In recent years, our understanding of the structure, mechanisms and functions of the endo-lysosomal TPC (two-pore channel) family have grown apace. Gated by the second messengers, NAADP and PI(3,5)P2, TPCs are an integral part of fundamental signal-transduction pathways, but their array and plasticity of cation conductances (Na+, Ca2+, H+) allow them to variously signal electrically, osmotically or chemically. Their relative tissue- and organelle-selective distribution, together with agonist-selective ion permeabilities provides a rich palette from which extracellular stimuli can choose. TPCs are emerging as mediators of immunity, cancer, metabolism, viral infectivity and neurodegeneration as this short review attests.

These H+-rich (acidic) vesicles encompass a spectrum of organelles that include endo-lysosomes, lysosome-related organelles and secretory vesicles which are endowed with the common ability to store and release Ca2+. That is, in addition to their roles of trafficking cargo, repairing membranes, degrading macromolecules and nutrient sensing, acidic vesicles generate Ca2+ signals. The purpose of this article is to update our previous overview [1] with more recent developments pertaining to one particular family of Ca2+-permeable channels found on such acidic vesicles — the TPCs (two-pore channels) — and we confine our remarks to the mammalian channels.

The free [Ca2+] within endosomes is tens of micromolar, whereas in lysosomes it is ∼300–600 µM [2,3] and comparable to that of the other major Ca2+ store, the endoplasmic reticulum (ER). The route of lysosomal Ca2+-filling remains unclear with candidates being either Ca2+/H+ exchange [2,4] (Figure 1), Ca2+ transfer from the ER via IP3 receptors (IP3Rs) [5] or the ATP13A2 transporter [3]. Whilst cytosolic [Na+] is ∼12 mM, the lysosomal luminal [Na+] is reported from 21 mM [6] to 150 mM [7], though 21 mM may be more reliable since low temperatures and the lack of ATP (in [7]) disrupt normal monovalent cation gradients [8,9]. The resting membrane potential (ΔΨ) across lysosomes is luminally positive (19–100 mV) [2,10,11] (Figure 1).

Second-messenger synthesis couples plasmalemmal receptors to endo-lysosomal TPCs.

Figure 1.
Second-messenger synthesis couples plasmalemmal receptors to endo-lysosomal TPCs.

A model whereby cell-surface receptors can promote the synthesis of either second messenger, the dinucleotide, NAADP or lysosome-specific lipid, PI(3,5)P2. Cations (X+) exit the acidic vesicle when TPCs are gated by NAADP via small accessory proteins, LSm12 or JPT2, or by PI(3,5)P2 that binds directly to TPCs. The lysosomal lumen is acidic by virtue of the V-H+-ATPase, which is also the primary drive of the luminally positive membrane potential (ΔΨ). In one model, the H+ gradient drives Ca2+ uptake via an unknown exchanger.

Figure 1.
Second-messenger synthesis couples plasmalemmal receptors to endo-lysosomal TPCs.

A model whereby cell-surface receptors can promote the synthesis of either second messenger, the dinucleotide, NAADP or lysosome-specific lipid, PI(3,5)P2. Cations (X+) exit the acidic vesicle when TPCs are gated by NAADP via small accessory proteins, LSm12 or JPT2, or by PI(3,5)P2 that binds directly to TPCs. The lysosomal lumen is acidic by virtue of the V-H+-ATPase, which is also the primary drive of the luminally positive membrane potential (ΔΨ). In one model, the H+ gradient drives Ca2+ uptake via an unknown exchanger.

Close modal

Two features distinguish the acidic Ca2+ stores from the ER: first, they are a diminutive Ca2+ source compared with the ER (typically 10% of the ER volume [12]) and therefore, the total amount of Ca2+ that is released by acidic Ca2+ stores is small by comparison with the ER's; second, acidic vesicles are arguably more motile, traversing large distances relative to their size. Both these features afford acidic Ca2+ stores the capability of substantially impacting physiology and ‘punching above their weight’.

Analogous to IP3Rs evoking Ca2+ release from the ER, the gating of Ca2+-permeable channels on acidic vesicles increase cytosolic Ca2+. There are multiple families of channels found across the vesicular continuum and include the TPCs, mucolipins (TRPMLs) and P2X4 receptors. Their pattern of expression is not only cell-type dependent but also aligned with certain vesicle populations. Mouse and human each contain two isoforms, TPC1 and TPC2, that are ubiquitously expressed throughout the body and particularly high in kidney and immune cells [2]. Generally, TPC1 predominates in mildly acidic vesicles (recycling endosomes, early endosomes; pH 5.7–6.9) whilst TPC2 is mainly found in more acidic late-endosomes/lysosomes or secretory vesicles (pH 4.0–5.6) [2] (Figure 2).

Distribution of TPC channels throughout the endo-lysosomal system.

Figure 2.
Distribution of TPC channels throughout the endo-lysosomal system.

TPC1 channels are predominantly found in less acidic, earlier compartments, whereas TPC2 is found in later, more acidic vesicles. Trafficking of cargo through the endo-lysosomal system (e.g. endocytosis, viruses, bacterial toxins) as well as vesicle movement and fusion is also subject to TPC control.

Figure 2.
Distribution of TPC channels throughout the endo-lysosomal system.

TPC1 channels are predominantly found in less acidic, earlier compartments, whereas TPC2 is found in later, more acidic vesicles. Trafficking of cargo through the endo-lysosomal system (e.g. endocytosis, viruses, bacterial toxins) as well as vesicle movement and fusion is also subject to TPC control.

Close modal

Hot on the heels of the plant TPC atomic structure [13], mammalian TPC1 [14] and TPC2 [15] were resolved by cryogenic electron microscopy and complemented recently by a zebrafish TPC3 structure [16]. The structure of all three isoforms is a similar TPC dimer that strikingly resembles voltage-gated ion channels, to which TPCs are evolutionarily related [17] (Figure 3). However, mammalian TPCs are unusual in the cation-channel pantheon in having a selectivity filter without charged residues [18] so that, instead, the main barrier to permeation is a steric hydrophobic gate that is relieved upon ligand binding [19]. Expression of the pore-forming region alone results in a constitutively active cation channel [20]. Unlike TPC2, TPC1 is voltage-dependent by virtue of a unique voltage-sensing S4 domain [14] (Figure 3).

Structure of TPCs.

Figure 3.
Structure of TPCs.

(A) Topology cartoon of a TPC monomer with tandem repeats of two ‘Shaker’ domains (six transmembrane domains each). The positively charged amino acids (+++) in the S4 domains confer voltage sensitivity in TPC1. Magenta branches depict luminal glycosylation. (B) Cryo-EM structure of the human TPC2 dimer (PDB: 6NQ0) with the surface structure of A and B chains in blue and pink, respectively. The lipid, PI(3,5)P2, is shown as a space-filling model (yellow, red and orange) bound to a pocket in the A chain (a second lipid molecule, bound to the equivalent pocket on the B chain, cannot be seen behind). X+ represents the direction of cation flow. (C) human TPC2 as a ribbon diagram flipped 90° compared with (B) and viewed end-on from the cytosolic face. The central pore (green shading) is contributed to by both monomers. Both PI(3,5)P2 molecules bound are visible. Structures were generated using UCSF Chimera X [132].

Figure 3.
Structure of TPCs.

(A) Topology cartoon of a TPC monomer with tandem repeats of two ‘Shaker’ domains (six transmembrane domains each). The positively charged amino acids (+++) in the S4 domains confer voltage sensitivity in TPC1. Magenta branches depict luminal glycosylation. (B) Cryo-EM structure of the human TPC2 dimer (PDB: 6NQ0) with the surface structure of A and B chains in blue and pink, respectively. The lipid, PI(3,5)P2, is shown as a space-filling model (yellow, red and orange) bound to a pocket in the A chain (a second lipid molecule, bound to the equivalent pocket on the B chain, cannot be seen behind). X+ represents the direction of cation flow. (C) human TPC2 as a ribbon diagram flipped 90° compared with (B) and viewed end-on from the cytosolic face. The central pore (green shading) is contributed to by both monomers. Both PI(3,5)P2 molecules bound are visible. Structures were generated using UCSF Chimera X [132].

Close modal

As important signalling switches, Ca2+ channels exhibit sensitivity to multiple inputs, be they ions, ligands or proteins, and TPCs are modulated by all three classes.

Ca2+ feedback

Classically, IP3Rs are regulated strongly by both cytosolic and luminal Ca2+ which amplify or dampen signals [21]. The Ca2+ effects upon TPCs are, however, more variably reported. On the one hand, TPC1 was stimulated by cytosolic Ca2+ [22] via a shift in its voltage activation [23] which might reinforce endosomal Ca2+ release. On the other hand, others do not find any effect of cytosolic Ca2+ on TPC1 [24]. An effect of cytosolic Ca2+ on TPC2 is not reported, and its vestigial cytosolic EF-hands lack the residues for Ca2+-binding [15]. Regulation by luminal Ca2+ is equally inconsistent. Luminal Ca2+ has been shown to have no effect on TPC1 [22], an inhibitory one (by locking into a closed state) [23] or a stimulatory one [25]. For the other isoform, TPC2 is activated by luminal Ca2+ [26]. The reason for the discrepancies is unknown.

Second messengers

Unlike monogamous IP3Rs, TPCs are more promiscuous and respond to either of two second-messengers. One is the cytosolic soluble second messenger, NAADP (nicotinic acid adenine dinucleotide phosphate), the other is a lysosome-specific lipid, PI(3,5)P2 (phosphatidylinositol 3,5-bisphosphate) (Figure 1). Befitting a messenger role, their levels increase in response to different cell stimuli e.g. [27–29], and TPC-dependent Ca2+ signals (or currents) can be evoked by either ligand. Whilst the route of PI(3,5)P2 synthesis is clear (PIKfyve [30]), that for NAADP has, historically, been uncertain. Candidates include CD38 (or related ADP-ribosyl cyclases) [31–33], SARM1 [34] and, more recently, the NADPH oxidases, DUOX1/2 [35].

Thanks to the atomic structures, we know that PI(3,5)P2 binds directly to TPC1 and TPC2 (Figure 3) and how this gates the channel [14,15]; mutagenesis of complementary basic amino acids in the binding pocket abolishes activation by the phospholipid [14,15,36]. For NAADP, TPC stimulation is indirect, with NAADP binding to a smaller, accessory protein(s) (Figure 1). Recent screens have finally identified NAADP-binding proteins that mediate the gating of TPC, namely LSm12 [37] and JPT2 [38] (note: JPT2/HN1L was also reported to activate ryanodine receptors (RyRs) [39]). These proteins were unexpected candidates given that LSm12 is an RNA-binding protein and JPT2 is otherwise mechanistically orphaned (although linked to cancers) [40]. Attesting to its importance, LSm12 deletion is embryonic lethal [37]. Where and how these proteins bind to TPCs (and whether there is any isoform selectivity [40]) will prove a key future direction.

The potential for two molecular messengers to converge upon one channel is unusual, and the physiological consequence of this duality is ill-defined. Whether either (or both) messengers is required for TPC activation requires further work and may also be context-sensitive. For example, in the same macrophage, TPC2 responds to PI(3,5)P2 for macropinosome resolution (i.e. shrinkage and resorption) [41], but to NAADP for TPC-dependent phagocytosis [42]. Moreover, inhibition of PI(3,5)P2 synthesis with vacuolin-1 [43] did not alter NAADP-induced Ca2+ release [44] implying there is little interaction between the messengers, at least in fibroblasts. The messengers’ kinetics, uniqueness, redundancy or potential synergy may shape the signalling palette from which stimuli can choose.

Ion permeabilities

Ionic permeabilities inform us as to the possible (and multiple) roles of TPCs. For mammalian TPC1, its permeability sequence has been reported as Na+ > K+ > Ca2+ [24] or H+ > K+ > Na+ ≥ Ca2+ [22], whereas for TPC2 its rank order has been given as Na+ > Ca2+ > K+ > Cs+ [7,26,44–47]. In spite of common trends, the absolute permeability ratios recorded for a given TPC isoform perplexingly vary, e.g. the PCa/PNa for TPC1 is recorded as 0.98 [22], ∼0.05 [23] and 0.005 [24]. As with the Ca2+ feedback above, the different methodologies (e.g. lipid bilayers, whole-lysosome recording, ectopic expression in plant vacuoles) may be a contributing factor to some of the discrepancies.

Just as the plasmalemmal NMDA receptor is a transducer of both electrical (Na+) and chemical signals (Ca2+), so too may TPCs be multi-functional and alter endo-lysosomal ΔΨ and osmolarity (Na+), cytosolic Ca2+ signals or vesicular pH (pHL) (Figure 5). Note that egress of Na+ depolarises the endo-lysosomal membrane [48], which impacts both Ca2+-refilling and -release [2,49,50], and the ability of the electrogenic V-H+-ATPase to acidify the lumen [51]. Acidic vesicle ΔΨ is manifestly important physiologically e.g. for vesicular fusion [52], cholesterol storage [49] and phagocytosis [50].

When a Na+ conductor, TPC1 modulates vesicular ΔΨ and electrical excitability [24]. In an osmotic modality, TPC1/TPC2 co-ordinate macropinosome resolution when their Na+ fluxes drive Cl co-transport, water movement and pinosome shrinkage [41,53]. As Ca2+-permeable channels, TPCs have arguably garnered more attention physiologically (see below). Experimentally, it is currently not trivial to distinguish between the Na+ and Ca2+ modalities of TPC signalling in driving biological processes, in part due to our inability to monitor Na+ fluxes in situ.

However, the permeability sequence of TPCs is not immutable and depends on the stimulating messenger. Activation of TPC2 via the PI(3,5)P2-pathway promotes a predominantly Na+ current (PCa/PNa ∼ 0.08), whereas the NAADP pathway evokes an eight-fold larger Ca2+ conductance (PCa/PNa ∼ 0.65) [54] (Figure 4). TPC1 may also exhibit ligand-dependent permeability, albeit more modestly, with PI(3,5)P2 shifting the PCa/PNa from 0.98 to 0.42 [22]. Ligand-induced permeability changes are a unique feature of TPCs and thereby resolve early controversies as to the permeant ions. Thus, by the judicious selection of messenger, TPCs may be recruited to signal via Na+ (osmolarity, ΔΨ) or Ca2+ or pHL.

Ionic permeabilities are a function of the TPC isoform and stimulus.

Figure 4.
Ionic permeabilities are a function of the TPC isoform and stimulus.

Models depicting the relative permeabilities to Ca2+, Na+ and H+ are conveyed by the size of the coloured plumes. For TPC2, soluble NAADP evokes TPC2 currents with comparable Ca2+ and Na+ conductances, whereas the lipid PI(3,5)P2 stimulates Na+-selective currents.

Figure 4.
Ionic permeabilities are a function of the TPC isoform and stimulus.

Models depicting the relative permeabilities to Ca2+, Na+ and H+ are conveyed by the size of the coloured plumes. For TPC2, soluble NAADP evokes TPC2 currents with comparable Ca2+ and Na+ conductances, whereas the lipid PI(3,5)P2 stimulates Na+-selective currents.

Close modal

How NAADP elevated the pHL of acidic Ca2+ stores was unclear [29,55] until the demonstrations that both TPC1 and TPC2 conduct H+, i.e. efflux pathways from vesicles [22,54] (Figure 5). Therefore, NAADP may signal not just by an increase in cytosolic Ca2+, but by a coincident alkalinization of endo-lysosomes. Interestingly, pHL changes parallel the Ca2+ signals in that H+ fluxes are stimulated by the NAADP- but not the PI(3,5)P2-pathway with TPC2 [54]. In part via effects on vesicular pH, TPC2 influences melanosome pigmentation [47,56] and autophagy [57].

In summary, different messengers evoke different ionic signals. This may explain, for example, why PI(3,5)P2 is selected for macropinosome resolution: the lipid favourably stimulates fluxes of Na+ (but not H+) to drive Cl and water loss [41]; NAADP would have been unfavourable since it evokes smaller Na+ fluxes and an increase in pHL that could inhibit the essential Cl co-transport [53].

Models of different ionic signalling modalities for TPC2.

Figure 5.
Models of different ionic signalling modalities for TPC2.

With NAADP as the messenger, it binds to its accessory protein (LSm12 or JPT2, red hexagons) to evoke local Ca2+ nanodomains that are uniquely sensed by closely associated Ca2+-binding proteins (‘decoders’, brown hexagon)). When PI(3,5)P2 is the stimulus, Na+-selective currents are evoked which can depolarise the lysosome (ΔΨ) or promote osmotic changes and vesicle shrinkage (by Cl co-transport and concomitant water loss). NAADP can also promote H+ efflux through TPC2 and increase the lysosomal luminal pH (pHL).

Figure 5.
Models of different ionic signalling modalities for TPC2.

With NAADP as the messenger, it binds to its accessory protein (LSm12 or JPT2, red hexagons) to evoke local Ca2+ nanodomains that are uniquely sensed by closely associated Ca2+-binding proteins (‘decoders’, brown hexagon)). When PI(3,5)P2 is the stimulus, Na+-selective currents are evoked which can depolarise the lysosome (ΔΨ) or promote osmotic changes and vesicle shrinkage (by Cl co-transport and concomitant water loss). NAADP can also promote H+ efflux through TPC2 and increase the lysosomal luminal pH (pHL).

Close modal

Pharmacology

Compounds that modulate TPCs are a growing family, with currently more inhibitors to choose from than activators.

Inhibitors

Most of the inhibitors are pore-blockers. At high concentrations, traditional voltage-gated Ca2+-channel blockers interact with TPCs, e.g. verapamil inhibits TPC1 [54] and TPC2 [7] currents and NAADP-induced Ca2+ release [58,59]. Screens have re-purposed drugs as inhibitors of NAADP-induced Ca2+ release (by implication and by modelling, as TPC blockers) [60–62]. One broad-spectrum channel blocker, tetrandrine, is used to inhibit TPC2 [54,58,59] and TPC1 [63], and refinement of its structure has revealed more potent analogues towards TPC2, albeit with variable discrimination from TPC1 or TRPMLs [64]. The natural dietary flavonoid, naringenin, inhibits TPC1 and TPC2 with low affinity [65] and other Asian-plant flavonoids (pratensein and duartin) block TPC2 [66].

In terms of antagonising the messengers, the only cell-permeant NAADP antagonists that we have are BZ194 [67], the original Ned-19 [68], and its minimally modified analogue, Ned-K [69,70]. We do not yet have any specific inhibitors of the lipid-activation site, although high concentrations of Ned-19 unexpectedly block PI(3,5)P2-induced TPC2 currents [58].

Activators

Historically, activation of TPCs in intact-cell populations has been limited to NAADP delivery via liposomes [71,72] or cell-permeant NAADP (NAADP/AM) [73] which is notoriously labile. The recent discovery of stable, cell-permeant agonists that mimic these two TPC activators will open up the field, even if robust Ca2+ responses require the ectopic expression of TPC2 [54]. Each mimetic targets the TPC2 isoform (TPC2-A1-N [NAADP mimetic] and TPC2-A1-P [PI(3,5)P2 mimetic]), and are selective for TPC2 over TPC1 and TRPMLs [54]. TPC2-A1-P requires the lipid-binding site on TPC2 [54] (Figure 3) — likely a direct interaction with the channel — but the TPC2-A1-N activation mechanism is currently unclear. Does it bind to the NAADP accessory proteins LSm12/JPT2, or does it bind to TPC2 directly and mimic their interactions?

Interestingly, photo-release of another lipid, sphingosine, acutely evoked Ca2+ signals via TPC1 (but not TPC2) [74]; is this an underexplored new pathway? Surprisingly, tricyclic antidepressants (TCAs) and the motor-neuron-disease medication, riluzole, are TPC agonists [75], but their poly-pharmacology towards other transporters will probably limit their usefulness in intact cells.

Of broad interest, the mTOR inhibitor, rapamycin, evokes TPC2-dependent Ca2+ transients in myocytes [59] and promotes TPC2-mediated currents in synergy with PI(3,5)P2 [76]. Likewise, rapamycin activates TRPML1 by binding to the channel and synergises with the messenger, PI(3,5)P2 [77,78]. Whilst rapamycin activation of TRPML1 is direct, activation of TPC2 is suggested to be indirect via inhibition of mTOR [59,76,79].

Protein regulators

Other signalling inputs may interact with TPCs including protein kinases such as LRKK [80], JNK/p38 [81], mTOR [79] and the small GTPase, Rab7 [82]. Protein kinase A was proposed to modulate TPC2 currents via phosphorylation of Ser666 [46] although, curiously, this residue lies within the lysosomal lumen and not accessible to cytosolic cAMP signals. In some cases, the physiological context for these modifiers is poorly defined.

Polymorphisms

In the global population, TPC2 naturally occurs with a spectrum of different polymorphisms [83] and some impact TPC2 function. The degree of melanin pigmentation is inversely related to TPC2 activity [66] and two gain-of-function (GOF) polymorphisms (in different regions of the TPC2) each promoted blond-hair colour by independent mechanisms [76]. More recently, it was shown that the M484L mutation required an additional ‘permissive’ L564P polymorphism [83]. GOF mutants certainly produce larger currents in response to PI(3,5)P2 [54,76,83] or to the TPC2 agonists TPC2-A1-N and TPC2-A1-P [54]. However, it is less certain which signalling modality of TPC2 these polymorphisms produces the phenotype when roles for Ca2+ [56], Na+ (ΔΨ) [47,83] and pH [47,56,83] have all been posited for pigmentation.

How are TPC-dependent Ca2+ signals converted (decoded) into downstream responses? A common theme is that TPCs affect vesicle formation, trafficking, maturation and movement, many of which are sensitive to Ca2+ and lysosomal membrane potential. Being small Ca2+ stores, endo-lysosomes are designed to generate local rather than global Ca2+ signals, and TPCs couple to downstream physiology via local Ca2+ signals for which other Ca2+ sources cannot substitute, as exemplified by phagocytosis [41,42], exocytosis [84], membrane contact site (MCS) formation [85], receptor trafficking [86,87], development [31], vesicular fusion/motility [88,89]. Ca2+-decoders include Ca2+-dependent channels, protein kinases/phosphatases and membrane-fusion machinery.

Via Ca2+ release, TPCs trans-activate Ca2+-regulated ion channels on other membranes that are closely apposed, probably at MCSs. Via Ca2+-induced Ca2+ release (CICR), IP3Rs or RyRs on the ER can amplify the small Ca2+ release from endo-lysosomal TPCs to evoke global Ca2+ signals [90]. At the plasma membrane, Ca2+-sensitive channels (e.g. TRPM4/5 [91]) depolarise pancreatic β-cells following glucose-induced bursts of local NAADP/TPC Ca2+ signalling [91] under the plasma membrane [92]. Although there are also Ca2+-sensitive K+ channels on lysosomes that regulate vesicular ΔΨ, so far only Ca2+ released by TRPML1 has been linked to their activation [49].

How local TPC Ca2+ signals are otherwise decoded is underexplored. Privileged TPC-coupling to downstream processes implies that Ca2+-sensitive decoding proteins are intimately associated with TPCs and sense these high Ca2+ nanodomains. Several TPC interactomes have been published (reviewed in [93]), but surprisingly few Ca2+-binding proteins have been pulled out (e.g. annexins, although interactions have not always been validated). Phagocytosis is uniquely driven by local Ca2+ from TPCs (but not global Ca2+ signals) [42], where the Ca2+-dependent phosphatase, calcineurin, may be the Ca2+ decoder [42].

The molecular switches downstream of the immediate Ca2+-binding decoders are growing, and the GTPase, dynamin, has been linked to the NAADP/TPC axis during ‘inward’ trafficking at phagocytosis [42] and endocytosis of the glucose transporter, GLUT1 [94]. Regarding downstream phosphorylation, the MAP kinase, ERK1/2, mediates cell proliferation driven by TPC2 [95], although ERKs are not themselves Ca2+-binding proteins. In neuronal cells, the NAADP/TPC axis activates AMPK during autophagy [96]. In melanomas, TPC2 activates MITF (microphthalmia-associated transcription factor) via a GSK3β phosphorylation pathway [66]. Affirming a role in trafficking, fusion and vesicle motility, TPC1/2 interactomes are heavily biased towards SNARE complex proteins such as VAMPs and syntaxins [93].

Our appreciation of the importance of TPCs is expanding, but the following, recent examples incidentally reinforce that the precise molecular details of the circuitry are often lacking and we do not know the messenger, permeant ion or the decoders. Our understanding of the roles of TPCs is still in its infancy.

TPCs contribute to neuronal homeostasis [97]. The neurotransmitter, glutamate, uses an NAADP pathway to drive Ca2+ signals [98–100] which in turn can drive neuronal autophagy via TPC1/2 [96]. TPCs are important for memory and long-term potentiation [99,101], neuroprotection [98] and axonal/neurite extension [72,102]. Aberrant TPC signalling may contribute to neurodegeneration (see below).

In the vasculature, the role of TPCs is growing and, in particular, at vasculogenesis. The proliferation of endothelial precursors cells is dependent upon NAADP and TPC1 [72,103], and several studies implicate TPC2 in angiogenesis e.g. [65,104]. During embryogenesis, TPC1 and TPC2 promote different aspects of muscle development, namely myoseptal junction formation [105] and myogenesis, respectively [106]. Moreover, innervation of the muscle likewise relies on the NAADP/TPC2 axis [102].

Metabolically, the nutrient-sensing kinase complex of mTOR inhibits TPC2 which thereby responds to nutrient status [79]. Reciprocally, TPC2-KO enhances mTOR activity [107]. Manipulation of TPC1 expression reveals a potential link to glucose and fat metabolism [108], and the net surface expression of GLUT1 [94] and GLUT4 [108] glucose transporters are under the control of endosomal TPC1, probably by regulating endocytosis. Deletion of TPC2 in mice exacerbates the effects of a high-fat diet by reducing cholesterol/triglyceride clearance [87,109], although this does not translate into weight gain [109], partly due to enhanced insulin sensitivity in the absence of TPC2 [109].

TPCs are abundantly expressed in immune cells and are involved in often complex Ca2+ circuitry to regulate vesicular trafficking events in an immune context [110]. Extracellular particle clearance and fluid sampling during ‘inward’ trafficking events like phagocytosis [42] and macropinocytosis [41] in macrophages are mirrored by TPCs controlling ‘outward’ events like exocytotic secretion of histamine in mast cells [63], of cytolytic factors in cytotoxic T-cells [84] and the surface-presentation of chemokine signalling molecules [111]. Thus, TPCs may be invaluable during anaphylaxis, pathogen clearance by the innate immune system and T-cell clonal expansion.

Given their physiological roles — particularly of membrane and protein trafficking — TPCs are implicated in a wide range of diseases that are a significant health burden [112]. For neurodegenerative conditions like Alzheimer's (AD) and Parkinson's (PD) diseases, endo-lysosomes seem to play a critical role [113]. Accordingly, TPC2, in particular, has been linked to both AD [114] and PD [115,116], perhaps a result of aberrant trafficking, and LRKK2 mutation in the case of PD.

TPCs contribute to cardiovascular complications. In blood vessels, TPCs exacerbate hypoxia-induced hypertension [117,118] or macular degeneration [119]. In the heart, the NAADP/TPC axis mediates adrenaline-evoked ionotropy [33] via local Ca2+ signals at MCSs [120], and TPC2-KO mice manifest cardiac arrhythmias [121]. Similarly, NAADP/TPCs aggravate ischaemia-reperfusion injury: a regulatory subunit of protein kinase A senses the injury-induced redox changes to switch off the NAADP/TPC-dependent Ca2+ release [122] that might otherwise couple to lethal mitochondrial permeability transition [69]; therefore, pharmacological or genetic ablation of NAADP/TPCs protects against ischaemia-reperfusion injury [69,122].

Pathogens deliver toxins and/or enter the host cells to replicate, often gaining access via the endocytic pathway where they traffic through the endo-lysosomal system by co-opting host pathways. Since TPCs regulate vesicular uptake pathways (endocytosis, macropinocytosis, phagocytosis) [41,42,123] and are important for trafficking [86,87,124], their importance in contributing to pathogenicity was likely. Accordingly, inhibiting TPCs reduces infectivity of the Ebola virus [58], and of the Coronaviruses causing MERS [89] and Covid-19 [125,126]. Likewise, reducing the expression of the essential NAADP-binding protein, JPT2, also reduces viral uptake [38]. For HIV-1 replication, the virus subverts TPCs to allow essential Tat protein release [127]. Bacteria require toxins to traffic through the endo-lysosomal system and those for cholera, diphtheria and anthrax rely on TPCs [123,124,128].

An increasing field is that of TPCs in cancer [129] where, remarkably, TPCs impact different aspects. Feeding the tumour requires a blood supply and TPCs help drive angiogenesis [65,104]. Metastatic invasion and migration of the tumour cells themselves is another TPC-dependent process [66,104,130], and finally, tumour proliferation is under TPC2 control [64,66]. Consequently, TPC inhibition reduces tumour mass [64,104], and TPC1 and TPC2 may differentially contribute [131]. It is germane that the NAADP-binding protein, JPT2, is implicated in cancer progression [40].

In this brief overview, we have highlighted the diversity of both the ionic nature of the TPC signals and the breadth of the (patho)physiological processes in which TPCs play an important role, and this is only set to grow. A common theme is the involvement of TPCs in vesicular trafficking. With still so many unknowns, and the likely intersection with hitherto unsuspected pathways, the field of endo-lysosomal ionic signalling will continue to be a rich source to mine.

  • TPCs are endo-lysosomal Ca2+-permeable channels that are emerging as important signal transducers across biology and phyla.

  • Unusually, their ion conductances depend on the stimulus: they are plastic channels.

  • Different conductances confer the ability of TPCs to signal in different modalities (e.g. Ca2+, electrical, lysosomal pH).

  • New TPC protein regulators have recently emerged.

The authors declare that there are no competing interests associated with the manuscript.

AD

Alzheimer's diseases

ER

endoplasmic reticulum

GOF

gain-of-function

PD

Parkinson's diseases

TPC

two-pore channel

1
Morgan
,
A.J.
,
Davis
,
L.C.
,
Ruas
,
M.
and
Galione
,
A.
(
2015
)
TPC: the NAADP discovery channel?
Biochem. Soc. Trans.
43
,
384
389
2
Morgan
,
A.J.
,
Platt
,
F.M.
,
Lloyd-Evans
,
E.
and
Galione
,
A.
(
2011
)
Molecular mechanisms of endolysosomal Ca2+ signalling in health and disease
.
Biochem. J.
439
,
349
374
3
Narayanaswamy
,
N.
,
Chakraborty
,
K.
,
Saminathan
,
A.
,
Zeichner
,
E.
,
Leung
,
K.
,
Devany
,
J.
et al (
2019
)
A pH-correctable, DNA-based fluorescent reporter for organellar calcium
.
Nat. Methods
16
,
95
102
4
Melchionda
,
M.
,
Pittman
,
J.K.
,
Mayor
,
R.
and
Patel
,
S.
(
2016
)
Ca2+/H+ exchange by acidic organelles regulates cell migration in vivo
.
J. Cell Biol.
212
,
803
813
5
Garrity
,
A.G.
,
Wang
,
W.
,
Collier
,
C.M.
,
Levey
,
S.A.
,
Gao
,
Q.
and
Xu
,
H.
(
2016
)
The endoplasmic reticulum, not the pH gradient, drives calcium refilling of lysosomes
.
eLife
5
,
e15887
6
Steinberg
,
B.E.
,
Huynh
,
K.K.
,
Brodovitch
,
A.
,
Jabs
,
S.
,
Stauber
,
T.
,
Jentsch
,
T.J.
et al (
2010
)
A cation counterflux supports lysosomal acidification
.
J. Cell Biol.
189
,
1171
1186
7
Wang
,
X.
,
Zhang
,
X.
,
Dong
,
X.-P.
,
Samie
,
M.
,
Li
,
X.
,
Cheng
,
X.
et al (
2012
)
TPC proteins are phosphoinositide-activated sodium-selective ion channels in endosomes and lysosomes
.
Cell
151
,
372
383
8
Jentsch
,
T.J.
,
Hoegg-Beiler
,
M.B.
and
Vogt
,
J.
(
2015
)
Departure gate of acidic Ca2+ confirmed
.
EMBO J.
34
,
1737
1739
9
Reijngoud
,
D.
and
Tager
,
J.M.
(
1975
)
Effect of ionophores and temperature on intralysosomal pH
.
FEBS Lett.
54
,
76
79
10
Saminathan
,
A.
,
Devany
,
J.
,
Veetil
,
A.T.
,
Suresh
,
B.
,
Pillai
,
K.S.
,
Schwake
,
M.
et al (
2021
)
A DNA-based voltmeter for organelles
.
Nat. Nanotechnol.
16
,
96
103
11
Koivusalo
,
M.
,
Steinberg
,
B.E.
,
Mason
,
D.
and
Grinstein
,
S.
(
2011
)
In situ measurement of the electrical potential across the lysosomal membrane using FRET
.
Traffic (Copenhagen, Denmark)
12
,
972
982
12
Morgan
,
A.J.
(
2016
)
Ca2+ dialogue between acidic vesicles and ER
.
Biochem. Soc. Trans.
44
,
546
553
13
Kintzer
,
A.F.
and
Stroud
,
R.M.
(
2016
)
Structure, inhibition and regulation of two-pore channel TPC1 from Arabidopsis thaliana
.
Nature
531
,
258
264
14
She
,
J.
,
Guo
,
J.
,
Chen
,
Q.
,
Zeng
,
W.
,
Jiang
,
Y.
and
Bai
,
X.C.
(
2018
)
Structural insights into the voltage and phospholipid activation of the mammalian TPC1 channel
.
Nature
556
,
130
134
15
She
,
J.
,
Zeng
,
W.
,
Guo
,
J.
,
Chen
,
Q.
,
Bai
,
X.C.
and
Jiang
,
Y.
(
2019
)
Structural mechanisms of phospholipid activation of the human TPC2 channel
.
eLife
8
,
e45222
16
Dickinson
,
M.S.
,
Myasnikov
,
A.
,
Eriksen
,
J.
,
Poweleit
,
N.
and
Stroud
,
R.M.
(
2020
)
Resting state structure of the hyperdepolarization activated two-pore channel 3
.
Proc. Natl Acad. Sci. U.S.A.
117
,
1988
1993
17
Jaslan
,
D.
,
Bock
,
J.
,
Krogsaeter
,
E.
and
Grimm
,
C.
(
2020
)
Evolutionary aspects of TRPMLs and TPCs
.
Int. J. Mol. Sci.
21
,
4181
18
Milenkovic
,
S.
,
Bodrenko
,
I.V.
,
Carpaneto
,
A.
and
Ceccarelli
,
M.
(
2021
)
The key role of the central cavity in sodium transport through ligand-gated two-pore channels
.
Phys. Chem. Chem. Phys.
23
,
18461
18474
19
Milenkovic
,
S.
,
Bodrenko
,
I.V.
,
Lagostena
,
L.
,
Gradogna
,
A.
,
Serra
,
G.
,
Bosin
,
A.
et al (
2020
)
The mechanism and energetics of a ligand-controlled hydrophobic gate in a mammalian two pore channel
.
Phys. Chem. Chem. Phys.
22
,
15664
15674
20
Penny
,
C.J.
,
Rahman
,
T.
,
Sula
,
A.
,
Miles
,
A.J.
,
Wallace
,
B.A.
and
Patel
,
S.
(
2016
)
Isolated pores dissected from human two-pore channel 2 are functional
.
Sci. Rep.
6
,
38426
21
Prole
,
D.L.
and
Taylor
,
C.W.
(
2019
)
Structure and function of IP3 receptors
.
Cold Spring Harb. Perspect. Biol.
11
,
a035063
22
Pitt
,
S.J.
,
Lam
,
A.K.
,
Rietdorf
,
K.
,
Galione
,
A.
and
Sitsapesan
,
R.
(
2014
)
Reconstituted human TPC1 is a proton-permeable ion channel and is activated by NAADP or Ca2+
.
Sci. Signal.
7
,
ra46
23
Lagostena
,
L.
,
Festa
,
M.
,
Pusch
,
M.
and
Carpaneto
,
A.
(
2017
)
The human two-pore channel 1 is modulated by cytosolic and luminal calcium
.
Sci. Rep.
7
,
43900
24
Cang
,
C.
,
Bekele
,
B.
and
Ren
,
D.
(
2014
)
The voltage-gated sodium channel TPC1 confers endolysosomal excitability
.
Nat. Chem. Biol.
10
,
463
469
25
Rybalchenko
,
V.
,
Ahuja
,
M.
,
Coblentz
,
J.
,
Churamani
,
D.
,
Patel
,
S.
,
Kiselyov
,
K.
et al (
2012
)
Membrane potential regulates nicotinic acid adenine dinucleotide phosphate (NAADP) dependence of the pH- and Ca2+-sensitive organellar two-pore channel TPC1
.
J. Biol. Chem.
287
,
20407
20416
26
Pitt
,
S.J.
,
Funnell
,
T.
,
Sitsapesan
,
M.
,
Venturi
,
E.
,
Rietdorf
,
K.
,
Ruas
,
M.
et al (
2010
)
TPC2 is a novel NAADP-sensitive Ca2+-release channel, operating as a dual sensor of luminal pH and Ca2+
.
J. Biol. Chem.
285
,
35039
35046
27
Samie
,
M.
,
Wang
,
X.
,
Zhang
,
X.
,
Goschka
,
A.
,
Li
,
X.
,
Cheng
,
X.
et al (
2013
)
A TRP channel in the lysosome regulates large particle phagocytosis via focal exocytosis
.
Dev. Cell.
26
,
511
524
28
Gul
,
R.
,
Park
,
D.R.
,
Shawl
,
A.I.
,
Im
,
S.Y.
,
Nam
,
T.S.
,
Lee
,
S.H.
et al (
2016
)
Nicotinic acid adenine dinucleotide phosphate (NAADP) and cyclic ADP-ribose (cADPR) mediate Ca2+ signaling in cardiac hypertrophy induced by beta-adrenergic stimulation
.
PLoS One
11
,
e0149125
29
Morgan
,
A.J.
,
Davis
,
L.C.
,
Wagner
,
K.T.Y.
,
Lewis
,
A.M.
,
Parrington
,
J.
,
Churchill
,
G.C.
et al (
2013
)
Bidirectional Ca2+ signaling occurs between the endoplasmic reticulum and acidic organelles
.
J. Cell Biol.
200
,
789
805
30
Zolov
,
S.N.
,
Bridges
,
D.
,
Zhang
,
Y.
,
Lee
,
W.W.
,
Riehle
,
E.
,
Verma
,
R.
et al (
2012
)
In vivo, Pikfyve generates PI(3,5)P2, which serves as both a signaling lipid and the major precursor for PI5P
.
Proc. Natl Acad. Sci. U.S.A.
109
,
17472
17477
31
Kelu
,
J.J.
,
Webb
,
S.E.
,
Galione
,
A.
and
Miller
,
A.L.
(
2019
)
Characterization of ADP-ribosyl cyclase 1-like (ARC1-like) activity and NAADP signaling during slow muscle cell development in zebrafish embryos
.
Dev. Biol.
445
,
211
225
32
Lee
,
H.C.
and
Zhao
,
Y.J.
(
2019
)
Resolving the topological enigma in Ca2+ signaling by cyclic ADP-ribose and NAADP
.
J. Biol. Chem.
294
,
19831
19843
33
Lin
,
W.K.
,
Bolton
,
E.L.
,
Cortopassi
,
W.A.
,
Wang
,
Y.
,
O'Brien
,
F.
,
Maciejewska
,
M.
et al (
2017
)
Synthesis of the Ca2+-mobilizing messengers NAADP and cADPR by intracellular CD38 enzyme in the mouse heart: role in beta-adrenoceptor signaling
.
J. Biol. Chem.
292
,
13243
13257
34
Angeletti
,
C.
,
Amici
,
A.
,
Gilley
,
J.
,
Loreto
,
A.
,
Trapanotto
,
A.G.
,
Antoniou
,
C.
et al (
2022
)
SARM1 is a multi-functional NAD(P)ase with prominent base exchange activity, all regulated bymultiple physiologically relevant NAD metabolites
.
iScience
25
,
103812
35
Gu
,
F.
,
Kruger
,
A.
,
Roggenkamp
,
H.G.
,
Alpers
,
R.
,
Lodygin
,
D.
,
Jaquet
,
V.
et al (
2021
)
Dual NADPH oxidases DUOX1 and DUOX2 synthesize NAADP and are necessary for Ca2+ signaling during T cell activation
.
Sci. Signal.
14
,
eabe3800
36
Kirsch
,
S.A.
,
Kugemann
,
A.
,
Carpaneto
,
A.
,
Bockmann
,
R.A.
and
Dietrich
,
P.
(
2018
)
Phosphatidylinositol-3,5-bisphosphate lipid-binding-induced activation of the human two-pore channel 2
.
Cell. Mol. Life Sci.
75
,
3803
3815
37
Zhang
,
J.
,
Guan
,
X.
,
Shah
,
K.
and
Yan
,
J.
(
2021
)
Lsm12 is an NAADP receptor and a two-pore channel regulatory protein required for calcium mobilization from acidic organelles
.
Nat. Commun.
12
,
4739
38
Gunaratne
,
G.S.
,
Brailoiu
,
E.
,
He
,
S.
,
Unterwald
,
E.M.
,
Patel
,
S.
,
Slama
,
J.T.
et al (
2021
)
Essential requirement for JPT2 in NAADP-evoked Ca2+ signaling
.
Sci. Signal.
14
,
eabd5605
39
Roggenkamp
,
H.G.
,
Khansahib
,
I.
,
Hernandez
,
C.L.
,
Zhang
,
Y.
,
Lodygin
,
D.
,
Kruger
,
A.
et al (
2021
)
HN1L/JPT2: a signaling protein that connects NAADP generation to Ca2+ microdomain formation
.
Sci. Signal.
14
,
eabd5647
40
Marchant
,
J.S.
,
Gunaratne
,
G.S.
,
Cai
,
X.
,
Slama
,
J.T.
and
Patel
,
S.
(
2022
)
NAADP-binding proteins find their identity
.
Trends Biochem. Sci.
47
,
235
249
41
Freeman
,
S.A.
,
Uderhardt
,
S.
,
Saric
,
A.
,
Collins
,
R.F.
,
Buckley
,
C.M.
,
Mylvaganam
,
S.
et al (
2020
)
Lipid-gated monovalent ion fluxes regulate endocytic traffic and support immune surveillance
.
Science
367
,
301
305
42
Davis
,
L.C.
,
Morgan
,
A.J.
and
Galione
,
A.
(
2020
)
NAADP-regulated two-pore channels drive phagocytosis through endo-lysosomal Ca2+ nanodomains, calcineurin and dynamin
.
EMBO J.
39
,
e104058
43
Sano
,
O.
,
Kazetani
,
K.
,
Funata
,
M.
,
Fukuda
,
Y.
,
Matsui
,
J.
and
Iwata
,
H.
(
2016
)
Vacuolin-1 inhibits autophagy by impairing lysosomal maturation via PIKfyve inhibition
.
FEBS Lett.
590
,
1576
1585
44
Ruas
,
M.
,
Davis
,
L.C.
,
Chen
,
C.C.
,
Morgan
,
A.J.
,
Chuang
,
K.T.
,
Walseth
,
T.F.
et al (
2015
)
Expression of Ca2+-permeable two-pore channels rescues NAADP signalling in TPC-deficient cells
.
EMBO J.
34
,
1743
1758
45
Schieder
,
M.
,
Rotzer
,
K.
,
Bruggemann
,
A.
,
Biel
,
M.
and
Wahl-Schott
,
C.A.
(
2010
)
Characterization of two-pore channel 2 (TPCN2)-mediated Ca2+ currents in isolated lysosomes
.
J. Biol. Chem.
285
,
21219
21222
46
Lee
,
C.S.
,
Tong
,
B.C.
,
Cheng
,
C.W.
,
Hung
,
H.C.
and
Cheung
,
K.H.
(
2016
)
Characterization of two-pore channel 2 by nuclear membrane electrophysiology
.
Sci. Rep.
6
,
20282
47
Bellono
,
N.W.
,
Escobar
,
I.E.
and
Oancea
,
E.
(
2016
)
A melanosomal two-pore sodium channel regulates pigmentation
.
Sci. Rep.
6
,
26570
48
Morgan
,
A.J.
and
Galione
,
A.
(
2014
)
Two-pore channels (TPCs): current controversies
.
Bioessays
36
,
173
183
49
Wang
,
W.
,
Zhang
,
X.
,
Gao
,
Q.
,
Lawas
,
M.
,
Yu
,
L.
,
Cheng
,
X.
et al (
2017
)
A voltage-dependent K+ channel in the lysosome is required for refilling lysosomal Ca2+ stores
.
J. Cell Biol.
216
,
1715
1730
50
Sun
,
X.
,
Xu
,
M.
,
Cao
,
Q.
,
Huang
,
P.
,
Zhu
,
X.
and
Dong
,
X.P.
(
2020
)
A lysosomal K+ channel regulates large particle phagocytosis by facilitating lysosome Ca2+ release
.
Sci. Rep.
10
,
1038
51
Mindell
,
J.A.
(
2012
)
Lysosomal acidification mechanisms
.
Annu. Rev. Physiol.
74
,
69
86
52
Cang
,
C.
,
Aranda
,
K.
,
Seo
,
Y.J.
,
Gasnier
,
B.
and
Ren
,
D.
(
2015
)
TMEM175 is an organelle K+ channel regulating lysosomal function
.
Cell
162
,
1101
1112
53
Zeziulia
,
M.
,
Blin
,
S.
,
Schmitt
,
F.W.
,
Lehmann
,
M.
and
Jentsch
,
T.J.
(
2022
)
Proton-gated anion transport governs macropinosome shrinkage
.
Nat. Cell Biol.
24
,
885
895
54
Gerndt
,
S.
,
Chen
,
C.C.
,
Chao
,
Y.K.
,
Yuan
,
Y.
,
Burgstaller
,
S.
,
Scotto Rosato
,
A.
et al (
2020
)
Agonist-mediated switching of ion selectivity in TPC2 differentially promotes lysosomal function
.
eLife
9
,
e54712
55
Morgan
,
A.J.
and
Galione
,
A.
(
2007
)
NAADP induces pH changes in the lumen of acidic Ca2+ stores
.
Biochem. J.
402
,
301
310
56
Ambrosio
,
A.L.
,
Boyle
,
J.A.
,
Aradi
,
A.E.
,
Christian
,
K.A.
and
Di Pietro
,
S.M.
(
2016
)
TPC2 controls pigmentation by regulating melanosome pH and size
.
Proc. Natl Acad. Sci. U.S.A.
113
,
5622
5627
57
Lin
,
P.H.
,
Duann
,
P.
,
Komazaki
,
S.
,
Park
,
K.H.
,
Li
,
H.
,
Sun
,
M.
et al (
2015
)
Lysosomal two-pore channel subtype 2 (TPC2) regulates skeletal muscle autophagic signaling
.
J. Biol. Chem.
290
,
3377
3389
58
Sakurai
,
Y.
,
Kolokoltsov
,
A.A.
,
Chen
,
C.C.
,
Tidwell
,
M.W.
,
Bauta
,
W.E.
,
Klugbauer
,
N.
et al (
2015
)
Ebola virus. Two-pore channels control Ebola virus host cell entry and are drug targets for disease treatment
.
Science
347
,
995
998
59
Ogunbayo
,
O.A.
,
Duan
,
J.
,
Xiong
,
J.
,
Wang
,
Q.
,
Feng
,
X.
,
Ma
,
J.
et al (
2018
)
mTORC1 controls lysosomal Ca2+ release through the two-pore channel TPC2
.
Sci. Signal.
11
,
eeaao5775
60
Penny
,
C.J.
,
Vassileva
,
K.
,
Jha
,
A.
,
Yuan
,
Y.
,
Chee
,
X.
,
Yates
,
E.
et al (
2018
)
Mining of Ebola virus entry inhibitors identifies approved drugs as two-pore channel pore blockers
.
Biochim. Biophys. Acta Mol. Cell Res.
1866
,
1151
1161
61
Gunaratne
,
G.S.
,
Johns
,
M.E.
,
Hintz
,
H.M.
,
Walseth
,
T.F.
and
Marchant
,
J.S.
(
2018
)
A screening campaign in sea urchin egg homogenate as a platform for discovering modulators of NAADP-dependent Ca2+ signaling in human cells
.
Cell Calcium
75
,
42
52
62
Rahman
,
T.
,
Cai
,
X.
,
Brailoiu
,
G.C.
,
Abood
,
M.E.
,
Brailoiu
,
E.
and
Patel
,
S.
(
2014
)
Two-pore channels provide insight into the evolution of voltage-gated Ca2+ and Na+ channels
.
Sci. Signal.
7
,
ra109
63
Arlt
,
E.
,
Fraticelli
,
M.
,
Tsvilovskyy
,
V.
,
Nadolni
,
W.
,
Breit
,
A.
,
O'Neill
,
T.J.
et al (
2020
)
TPC1 deficiency or blockade augments systemic anaphylaxis and mast cell activity
.
Proc. Natl Acad. Sci. U.S.A.
117
,
18068
18078
64
Muller
,
M.
,
Gerndt
,
S.
,
Chao
,
Y.K.
,
Zisis
,
T.
,
Nguyen
,
O.N.P.
,
Gerwien
,
A.
et al (
2021
)
Gene editing and synthetically accessible inhibitors reveal role for TPC2 in HCC cell proliferation and tumor growth
.
Cell Chem. Biol.
28
,
1119
1131.e27
65
Pafumi
,
I.
,
Festa
,
M.
,
Papacci
,
F.
,
Lagostena
,
L.
,
Giunta
,
C.
,
Gutla
,
V.
et al (
2017
)
Naringenin impairs two-pore channel 2 activity and inhibits VEGF-induced angiogenesis
.
Sci. Rep.
7
,
5121
66
Netcharoensirisuk
,
P.
,
Abrahamian
,
C.
,
Tang
,
R.
,
Chen
,
C.C.
,
Rosato
,
A.S.
,
Beyers
,
W.
et al (
2021
)
Flavonoids increase melanin production and reduce proliferation, migration and invasion of melanoma cells by blocking endolysosomal/melanosomal TPC2
.
Sci. Rep.
11
,
8515
67
Dammermann
,
W.
,
Zhang
,
B.
,
Nebel
,
M.
,
Cordiglieri
,
C.
,
Odoardi
,
F.
,
Kirchberger
,
T.
et al (
2009
)
NAADP-mediated Ca2+ signaling via type 1 ryanodine receptor in T cells revealed by a synthetic NAADP antagonist
.
Proc. Natl Acad. Sci. U.S.A.
106
,
10678
10683
68
Naylor
,
E.
,
Arredouani
,
A.
,
Vasudevan
,
S.R.
,
Lewis
,
A.M.
,
Parkesh
,
R.
,
Mizote
,
A.
et al (
2009
)
Identification of a chemical probe for NAADP by virtual screening
.
Nat. Chem. Biol.
5
,
220
226
69
Davidson
,
S.M.
,
Foote
,
K.
,
Kunuthur
,
S.
,
Gosain
,
R.
,
Tan
,
N.
,
Tyser
,
R.
et al (
2015
)
Inhibition of NAADP signalling on reperfusion protects the heart by preventing lethal calcium oscillations via two-pore channel 1 and opening of the mitochondrial permeability transition pore
.
Cardiovasc. Res.
108
,
357
366
70
Pozo-Guisado
,
E.
,
Casas-Rua
,
V.
,
Tomas-Martin
,
P.
,
Lopez-Guerrero
,
A.M.
,
Alvarez-Barrientos
,
A.
and
Martin-Romero
,
F.J.
(
2013
)
Phosphorylation of STIM1 at ERK1/2 target sites regulates interaction with the microtubule plus-end binding protein EB1
.
J. Cell Sci.
126
,
3170
3180
71
Di Nezza
,
F.
,
Zuccolo
,
E.
,
Poletto
,
V.
,
Rosti
,
V.
,
De Luca
,
A.
,
Moccia
,
F.
et al (
2017
)
Liposomes as a putative tool to investigate NAADP signaling in vasculogenesis
.
J. Cell. Biochem.
118
,
3722
3729
72
Brailoiu
,
E.
,
Hoard
,
J.L.
,
Filipeanu
,
C.M.
,
Brailoiu
,
G.C.
,
Dun
,
S.L.
,
Patel
,
S.
et al (
2005
)
Nicotinic acid adenine dinucleotide phosphate potentiates neurite outgrowth
.
J. Biol. Chem.
280
,
5646
5650
73
Parkesh
,
R.
,
Lewis
,
A.
,
Aley
,
P.
,
Arredouani
,
A.
,
Rossi
,
S.
,
Tavares
,
R.
et al (
2008
)
Cell-permeant NAADP: a novel chemical tool enabling the study of Ca2+ signalling in intact cells
.
Cell Calcium
43
,
531
538
74
Hoglinger
,
D.
,
Haberkant
,
P.
,
Aguilera-Romero
,
A.
,
Riezman
,
H.
,
Porter
,
F.D.
,
Platt
,
F.M.
et al (
2015
)
Intracellular sphingosine releases calcium from lysosomes
.
eLife
4
,
e10616
75
Zhang
,
X.
,
Chen
,
W.
,
Li
,
P.
,
Calvo
,
R.
,
Southall
,
N.
,
Hu
,
X.
et al (
2019
)
Agonist-specific voltage-dependent gating of lysosomal two-pore Na+ channels
.
eLife
8
,
e51423
76
Chao
,
Y.K.
,
Schludi
,
V.
,
Chen
,
C.C.
,
Butz
,
E.
,
Nguyen
,
O.N.P.
,
Muller
,
M.
et al (
2017
)
TPC2 polymorphisms associated with a hair pigmentation phenotype in humans result in gain of channel function by independent mechanisms
.
Proc. Natl Acad. Sci. U.S.A.
114
,
E8595
E8602
77
Zhang
,
X.
,
Chen
,
W.
,
Gao
,
Q.
,
Yang
,
J.
,
Yan
,
X.
,
Zhao
,
H.
et al (
2019
)
Rapamycin directly activates lysosomal mucolipin TRP channels independent of mTOR
.
PLoS Biol.
17
,
e3000252
78
Gan
,
N.
,
Han
,
Y.
,
Zeng
,
W.
,
Wang
,
Y.
,
Xue
,
J.
and
Jiang
,
Y.
(
2022
)
Structural mechanism of allosteric activation of TRPML1 by PI(3,5)P2 and rapamycin
.
Proc. Natl Acad. Sci. U.S.A.
119
,
e2120404119
79
Cang
,
C.
,
Zhou
,
Y.
,
Navarro
,
B.
,
Seo
,
Y.-J.
,
Aranda
,
K.
,
Shi
,
L.
et al (
2013
)
mTOR regulates lysosomal ATP-sensitive two-pore Na+ channels to adapt to metabolic state
.
Cell
152
,
778
790
80
Gomez-Suaga
,
P.
,
Luzon-Toro
,
B.
,
Churamani
,
D.
,
Zhang
,
L.
,
Bloor-Young
,
D.
,
Patel
,
S.
et al (
2012
)
Leucine-rich repeat kinase 2 regulates autophagy through a calcium-dependent pathway involving NAADP
.
Hum. Mol. Genet.
21
,
511
525
81
Jha
,
A.
,
Ahuja
,
M.
,
Patel
,
S.
,
Brailoiu
,
E.
and
Muallem
,
S.
(
2014
)
Convergent regulation of the lysosomal two-pore channel-2 by Mg2+, NAADP, PI(3,5)P2 and multiple protein kinases
.
EMBO J.
33
,
501
511
82
Lin-Moshier
,
Y.
,
Keebler
,
M.V.
,
Hooper
,
R.
,
Boulware
,
M.J.
,
Liu
,
X.
,
Churamani
,
D.
et al (
2014
)
The Two-pore channel (TPC) interactome unmasks isoform-specific roles for TPCs in endolysosomal morphology and cell pigmentation
.
Proc. Natl Acad. Sci. U.S.A.
111
,
13087
13092
83
Bock
,
J.
,
Krogsaeter
,
E.
,
Passon
,
M.
,
Chao
,
Y.K.
,
Sharma
,
S.
,
Grallert
,
H.
et al (
2021
)
Human genome diversity data reveal that L564P is the predominant TPC2 variant and a prerequisite for the blond hair associated M484L gain-of-function effect
.
PLoS Genet.
17
,
e1009236
84
Davis
,
L.C.
,
Morgan
,
A.J.
,
Chen
,
J.L.
,
Snead
,
C.M.
,
Bloor-Young
,
D.
,
Shenderov
,
E.
et al (
2012
)
NAADP activates two-pore channels on T cell cytolytic granules to stimulate exocytosis and killing
.
Curr. Biol.
22
,
2331
2337
85
Kilpatrick
,
B.S.
,
Eden
,
E.R.
,
Hockey
,
L.N.
,
Yates
,
E.
,
Futter
,
C.E.
and
Patel
,
S.
(
2017
)
An endosomal NAADP-sensitive two-pore Ca2+ channel regulates ER-endosome membrane contact sites to control growth factor signaling
.
Cell Rep.
18
,
1
10
86
Muller
,
T.
,
Grossmann
,
S.
,
Mallmann
,
R.T.
,
Rommel
,
C.
,
Hein
,
L.
and
Klugbauer
,
N.
(
2021
)
Two-pore channels affect EGF receptor signaling by receptor trafficking and expression
.
iScience
24
,
102099
87
Grimm
,
C.
,
Holdt
,
L.M.
,
Chen
,
C.C.
,
Hassan
,
S.
,
Muller
,
C.
,
Jors
,
S.
et al (
2014
)
High susceptibility to fatty liver disease in two-pore channel 2-deficient mice
.
Nat. Commun.
5
,
4699
88
Ambrosio
,
A.L.
,
Boyle
,
J.A.
and
Di Pietro
,
S.M.
(
2015
)
TPC2 mediates new mechanisms of platelet dense granule membrane dynamics through regulation of Ca2+ release
.
Mol. Biol. Cell
26
,
3263
3274
89
Gunaratne
,
G.S.
,
Yang
,
Y.
,
Li
,
F.
,
Walseth
,
T.F.
and
Marchant
,
J.S.
(
2018
)
NAADP-dependent Ca2+ signaling regulates Middle East respiratory syndrome-coronavirus pseudovirus translocation through the endolysosomal system
.
Cell Calcium
75
,
30
41
90
Patel
,
S.
and
Brailoiu
,
E.
(
2012
)
Triggering of Ca2+ signals by NAADP-gated two-pore channels: a role for membrane contact sites?
Biochem. Soc. Trans.
40
,
153
157
91
Arredouani
,
A.
,
Ruas
,
M.
,
Collins
,
S.C.
,
Parkesh
,
R.
,
Clough
,
F.
,
Pillinger
,
T.
et al (
2015
)
Nicotinic acid adenine dinucleotide phosphate (NAADP) and endolysosomal two-pore channels modulate membrane excitability and stimulus-secretion coupling in mouse pancreatic beta cells
.
J. Biol. Chem.
290
,
21376
21392
92
Heister
,
P.M.
,
Powell
,
T.
and
Galione
,
A.
(
2021
)
Glucose and NAADP trigger elementary intracellular beta-cell Ca2+ signals
.
Sci. Rep.
11
,
10714
93
Krogsaeter
,
E.K.
,
Biel
,
M.
,
Wahl-Schott
,
C.
and
Grimm
,
C.
(
2019
)
The protein interaction networks of mucolipins and two-pore channels
.
Biochim. Biophys. Acta Mol. Cell Res.
1866
,
1111
1123
94
Fujii
,
T.
,
Katoh
,
M.
,
Ootsubo
,
M.
,
Nguyen
,
O.T.T.
,
Iguchi
,
M.
,
Shimizu
,
T.
et al (
2022
)
Cardiac glycosides stimulate endocytosis of GLUT1 via intracellular Na+, K+ -ATPase alpha3-isoform in human cancer cells
.
J. Cell Physiol.
237
,
2980
2991
95
Faris
,
P.
,
Casali
,
C.
,
Negri
,
S.
,
Iengo
,
L.
,
Biggiogera
,
M.
,
Maione
,
A.S.
et al (
2022
)
Nicotinic acid adenine dinucleotide phosphate induces intracellular Ca2+ signalling and stimulates proliferation in human cardiac mesenchymal stromal cells
.
Front. Cell Dev. Biol.
10
,
874043
96
Pereira
,
G.J.
,
Antonioli
,
M.
,
Hirata
,
H.
,
Ureshino
,
R.P.
,
Nascimento
,
A.R.
,
Bincoletto
,
C.
et al (
2017
)
Glutamate induces autophagy via the two-pore channels in neural cells
.
Oncotarget
8
,
12730
12740
97
Martucci
,
L.L.
and
Cancela
,
J.M.
(
2022
)
Neurophysiological functions and pharmacological tools of acidic and non-acidic Ca2+ stores
.
Cell Calcium
104
,
102582
98
Hermann
,
J.
,
Bender
,
M.
,
Schumacher
,
D.
,
Woo
,
M.S.
,
Shaposhnykov
,
A.
,
Rosenkranz
,
S.C.
et al (
2020
)
Contribution of NAADP to glutamate-evoked changes in Ca2+ homeostasis in mouse hippocampal neurons
.
Front. Cell Dev. Biol.
8
,
496
99
Foster
,
W.J.
,
Taylor
,
H.B.C.
,
Padamsey
,
Z.
,
Jeans
,
A.F.
,
Galione
,
A.
and
Emptage
,
N.J.
(
2018
)
Hippocampal mGluR1-dependent long-term potentiation requires NAADP-mediated acidic store Ca2+ signaling
.
Sci. Signal.
11
,
eaat9093
100
Pandey
,
V.
,
Chuang
,
C.C.
,
Lewis
,
A.M.
,
Aley
,
P.
,
Brailoiu
,
E.
,
Dun
,
N.
et al (
2009
)
Recruitment of NAADP-sensitive acidic Ca2+ stores by glutamate
.
Biochem. J.
422
,
503
512
101
Mallmann
,
R.T.
and
Klugbauer
,
N.
(
2020
)
Genetic inactivation of two-pore channel 1 impairs spatial learning and memory
.
Behav. Genet.
50
,
401
410
102
Guo
,
C.
,
Webb
,
S.E.
,
Chan
,
C.M.
and
Miller
,
A.L.
(
2020
)
TPC2-mediated ca2+ signaling is required for axon extension in caudal primary motor neurons in zebrafish embryos
.
J. Cell Sci.
133
,
jcs244780
103
Moccia
,
F.
,
Zuccolo
,
E.
,
Di Nezza
,
F.
,
Pellavio
,
G.
,
Faris
,
P.S.
,
Negri
,
S.
et al (
2020
)
Nicotinic acid adenine dinucleotide phosphate activates two-pore channel TPC1 to mediate lysosomal Ca2+ release in endothelial colony-forming cells
.
J. Cell Physiol.
236
,
688
705
104
Favia
,
A.
,
Pafumi
,
I.
,
Desideri
,
M.
,
Padula
,
F.
,
Montesano
,
C.
,
Passeri
,
D.
et al (
2016
)
NAADP-dependent Ca2+ signaling controls melanoma progression, metastatic dissemination and neoangiogenesis
.
Sci. Rep.
6
,
18925
105
Rice
,
K.L.
,
Webb
,
S.E.
and
Miller
,
A.L.
(
2022
)
Localized TPC1-mediated Ca2+ release from endolysosomes contributes to myoseptal junction development in zebrafish
.
J. Cell Sci.
135
,
jcs259564
106
Kelu
,
J.J.
,
Chan
,
H.L.
,
Webb
,
S.E.
,
Cheng
,
A.H.
,
Ruas
,
M.
,
Parrington
,
J.
et al (
2015
)
Two-pore channel 2 activity is required for slow muscle cell-generated Ca (2+)signaling during myogenesis in intact zebrafish
.
Int. J. Dev. Biol.
59
,
313
325
107
Chang
,
F.S.
,
Wang
,
Y.
,
Dmitriev
,
P.
,
Gross
,
J.
,
Galione
,
A.
and
Pears
,
C.
(
2020
)
A two-pore channel protein required for regulating mTORC1 activity on starvation
.
BMC Biol.
18
,
8
108
Garcia-Rua
,
V.
,
Feijoo-Bandin
,
S.
,
Garcia-Vence
,
M.
,
Aragon-Herrera
,
A.
,
Bravo
,
S.B.
,
Rodriguez-Penas
,
D.
et al (
2016
)
Metabolic alterations derived from absence of two-pore channel 1 at cardiac level
.
J. Biosci.
41
,
643
658
109
He
,
H.
,
Holl
,
K.
,
DeBehnke
,
S.
,
Yeo
,
C.T.
,
Hansen
,
P.
,
Gebre
,
A.K.
et al (
2018
)
Tpcn2 knockout mice have improved insulin sensitivity and are protected against high-fat diet-induced weight gain
.
Physiol. Genom.
50
,
605
614
110
Davis
,
L.C.
,
Morgan
,
A.J.
and
Galione
,
A.
(
2022
)
Acidic Ca2+ stores and immune-cell function
.
Cell Calcium
101
,
102516
111
He
,
T.
,
Yang
,
D.
,
Li
,
X.Q.
,
Jiang
,
M.
,
Islam
,
M.S.
,
Chen
,
S.
et al (
2020
)
Inhibition of two-pore channels in antigen-presenting cells promotes the expansion of TNFR2-expressing CD4+Foxp3+ regulatory T cells
.
Sci. Adv.
6
,
eaba6584
112
Patel
,
S.
and
Kilpatrick
,
B.S.
(
2018
)
Two-pore channels and disease
.
Biochim. Biophys. Acta Mol. Cell Res.
1865
,
1678
1686
113
Fraldi
,
A.
,
Klein
,
A.D.
,
Medina
,
D.L.
and
Settembre
,
C.
(
2016
)
Brain disorders due to lysosomal dysfunction
.
Annu. Rev. Neurosci.
39
,
277
295
114
Tong
,
B.C.
,
Wu
,
A.J.
,
Huang
,
A.S.
,
Dong
,
R.
,
Malampati
,
S.
,
Iyaswamy
,
A.
et al (
2022
)
Lysosomal TPCN (two pore segment channel) inhibition ameliorates beta-amyloid pathology and mitigates memory impairment in Alzheimer disease
.
Autophagy
18
,
624
642
115
Hockey
,
L.N.
,
Kilpatrick
,
B.S.
,
Eden
,
E.R.
,
Lin-Moshier
,
Y.
,
Brailoiu
,
G.C.
,
Brailoiu
,
E.
et al (
2015
)
Dysregulation of lysosomal morphology by pathogenic LRRK2 is corrected by TPC2 inhibition
.
J. Cell Sci.
128
,
232
238
116
Kilpatrick
,
B.S.
,
Magalhaes
,
J.
,
Beavan
,
M.S.
,
McNeill
,
A.
,
Gegg
,
M.E.
,
Cleeter
,
M.W.
et al (
2016
)
Endoplasmic reticulum and lysosomal Ca2+ stores are remodelled in GBA1-linked Parkinson disease patient fibroblasts
.
Cell Calcium
59
,
12
20
117
Hu
,
W.
,
Zhao
,
F.
,
Chen
,
L.
,
Ni
,
J.
and
Jiang
,
Y.
(
2021
)
NAADP-induced intracellular calcium ion is mediated by the TPCs (two-pore channels) in hypoxia-induced pulmonary arterial hypertension
.
J. Cell Mol. Med.
25
,
7485
7499
118
Jiang
,
Y.
,
Zhou
,
Y.
,
Peng
,
G.
,
Tian
,
H.
,
Pan
,
D.
,
Liu
,
L.
et al (
2018
)
Two-pore channels mediated receptor-operated Ca(2+) entry in pulmonary artery smooth muscle cells in response to hypoxia
.
Int. J. Biochem. Cell Biol.
97
,
28
35
119
Li
,
Y.
,
Schon
,
C.
,
Chen
,
C.C.
,
Yang
,
Z.
,
Liegl
,
R.
,
Murenu
,
E.
et al (
2021
)
TPC2 promotes choroidal angiogenesis and inflammation in a mouse model of neovascular age-related macular degeneration
.
Life Sci. Alliance
4
,
e202101047
120
Aston
,
D.
,
Capel
,
R.A.
,
Ford
,
K.L.
,
Christian
,
H.C.
,
Mirams
,
G.R.
,
Rog-Zielinska
,
E.A.
et al (
2017
)
High resolution structural evidence suggests the sarcoplasmic reticulum forms microdomains with acidic stores (lysosomes) in the heart
.
Sci. Rep.
7
,
40620
121
Capel
,
R.A.
,
Bolton
,
E.L.
,
Lin
,
W.K.
,
Aston
,
D.
,
Wang
,
Y.
,
Liu
,
W.
et al (
2015
)
Two pore channels (TPC2s) and nicotinic acid adenine dinucleotide phosphate (NAADP) at lysosomal-sarcoplasmic reticular junctions contribute to acute and chronic beta-adrenoceptor signaling in the heart
.
J. Biol. Chem.
290
,
30087
30098
122
Simon
,
J.N.
,
Vrellaku
,
B.
,
Monterisi
,
S.
,
Chu
,
S.M.
,
Rawlings
,
N.
,
Lomas
,
O.
et al (
2021
)
Oxidation of protein kinase A regulatory subunit PKARIalpha protects against myocardial ischemia-reperfusion injury by inhibiting lysosomal-triggered calcium release
.
Circulation
143
,
449
465
123
Castonguay
,
J.
,
Orth
,
J.H.C.
,
Müller
,
T.
,
Sleman
,
F.
,
Grimm
,
C.
,
Wahl-Schott
,
C.
et al (
2017
)
The two-pore channel TPC1 is required for efficient protein processing through early and recycling endosomes
.
Sci. Rep.
7
,
10038
124
Ruas
,
M.
,
Chuang
,
K.T.
,
Davis
,
L.C.
,
Al-Douri
,
A.
,
Tynan
,
P.W.
,
Tunn
,
R.
et al (
2014
)
TPC1 has two variant isoforms, and their removal has different effects on endo-lysosomal functions compared to loss of TPC2
.
Mol. Cell Biol.
34
,
3981
3992
125
Ou
,
X.
,
Liu
,
Y.
,
Lei
,
X.
,
Li
,
P.
,
Mi
,
D.
,
Ren
,
L.
et al (
2020
)
Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV
.
Nat. Commun.
11
,
1620
126
Clementi
,
N.
,
Scagnolari
,
C.
,
D'Amore
,
A.
,
Palombi
,
F.
,
Criscuolo
,
E.
,
Frasca
,
F.
et al (
2021
)
Naringenin is a powerful inhibitor of SARS-CoV-2 infection in vitro
.
Pharmacol. Res.
163
,
105255
127
Khan
,
N.
,
Halcrow
,
P.W.
,
Lakpa
,
K.L.
,
Afghah
,
Z.
,
Miller
,
N.M.
,
Dowdy
,
S.F.
et al (
2020
)
Two-pore channels regulate Tat endolysosome escape and Tat-mediated HIV-1 LTR transactivation
.
FASEB J.
34
,
4147
4162
128
Ruas
,
M.
,
Rietdorf
,
K.
,
Arredouani
,
A.
,
Davis
,
L.C.
,
Lloyd-Evans
,
E.
,
Koegel
,
H.
et al (
2010
)
Purified TPC isoforms form NAADP receptors with distinct roles for Ca2+ signaling and endolysosomal trafficking
.
Curr. Biol.
20
,
703
709
129
Chen
,
C.C.
,
Krogsaeter
,
E.
,
Kuo
,
C.Y.
,
Huang
,
M.C.
,
Chang
,
S.Y.
and
Biel
,
M.
(
2022
)
Endolysosomal cation channels point the way towards precision medicine of cancer and infectious diseases
.
Biomed. Pharmacother.
148
,
112751
130
Nguyen
,
O.N.
,
Grimm
,
C.
,
Schneider
,
L.S.
,
Chao
,
Y.K.
,
Atzberger
,
C.
,
Bartel
,
K.
et al (
2017
)
Two-pore channel function is crucial for the migration of invasive cancer cells
.
Cancer Res.
77
,
1427
1438
131
Jahidin
,
A.H.
,
Stewart
,
T.A.
,
Thompson
,
E.W.
,
Roberts-Thomson
,
S.J.
and
Monteith
,
G.R.
(
2016
)
Differential effects of two-pore channel protein 1 and 2 silencing in MDA-MB-468 breast cancer cells
.
Biochem. Biophys. Res. Commun.
477
,
731
736
132
Pettersen
,
E.F.
,
Goddard
,
T.D.
,
Huang
,
C.C.
,
Meng
,
E.C.
,
Couch
,
G.S.
,
Croll
,
T.I.
et al (
2021
)
UCSF chimerax: structure visualization for researchers, educators, and developers
.
Protein Sci.
30
,
70
82
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).