C, Du X, Wang J, Yang H, Miao HH, Li BL, Song BL. 2015. Cholesterol transport through lysosome–peroxisome membrane contacts. Cell 161: 291–306. 10.1016/j.cell.2015.02.019 [DOI] [PubMed] [Google Scholar]
- Coen K, Flannagan RS, Baron S, Carraro-Lacroix LR, Wang D, Vermeire W, Michiels C, Munck S, Baert V, Sugita S, et al. 2012. Lysosomal calcium homeostasis defects, not proton pump defects, cause endo-lysosomal dysfunction in PSEN-deficient cells. J Cell Biol 198: 23–35. 10.1083/jcb.201201076 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colina C, Cervino V, Benaim G. 2002. Ceramide and sphingosine have an antagonistic effect on the plasma-membrane Ca2+-ATPase from human erythrocytes. Biochem J 362: 247–251. 10.1042/bj3620247 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Colombo MI, Beron W, Stahl PD. 1997. Calmodulin regulates endosome fusion. J Biol Chem 272: 7707–7712. 10.1074/jbc.272.12.7707 [DOI] [PubMed] [Google Scholar]
- Conn S, Gilliham M. 2010. Comparative physiology of elemental distributions in plants. Ann Bot 105: 1081–1102. 10.1093/aob/mcq027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cox TM, Cachón-González MB. 2012. The cellular pathology of lysosomal diseases. J Pathol 226: 241–254. 10.1002/path.3021 [DOI] [PubMed] [Google Scholar]
- Curcio-Morelli C, Charles FA, Micsenyi MC, Cao Y, Venugopal B, Browning MF, Dobrenis K, Cotman SL, Walkley SU, Slaugenhaupt SA. 2010. Macroautophagy is defective in mucolipin-1-deficient mouse neurons. Neurobiol Dis 40: 370–377. 10.1016/j.nbd.2010.06.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davis LC, Morgan AJ, Chen JL, Snead CM, Bloor-Young D, Shenderov E, Stanton-Humphreys MN, Conway SJ, Churchill GC, Parrington J, et al. 2012. NAADP activates two-pore channels on T cell cytolytic granules to stimulate exocytosis and killing. Curr Biol 22: 2331–2337. 10.1016/j.cub.2012.10.035 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Paola S, Scotto-Rosato A, Medina DL. 2018. TRPML1: The Ca2+ retaker of the lysosome. Cell Calcium 69: 112–121. 10.1016/j.ceca.2017.06.006 [DOI] [PubMed] [Google Scholar]
- Dong XP, Cheng X, Mills E, Delling M, Wang F, Kurz T, Xu H. 2008. The type IV mucolipidosis-associated protein TRPML1 is an endolysosomal iron release channel. Nature 455: 992–996. 10.1038/nature07311 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dong XP, Shen D, Wang X, Dawson T, Li X, Zhang Q, Cheng X, Zhang Y, Weisman LS, Delling M, et al. 2010. PI(3,5)P2 controls membrane trafficking by direct activation of mucolipin Ca2+ release channels in the endolysosome. Nat Commun 1: 38 10.1038/ncomms1037 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duchen MR. 2000. Mitochondria and calcium: From cell signalling to cell death. J Physiol 529 (Pt 1): 57–68. 10.1111/j.1469-7793.2000.00057.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eichelsdoerfer JL, Evans JA, Slaugenhaupt SA, Cuajungco MP. 2010. Zinc dyshomeostasis is linked with the loss of mucolipidosis IV-associated TRPML1 ion channel. J Biol Chem 285: 34304–34308. 10.1074/jbc.C110.165480 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elbaz Y, Schuldiner M. 2011. Staying in touch: The molecular era of organelle contact sites. Trends Biochem Sci 36: 616–623. 10.1016/j.tibs.2011.08.004 [DOI] [PubMed] [Google Scholar]
- Fairbairn IP, Stober CB, Kumararatne DS, Lammas DA. 2001. ATP-mediated killing of intracellular mycobacteria by macrophages is a P2X7-dependent process inducing bacterial death by phagosome–lysosome fusion. J Immunol 167: 3300–3307. 10.4049/jimmunol.167.6.3300 [DOI] [PubMed] [Google Scholar]
- Feng X, Huang Y, Lu Y, Xiong J, Wong CO, Yang P, Xia J, Chen D, Du G, Venkatachalam K, et al. 2014a. Drosophila TRPML forms PI(3,5)P2-activated cation channels in both endolysosomes and plasma membrane. J Biol Chem 289: 4262–4272. 10.1074/jbc.M113.506501 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Feng X, Xiong J, Lu Y, Xia X, Zhu MX. 2014b. Differential mechanisms of action of the mucolipin synthetic agonist, ML-SA1, on insect TRPML and mammalian TRPML1. Cell Calcium 56: 446–456. 10.1016/j.ceca.2014.09.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fine M, Schmiege P, Li X. 2018. Structural basis for PtdInsP2-mediated human TRPML1 regulation. Nat Commun 9: 4192 10.1038/s41467-018-06493-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galione A, Churchill GC. 2002. Interactions between calcium release pathways: Multiple messengers and multiple stores. Cell Calcium 32: 343–354. 10.1016/S0143416002001902 [DOI] [PubMed] [Google Scholar]
- Garrity AG, Wang W, Collier CM, Levey SA, Gao Q, Xu H. 2016. The endoplasmic reticulum, not the pH gradient, drives calcium refilling of lysosomes. eLife 5: e15887 10.7554/eLife.15887 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gerasimenko JV, Tepikin AV, Petersen OH, Gerasimenko OV. 1998. Calcium uptake via endocytosis with rapid release from acidifying endosomes. Curr Biol 8: 1335–1338. 10.1016/S0960-9822(07)00565-9 [DOI] [PubMed] [Google Scholar]
- Gordon PB, Holen I, Fosse M, Rotnes JS, Seglen PO. 1993. Dependence of hepatocytic autophagy on intracellularly sequestered calcium. J Biol Chem 268: 26107–26112. [PubMed] [Google Scholar]
- Grimm C, Cuajungco MP, van Aken AF, Schnee M, Jors S, Kros CJ, Ricci AJ, Heller S. 2007. A helix-breaking mutation in TRPML3 leads to constitutive activity underlying deafness in the varitint-waddler mouse. Proc Natl Acad Sci 104: 19583–19588. 10.1073/pnas.0709846104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grimm C, Jörs S, Saldanha SA, Obukhov AG, Pan B, Oshima K, Cuajungco MP, Chase P, Hodder P, Heller S. 2010. Small molecule activators of TRPML3. Chem Biol 17: 135–148. 10.1016/j.chembiol.2009.12.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Halachmi D, Eilam Y. 1989. Cytosolic and vacuolar Ca2+ concentrations in yeast cells measured with the Ca2+-sensitive fluorescence dye indo-1. FEBS Lett 256: 55–61. 10.1016/0014-5793(89)81717-X [DOI] [PubMed] [Google Scholar]
- Harris NS, Bazydlo LAL, Needham M. 2014. Calcium, magnesium, and phosphate. Lab Med 45: e44–e50. 10.1309/LMGLMZ8CIYMFNOGX [DOI] [Google Scholar]
- Hockey LN, Kilpatrick BS, Eden ER, Lin-Moshier Y, Brailoiu GC, Brailoiu E, Futter CE, Schapira AH, Marchant JS, Patel S. 2015. Dysregulation of lysosomal morphology by pathogenic LRRK2 is corrected by TPC2 inhibition. J Cell Sci 128: 232–238. 10.1242/jcs.164152 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Höglinger D, Haberkant P, Aguilera-Romero A, Riezman H, Porter FD, Platt FM, Galione A, Schultz C. 2015. Intracellular sphingosine releases calcium from lysosomes. eLife 4: e10616 10.7554/eLife.10616 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Huang P, Zou Y, Zhong XZ, Cao Q, Zhao K, Zhu MX, Murrell-Lagnado R, Dong XP. 2014. P2X4 forms functional ATP-activated cation channels on lysosomal membranes regulated by luminal pH. J Biol Chem 289: 17658–17667. 10.1074/jbc.M114.552158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jozef K, Stephan K, Barbara K. 2013. Calcium in the early evolution of living systems: A biohistorical approach. Curr Org Chem 17: 1738–1750. 10.2174/13852728113179990081 [DOI] [Google Scholar]
- Katz AK, Glusker JP, Beebe SA, Bock CW. 1996. Calcium ion coordination: A comparison with that of beryllium, magnesium, and zinc. J Am Chem Soc 118: 5752–5763. 10.1021/ja953943i [DOI] [Google Scholar]
- Kilpatrick BS, Eden ER, Schapira AH, Futter CE, Patel S. 2013. Direct mobilisation of lysosomal Ca2+ triggers complex Ca2+ signals. J Cell Sci 126: 60–66. 10.1242/jcs.118836 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kilpatrick BS, Magalhaes J, Beavan MS, McNeill A, Gegg ME, Cleeter MW, Bloor-Young D, Churchill GC, Duchen MR, Schapira AH, et al. 2016a. Endoplasmic reticulum and lysosomal Ca2+ stores are remodelled in GBA1-linked Parkinson disease patient fibroblasts. Cell Calcium 59: 12–20. 10.1016/j.ceca.2015.11.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kilpatrick BS, Yates E, Grimm C, Schapira AH, Patel S. 2016b. Endo-lysosomal TRP mucolipin-1 channels trigger global ER Ca2+ release and Ca2+ influx. J Cell Sci 129: 3859–3867. 10.1242/jcs.190322 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kiselyov K, Chen J, Rbaibi Y, Oberdick D, Tjon-Kon-Sang S, Shcheynikov N, Muallem S, Soyombo A. 2005. TRP-ML1 is a lysosomal monovalent cation channel that undergoes proteolytic cleavage. J Biol Chem 280: 43218–43223. 10.1074/jbc.M508210200 [DOI] [PubMed] [Google Scholar]
- Klionsky DJ, Abdelmohsen K, Abe A, Abedin MJ, Abeliovich H, Acevedo Arozena A, Adachi H, Adams CM, Adams PD, Adeli K, et al. 2016. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy 12: 1–222. 10.1080/15548627.2015.1100356 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kotlikoff MI. 2007. Genetically encoded Ca2+ indicators: Using genetics and molecular design to understand complex physiology. J Physiol 578: 55–67. 10.1113/jphysiol.2006.120212 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kukic I, Lee JK, Coblentz J, Kelleher SL, Kiselyov K. 2013. Zinc-dependent lysosomal enlargement in TRPML1-deficient cells involves MTF-1 transcription factor and ZnT4 (Slc30a4) transporter. Biochem J 451: 155–163. 10.1042/BJ20121506 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lamason RL, Mohideen MA, Mest JR, Wong AC, Norton HL, Aros MC, Jurynec MJ, Mao X, Humphreville VR, Humbert JE, et al. 2005. SLC24A5, a putative cation exchanger, affects pigmentation in zebrafish and humans. Science 310: 1782–1786. 10.1126/science.1116238 [DOI] [PubMed] [Google Scholar]
- Lange J, Haslett LJ, Lloyd-Evans E, Pocock JM, Sands MS, Williams BP, Cooper JD. 2018. Compromised astrocyte function and survival negatively impact neurons in infantile neuronal ceroid lipofuscinosis. Acta Neuropathol Commun 6: 74 10.1186/s40478-018-0575-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- LaPlante JM, Falardeau J, Sun M, Kanazirska M, Brown EM, Slaugenhaupt SA, Vassilev PM. 2002. Identification and characterization of the single channel function of human mucolipin-1 implicated in mucolipidosis type IV, a disorder affecting the lysosomal pathway. FEBS Lett 532: 183–187. 10.1016/S0014-5793(02)03670-0 [DOI] [PubMed] [Google Scholar]
- Lee H, Lee JK, Min WK, Bae JH, He X, Schuchman EH, Bae JS, Jin HK. 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. 10.1002/stem.401 [DOI] [PubMed] [Google Scholar]
- Lee JH, McBrayer MK, Wolfe DM, Haslett LJ, Kumar A, Sato Y, Lie PP, Mohan P, Coffey EE, Kompella U, et al. 2015. Presenilin 1 maintains lysosomal Ca2+ homeostasis via TRPML1 by regulating vATPase-mediated lysosome acidification. Cell Rep 12: 1430–1444. 10.1016/j.celrep.2015.07.050 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lelouvier B, Puertollano R. 2011. Mucolipin-3 regulates luminal calcium, acidification, and membrane fusion in the endosomal pathway. J Biol Chem 286: 9826–9832. 10.1074/jbc.M110.169185 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lemons RM, Thoene JG. 1991. Mediated calcium transport by isolated human fibroblast lysosomes. J Biol Chem 266: 14378–14382. [PubMed] [Google Scholar]
- Li M, Zhang WK, Benvin NM, Zhou X, Su D, Li H, Wang S, Michailidis IE, Tong L, Li X, et al. 2017. Structural basis of dual Ca2+/pH regulation of the endolysosomal TRPML1 channel. Nat Struct Mol Biol 24: 205–213. 10.1038/nsmb.3362 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Linebaugh BE, Sameni M, Day NA, Sloane BF, Keppler D. 1999. Exocytosis of active cathepsin B enzyme activity at pH 7.0, inhibition and molecular mass. Eur J Biochem 264: 100–109. 10.1046/j.1432-1327.1999.00582.x [DOI] [PubMed] [Google Scholar]
- Lloyd-Evans E. 2016. Acidic Ca2+ stores in neurodegeneration. Messenger (Los Angel) 5: 37–55. 10.1166/msr.2016.1054 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lloyd-Evans E, Haslett LJ. 2016. The lysosomal storage disease continuum with ageing-related neurodegenerative disease. Ageing Res Rev 32: 104–121. 10.1016/j.arr.2016.07.005 [DOI] [PubMed] [Google Scholar]
- Lloyd-Evans E, Platt FM. 2010. Lipids on trial: The search for the offending metabolite in Niemann–Pick type C disease. Traffic 11: 419–428. 10.1111/j.1600-0854.2010.01032.x [DOI] [PubMed] [Google Scholar]
- Lloyd-Evans E, Platt FM. 2011. Lysosomal Ca2+ homeostasis: Role in pathogenesis of lysosomal storage diseases. Cell Calcium 50: 200–205. 10.1016/j.ceca.2011.03.010 [DOI] [PubMed] [Google Scholar]
- Lloyd-Evans E, Morgan AJ, He X, Smith DA, Elliot-Smith E, Sillence DJ, Churchill GC, Schuchman EH, Galione A, Platt FM. 2008. Niemann–Pick disease type C1 is a sphingosine storage disease that causes deregulation of lysosomal calcium. Nat Med 14: 1247–1255. 10.1038/nm.1876 [DOI] [PubMed] [Google Scholar]
- Lloyd-Evans E, Waller-Evans H, Peterneva K, Platt FM. 2010. Endolysosomal calcium regulation and disease. Biochem Soc Trans 38: 1458–1464. 10.1042/BST0381458 [DOI] [PubMed] [Google Scholar]
- Lodish HF. 2000. Molecular cell biology. Freeman, New York. [Google Scholar]
- López JJ, Jardín I, Bobe R, Pariente JA, Enouf J, Salido GM, Rosado JA. 2008. STIM1 regulates acidic Ca2+ store refilling by interaction with SERCA3 in human platelets. Biochem Pharmacol 75: 2157–2164. 10.1016/j.bcp.2008.03.010 [DOI] [PubMed] [Google Scholar]
- Luzio JP, Pryor PR, Gray SR, Gratian MJ, Piper RC, Bright NA. 2005. Membrane traffic to and from lysosomes. Biochem Soc Symp 72: 77–86. 10.1042/bss0720077 [DOI] [PubMed] [Google Scholar]
- Masson M, Spezzatti B, Chapman J, Battisti C, Baumann N. 1992. Calmodulin antagonists chlorpromazine and W-7 inhibit exogenous cholesterol esterification and sphingomyelinase activity in human skin fibroblast cultures. Similarities between drug-induced and Niemann–Pick type C lipidoses. J Neurosci Res 31: 84–88. 10.1002/jnr.490310112 [DOI] [PubMed] [Google Scholar]
- McCue HV, Wardyn JD, Burgoyne RD, Haynes LP. 2013. Generation and characterization of a lysosomally targeted, genetically encoded Ca2+-sensor. Biochem J 449: 449–457. 10.1042/BJ20120898 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Medina DL, Ballabio A. 2015. Lysosomal calcium regulates autophagy. Autophagy 11: 970–971. 10.1080/15548627.2015.1047130 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Medina DL, Fraldi A, Bouche V, Annunziata F, Mansueto G, Spampanato C, Puri C, Pignata A, Martina JA, Sardiello M, et al. 2011. Transcriptional activation of lysosomal exocytosis promotes cellular clearance. Dev Cell 21: 421–430. 10.1016/j.devcel.2011.07.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Medina DL, Di Paola S, Peluso I, Armani A, De Stefani D, Venditti R, Montefusco S, Scotto-Rosato A, Prezioso C, Forrester A, et al. 2015. Lysosomal calcium signalling regulates autophagy through calcineurin and TFEB. Nat Cell Biol 17: 288–299. 10.1038/ncb3114 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Melchionda M, Pittman JK, Mayor R, Patel S. 2016. Ca2+/H+ exchange by acidic organelles regulates cell migration in vivo. J Cell Biol 212: 803–813. 10.1083/jcb.201510019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mills IG, Urbe S, Clague MJ. 2001. Relationships between EEA1 binding partners and their role in endosome fusion. J Cell Sci 114: 1959–1965. [DOI] [PubMed] [Google Scholar]
- Mole SE, Williams RE, Goebel HH. 2011. The neuronal ceroid lipofuscinoses (Batten disease). Oxford University Press, Oxford. [Google Scholar]
- Morgan AJ, Davis LC, Wagner SK, Lewis AM, Parrington J, Churchill GC, Galione A. 2013. Bidirectional Ca2+ signaling occurs between the endoplasmic reticulum and acidic organelles. J Cell Biol 200: 789–805. 10.1083/jcb.201204078 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morgan AJ, Davis LC, Galione A. 2015. Imaging approaches to measuring lysosomal calcium. Methods Cell Biol 126: 159–195. 10.1016/bs.mcb.2014.10.031 [DOI] [PubMed] [Google Scholar]
- Murrell-Lagnado RD. 2018. A role for P2X4 receptors in lysosome function. J Gen Physiol 150: 185–187. 10.1085/jgp.201711963 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Onyenwoke RU, Sexton JZ, Yan F, Diaz MC, Forsberg LJ, Major MB, Brenman JE. 2015. The mucolipidosis IV Ca2+ channel TRPML1 (MCOLN1) is regulated by the TOR kinase. Biochem J 470: 331–342. 10.1042/BJ20150219 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Palmieri M, Impey S, Kang H, di Ronza A, Pelz C, Sardiello M, Ballabio A. 2011. Characterization of the CLEAR network reveals an integrated control of cellular clearance pathways. Hum Mol Genet 20: 3852–3866. 10.1093/hmg/ddr306 [DOI] [PubMed] [Google Scholar]
- Parviainen L, Dihanich S, Anderson GW, Wong AM, Brooks HR, Abeti R, Rezaie P, Lalli G, Pope S, Heales SJ, et al. 2017. Glial cells are functionally impaired in juvenile neuronal ceroid lipofuscinosis and detrimental to neurons. Acta Neuropathol Commun 5: 74 10.1186/s40478-017-0476-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patel S, Cai X. 2015. Evolution of acidic Ca2+ stores and their resident Ca2+-permeable channels. Cell Calcium 57: 222–230. 10.1016/j.ceca.2014.12.005 [DOI] [PubMed] [Google Scholar]
- Patergnani S, Suski JM, Agnoletto C, Bononi A, Bonora M, De Marchi E, Giorgi C, Marchi S, Missiroli S, Poletti F, et al. 2011. Calcium signaling around mitochondria associated membranes (MAMs). Cell Commun Signal 9: 19 10.1186/1478-811X-9-19 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pelled D, Lloyd-Evans E, Riebeling C, Jeyakumar M, Platt FM, Futerman AH. 2003. Inhibition of calcium uptake via the sarco/endoplasmic reticulum Ca2+-ATPase in a mouse model of Sandhoff disease and prevention by treatment with N-butyldeoxynojirimycin. J Biol Chem 278: 29496–29501. 10.1074/jbc.M302964200 [DOI] [PubMed] [Google Scholar]
- Pelled D, Trajkovic-Bodennec S, Lloyd-Evans E, Sidransky E, Schiffmann R, Futerman AH. 2005. Enhanced calcium release in the acute neuronopathic form of Gaucher disease. Neurobiol Dis 18: 83–88. 10.1016/j.nbd.2004.09.004 [DOI] [PubMed] [Google Scholar]
- Penny CJ, Kilpatrick BS, Eden ER, Patel S. 2015. Coupling acidic organelles with the ER through Ca2+ microdomains at membrane contact sites. Cell Calcium 58: 387–396. 10.1016/j.ceca.2015.03.006 [DOI] [PubMed] [Google Scholar]
- Peters C, Mayer A. 1998. Ca2+/calmodulin signals the completion of docking and triggers a late step of vacuole fusion. Nature 396: 575–580. 10.1038/25133 [DOI] [PubMed] [Google Scholar]
- Peters C, Andrews PD, Stark MJ, Cesaro-Tadic S, Glatz A, Podtelejnikov A, Mann M, Mayer A. 1999. Control of the terminal step of intracellular membrane fusion by protein phosphatase 1. Science 285: 1084–1087. 10.1126/science.285.5430.1084 [DOI] [PubMed] [Google Scholar]
- Pittman JK. 2011. Vacuolar Ca2+ uptake. Cell Calcium 50: 139–146. 10.1016/j.ceca.2011.01.004 [DOI] [PubMed] [Google Scholar]
- Platt FM, Walkley SU. 2004. Lysosomal disorders of the brain: Recent advances in molecular and cellular pathogenesis and treatment. Oxford University Press, Oxford. [Google Scholar]
- Pryor PR, Mullock BM, Bright NA, Gray SR, Luzio JP. 2000. The role of intraorganellar Ca2+ in late endosome–lysosome heterotypic fusion and in the reformation of lysosomes from hybrid organelles. J Cell Biol 149: 1053–1062. 10.1083/jcb.149.5.1053 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Qureshi OS, Paramasivam A, Yu JC, Murrell-Lagnado RD. 2007. Regulation of P2X4 receptors by lysosomal targeting, glycan protection and exocytosis. J Cell Sci 120: 3838–3849. 10.1242/jcs.010348 [DOI] [PubMed] [Google Scholar]
- Raychowdhury MK, Gonzalez-Perrett S, Montalbetti N, Timpanaro GA, Chasan B, Goldmann WH, Stahl S, Cooney A, Goldin E, Cantiello HF. 2004. Molecular pathophysiology of mucolipidosis type IV: pH dysregulation of the mucolipin-1 cation channel. Hum Mol Genet 13: 617–627. 10.1093/hmg/ddh067 [DOI] [PubMed] [Google Scholar]
- Rouf SM, Ohara-Nemoto Y, Ono T, Shimoyama Y, Kimura S, Nemoto TK. 2013. Phenylalanine 664 of dipeptidyl peptidase (DPP) 7 and Phenylalanine 671 of DPP11 mediate preference for P2-position hydrophobic residues of a substrate. FEBS Open Bio 3: 177–183. 10.1016/j.fob.2013.03.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ruas M, Rietdorf K, Arredouani A, Davis LC, Lloyd-Evans E, Koegel H, Funnell TM, Morgan AJ, Ward JA, Watanabe K, et al. 2010. Purified TPC isoforms form NAADP receptors with distinct roles for Ca2+ signaling and endolysosomal trafficking. Curr Biol 20: 703–709. 10.1016/j.cub.2010.02.049 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ruas M, Davis LC, Chen CC, Morgan AJ, Chuang KT, Walseth TF, Grimm C, Garnham C, Powell T, Platt N, et al. 2015. Expression of Ca2+-permeable two-pore channels rescues NAADP signalling in TPC-deficient cells. EMBO J 34: 1743–1758. 10.15252/embj.201490009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Saftig P, Klumperman J. 2009. Lysosome biogenesis and lysosomal membrane proteins: Trafficking meets function. Nat Rev Mol Cell Biol 10: 623–635. 10.1038/nrm2745 [DOI] [PubMed] [Google Scholar]
- Sahoo N, Gu M, Zhang X, Raval N, Yang J, Bekier M, Calvo R, Patnaik S, Wang W, King G, et al. 2017. Gastric acid secretion from parietal cells is mediated by a Ca2+ efflux channel in the tubulovesicle. Dev Cell 41: 262–273.e6. 10.1016/j.devcel.2017.04.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samie MA, Xu H. 2014. Lysosomal exocytosis and lipid storage disorders. J Lipid Res 55: 995–1009. 10.1194/jlr.R046896 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samie M, Wang X, Zhang X, Goschka A, Li X, Cheng X, Gregg E, Azar M, Zhuo Y, Garrity AG, et al. 2013. A TRP channel in the lysosome regulates large particle phagocytosis via focal exocytosis. Dev Cell 26: 511–524. 10.1016/j.devcel.2013.08.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sano R, Annunziata I, Patterson A, Moshiach S, Gomero E, Opferman J, Forte M, d'Azzo A. 2009. GM1-ganglioside accumulation at the mitochondria-associated ER membranes links ER stress to Ca2+-dependent mitochondrial apoptosis. Mol Cell 36: 500–511. 10.1016/j.molcel.2009.10.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schiffmann R, Dwyer NK, Lubensky IA, Tsokos M, Sutliff VE, Latimer JS, Frei KP, Brady RO, Barton NW, Blanchette-Mackie EJ, et al. 1998. Constitutive achlorhydria in mucolipidosis type IV. Proc Natl Acad Sci 95: 1207–1212. 10.1073/pnas.95.3.1207 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmiege P, Fine M, Blobel G, Li X. 2017. Human TRPML1 channel structures in open and closed conformations. Nature 550: 366–370. 10.1038/nature24036 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schondorf DC, Aureli M, McAllister FE, Hindley CJ, Mayer F, Schmid B, Sardi SP, Valsecchi M, Hoffmann S, Schwarz LK, et al. 2014. iPSC-derived neurons from GBA1-associated Parkinson's disease patients show autophagic defects and impaired calcium homeostasis. Nat Commun 5: 4028 10.1038/ncomms5028 [DOI] [PubMed] [Google Scholar]
- Schönknecht G. 2013. Calcium signals from the vacuole. Plants (Basel) 2: 589–614. 10.3390/plants2040589 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Settembre C, Fraldi A, Medina DL, Ballabio A. 2013. Signals from the lysosome: A control centre for cellular clearance and energy metabolism. Nat Rev Mol Cell Biol 14: 283–296. 10.1038/nrm3565 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shen D, Wang X, Li X, Zhang X, Yao Z, Dibble S, Dong XP, Yu T, Lieberman AP, Showalter HD, et al. 2012. Lipid storage disorders block lysosomal trafficking by inhibiting a TRP channel and lysosomal calcium release. Nat Commun 3: 731 10.1038/ncomms1735 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sidransky E, Nalls MA, Aasly JO, Aharon-Peretz J, Annesi G, Barbosa ER, Bar-Shira A, Berg D, Bras J, Brice A, et al. 2009. Multicenter analysis of glucocerebrosidase mutations in Parkinson's disease. N Engl J Med 361: 1651–1661. 10.1056/NEJMoa0901281 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soyombo AA, Tjon-Kon-Sang S, Rbaibi Y, Bashllari E, Bisceglia J, Muallem S, Kiselyov K. 2006. TRP-ML1 regulates lysosomal pH and acidic lysosomal lipid hydrolytic activity. J Biol Chem 281: 7294–7301. 10.1074/jbc.M508211200 [DOI] [PubMed] [Google Scholar]
- Speak AO, Te Vruchte D, Davis LC, Morgan AJ, Smith DA, Yanjanin NM, Simmons L, Hartung R, Runz H, Mengel E, et al. 2014. Altered distribution and function of natural killer cells in murine and human Niemann-Pick disease type C1. Blood 123: 51–60. 10.1182/blood-2013-03-488692 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Srinivas SP, Ong A, Goon L, Goon L, Bonanno JA. 2002. Lysosomal Ca2+ stores in bovine corneal endothelium. Invest Ophthalmol Vis Sci 43: 2341–2350. [PubMed] [Google Scholar]
- Steinberg BE, Huynh KK, Brodovitch A, Jabs S, Stauber T, Jentsch TJ, Grinstein S. 2010. A cation counterflux supports lysosomal acidification. J Cell Biol 189: 1171–1186. 10.1083/jcb.200911083 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sun T, Wang X, Lu Q, Ren H, Zhang H. 2011. CUP-5, the C. elegans ortholog of the mammalian lysosomal channel protein MLN1/TRPML1, is required for proteolytic degradation in autolysosomes. Autophagy 7: 1308–1315. 10.4161/auto.7.11.17759 [DOI] [PubMed] [Google Scholar]
- Takeshima H, Venturi E, Sitsapesan R. 2015. New and notable ion-channels in the sarcoplasmic/endoplasmic reticulum: Do they support the process of intracellular Ca2+ release? J Physiol 593: 3241–3251. 10.1113/jphysiol.2014.281881 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tessitore A, del PMM, Sano R, Ma Y, Mann L, Ingrassia A, Laywell ED, Steindler DA, Hendershot LM, d'Azzo A. 2004. GM1-ganglioside-mediated activation of the unfolded protein response causes neuronal death in a neurodegenerative gangliosidosis. Mol Cell 15: 753–766. 10.1016/j.molcel.2004.08.029 [DOI] [PubMed] [Google Scholar]
- Tian X, Gala U, Zhang Y, Shang W, Nagarkar Jaiswal S, di Ronza A, Jaiswal M, Yamamoto S, Sandoval H, Duraine L, et al. 2015. A voltage-gated calcium channel regulates lysosomal fusion with endosomes and autophagosomes and is required for neuronal homeostasis. PLoS Biol 13: e1002103 10.1371/journal.pbio.1002103 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Treusch S, Knuth S, Slaugenhaupt SA, Goldin E, Grant BD, Fares H. 2004. Caenorhabditis elegans functional orthologue of human protein h-mucolipin-1 is required for lysosome biogenesis. Proc Natl Acad Sci 101: 4483–4488. 10.1073/pnas.0400709101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Venkatachalam K, Long AA, Elsaesser R, Nikolaeva D, Broadie K, Montell C. 2008. Motor deficit in a Drosophila model of mucolipidosis type IV due to defective clearance of apoptotic cells. Cell 135: 838–851. 10.1016/j.cell.2008.09.041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Venkatachalam K, Wong CO, Zhu MX. 2015. The role of TRPMLs in endolysosomal trafficking and function. Cell Calcium 58: 48–56. 10.1016/j.ceca.2014.10.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vergarajauregui S, Connelly PS, Daniels MP, Puertollano R. 2008. Autophagic dysfunction in mucolipidosis type IV patients. Hum Mol Genet 17: 2723–2737. 10.1093/hmg/ddn174 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vienken H, Mabrouki N, Grabau K, Claas RF, Rudowski A, Schömel N, Pfeilschifter J, Lütjohann D, van Echten-Deckert G, Meyer Zu Heringdorf D. 2017. Characterization of cholesterol homeostasis in sphingosine-1-phosphate lyase-deficient fibroblasts reveals a Niemann-Pick disease type C-like phenotype with enhanced lysosomal Ca2+ storage. Sci Rep 7: 43575 10.1038/srep43575 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Visentin S, De Nuccio C, Bernardo A, Pepponi R, Ferrante A, Minghetti L, Popoli P. 2013. The stimulation of adenosine A2A receptors ameliorates the pathological phenotype of fibroblasts from Niemann-Pick type C patients. J Neurosci 33: 15388–15393. 10.1523/jneurosci.0558-13.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vitner EB, Platt FM, Futerman AH. 2010. Common and uncommon pathogenic cascades in lysosomal storage diseases. J Biol Chem 285: 20423–20427. 10.1074/jbc.R110.134452 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Waller-Evans H, Lloyd-Evans E. 2015. Regulation of TRPML1 function. Biochem Soc Trans 43: 442–446. 10.1042/BST20140311 [DOI] [PubMed] [Google Scholar]
- Wang S, McDonnell EH, Sedor FA, Toffaletti JG. 2002. pH effects on measurements of ionized calcium and ionized magnesium in blood. Arch Pathol Lab Med 126: 947–950. [DOI] [PubMed] [Google Scholar]
- Wang W, Gao Q, Yang M, Zhang X, Yu L, Lawas M, Li X, Bryant-Genevier M, Southall NT, Marugan J, et al. 2015. Up-regulation of lysosomal TRPML1 channels is essential for lysosomal adaptation to nutrient starvation. Proc Natl Acad Sci 112: E1373–E1381. 10.1073/pnas.1419669112 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Warnock A, Tan L, Li C, An Haack K, Narayan SB, Bennett MJ. 2013. Amlodipine prevents apoptotic cell death by correction of elevated intracellular calcium in a primary neuronal model of Batten disease (CLN3 disease). Biochem Biophys Res Commun 436: 645–649. 10.1016/j.bbrc.2013.04.113 [DOI] [PubMed] [Google Scholar]
- Williams RJ. 2006. The evolution of calcium biochemistry. Biochim Biophys Acta 1763: 1139–1146. 10.1016/j.bbamcr.2006.08.042 [DOI] [PubMed] [Google Scholar]
- Wong CO, Li R, Montell C, Venkatachalam K. 2012. Drosophila TRPML is required for TORC1 activation. Curr Biol 22: 1616–1621. 10.1016/j.cub.2012.06.055 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu H, Delling M, Li L, Dong X, Clapham DE. 2007. Activating mutation in a mucolipin transient receptor potential channel leads to melanocyte loss in varitint-waddler mice. Proc Natl Acad Sci 104: 18321–18326. 10.1073/pnas.0709096104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Xu M, Liu K, Swaroop M, Porter FD, Sidhu R, Firnkes S, Ory DS, Marugan JJ, Xiao J, Southall N, et al. 2012. δ-Tocopherol reduces lipid accumulation in Niemann-Pick type C1 and Wolman cholesterol storage disorders. J Biol Chem 287: 39349–39360. 10.1074/jbc.M112.357707 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yamaguchi S, Jha A, Li Q, Soyombo AA, Dickinson GD, Churamani D, Brailoiu E, Patel S, Muallem S. 2011. Transient receptor potential mucolipin 1 (TRPML1) and two-pore channels are functionally independent organellar ion channels. J Biol Chem 286: 22934–22942. 10.1074/jbc.M110.210930 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang X, Li X, Xu H. 2012. Phosphoinositide isoforms determine compartment-specific ion channel activity. Proc Natl Acad Sci 109: 11384–11389. 10.1073/pnas.1202194109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhang X, Cheng X, Yu L, Yang J, Calvo R, Patnaik S, Hu X, Gao Q, Yang M, Lawas M, et al. 2016. MCOLN1 is a ROS sensor in lysosomes that regulates autophagy. Nat Commun 7: 12109 10.1038/ncomms12109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhong XZ, Zou Y, Sun X, Dong G, Cao Q, Pandey A, Rainey JK, Zhu X, Dong XP. 2017. Inhibition of transient receptor potential channel mucolipin-1 (TRPML1) by lysosomal adenosine involved in severe combined immunodeficiency diseases. J Biol Chem 292: 3445–3455. 10.1074/jbc.M116.743963 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zou J, Hu B, Arpag S, Yan Q, Hamilton A, Zeng YS, Vanoye CG, Li J. 2015. Reactivation of lysosomal Ca2+ efflux rescues abnormal lysosomal storage in FIG4-deficient cells. J Neurosci 35: 6801–6812. 10.1523/jneurosci.4442-14.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]