Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Feb 1.
Published in final edited form as: Cell Calcium. 2010 Jan 6;47(2):103–111. doi: 10.1016/j.ceca.2009.12.007

Aberrant Ca2+ handling in lysosomal storage disorders

Kirill Kiselyov 1, Soichiro Yamaguchi 2, Christopher W Lyons 1, Shmuel Muallem 2
PMCID: PMC2838446  NIHMSID: NIHMS168525  PMID: 20053447

Abstract

Lysosomal storage diseases (LSDs) are caused by inability of cells to process the material captured during endocytosis. While they are essentially diseases of cellular “indigestion”, LSDs affect large number of cellular activities and, as such, they teach us about the integrative function of the cell, as well as about the gaps in our knowledge of the endocytic pathway and membrane transport. The present review summarizes recent findings on Ca2+ handling in LSDs and attempts to identify the key questions on alterations inCa2+ signaling and membrane transport in this group of diseases, answers to which may lie in delineating the cellular pathogeneses of LSDs.

Lysosomal storage diseases as a model for the integrative function of the cell

Lysosomal storage diseases (LSDs) area diverse set of conditions that impair the uptake, sorting, or digestion of the material captured by cells during endocytosis or claimed by autophagy [1, 2]. Degradation of endocytosed extracellular matter and plasma membrane components is a complex process that involves selective membrane fusion, protein and lipid sorting and degradation, and absorption of the products of digestion [3, 4]. LSDs occur due to mutations in genes that code for components of the cellular endocytic machinery, or they can be caused by environmental influences such as toxic metals or drugs [57]. LSDs caused by gene mutations result in improperly delivered, structurally dysfunctional, or acutely inhibited lysosomal digestive enzymes; in some cases LSDs-causing mutations affect absorption of the products of digestion. Additionally, recently accumulated data suggests impaired membrane flow in the endocytic pathway, whether directly or indirectly induced by the genetic mutations, as a contributing factor in LSDs pathogenesis [8].

Although they are rarely discussed in the context of LSDs, chemically-induced dysregulated lysosomal function due to acute poisoning or chronic buildup of inhibitors (such as in iron-dependent lipofuscin buildup in aging cells [9, 10], and, perhaps, in Mucolipidosis type IV [11]), share elements of causality as well as cellular and clinical manifestations with the “genetically-induced” LSDs.

The inability of cells affected by LSDs to properly handle endocytosed material results in buildup of storage bodies, which are malformed endocytic organelles filled with undigested or unabsorbed material. Without exception, LSDs have detrimental effect on the cells and on the organism in general. Perhaps the best-known clinical manifestation of LSDs is neurodegeneration that is evident in most LSDs, with the severity varies between LSDs [12]. Although the LSDs-induced neurodegeneration has been convincingly linked to cell death, it is important to note that, at least in some cases, the ultrastructural readout of LSDs severity (number of storage bodies) do not seem to correlate with the extent of cell death or with the clinical manifestation of the disease. In addition, mutations in different components of the endocytic pathway may result in similar phenotype [13]. This suggests impairment in a crucial integrative aspect of cellular function, rather than a general toxic effect of storage bodies as the key cause of cell death in LSDs.

The latter notion points to an exciting recent novel paradigm development in the LSDs field that is spurred by the rapidly accumulating body of knowledge made possible by the use of LSDs as model systems to dissect the molecular mechanisms that translate lysosomal deficiency to cell death [1420]. These developments highlight the integrative function of the cells and interconnectedness of the mechanisms that make the cell work as a whole. Among the more interesting developments in this field is the understanding that lysosomal activity is directly responsible for maintaining higher household functions such as autophagy, organellar recycling, and cellular signaling [1, 9, 21, 22]. The analysis that led to identification of the molecular determinants of LSDs was and continues to be a major driving force in discovery of the critical components of the endocytic pathway. It will undoubtedly lead to better understanding of cell function.

Ca2+ handling by lysosomes – why and how

The role of Ca2+ in lysosomal function is supported by the well-established paradigm of its role in organellar fusion and by a series of reports, which suggested that Ca2+ release from the lysosomes is required for the content exchange between membranous vesicles derived from endocytic organelles [23]. Based on these two premises, the current model of lysosomal-endosomal fusion postulates that Ca2+ release from lysosomes through channels, whose possible molecular identity will be discussed below, drives the fusion of lysosomes with late endosomes and, therefore, the exposure of the endocytosis material to lysosomal digestive enzymes [24]. Below we will discuss implications, problems, and possible future developments of this model.

The postulate of Ca2+ involvement in lysosomal-endosomal fusion is similar to its role in membrane fusion in the presynaptic terminal. In synapses, vesicles containing neurotransmitter are docked at the plasma membrane by the SNARE complex [25]. Ca2+ influx resulting from the activation of voltage-dependent Ca2+ channels by the action potential causes conformational change in the SNARE proteins that prompts the fusion of vesicular membranes with the plasma membrane. As expected form the signal-reaction paradigm nature of neuronal transduction, synaptic transmission is “signal-centric”: it involves receiving the signal (action potential), signal interpretation (opening of the Ca2+ channels) and amplification (Ca2+ influx), which is followed by a reaction to the signal in the form of neurotransmitter release. When extended to lysosomal-endosomal fusion, it gives rise to a series of intriguing questions: if Ca2+ release from lysosomes drives lysosomal-endosomal fusion, then what is the “action potential” or signal that initiates such a release? If endosomal proximity to lysosomes is the signal for Ca2+ release from lysosomes, then what is the sensor that promotes such proximity? Is there a maturation signal indicating that a given endosome is ready to receive the digestive enzymes? Answering these question promises gaining new knowledge related to endocytic function in particular and to cellular material flow in general. Figure 1 summarizes the questions pertaining to Ca2+ regulation in LSDs that will be discussed in this review.

Figure 1.

Figure 1

The central questions in Ca2+ regulation in LSDs that are discussed in this review. Numbered circles correspond to: 1 – Regulation of Ca2+ entry, 2 – mitochondrial Ca2+ uptake, 3 – mitochondrial biogenesis and recycling, 4 – lysosomal Ca2+ uptake, 5 – lysosomal Ca2+ release, 6 – interaction of lysosomal Ca2+ release and Ca2+ release from endoplasmic reticulum.

Support for the Ca2+-dependent fusion model was developed using the in vitro mixing assays, in which the exchange of material between isolated lysosomal and endosomal fractions obtained from cells preloaded with specific markers was used as readout of the efficacy of organellar fusion [23, 26]. Using this system, it was established that in the absence of Ca2+in the reaction medium drastically reduced the rates of vesicle content mixing, indicating that Ca2+ is necessary for the fusion. A compound called U1866A, known to mimic the Niemann-Pick type C1 (NPC1) phenotype, induced alterations in Ca2+signaling that were similar to those observed in NPC1, and cytoplasmic Ca2+([Ca2+]i) elevation appears to correct the NPC1 phenotype [27]. Our data presented in Figure 2illustrate the possible role of Ca2+ in the lysosomal-endosomal fusion: loading cells with low µM concentration of the Ca2+ chelator BAPTA-AM to buffer cytoplasmic Ca2+ and prevent fusion of cellular organelles results in a buildup of storage bodies with a specific ultrastructural profile that is somewhat different from those induced by lysosomal inhibition by pharmacological means, or by acutely modeling the lysosomal storage disorder Mucolipidosis type IV (MLIV) [28]. Together, the available data suggest that a minimal level of Ca2+ in the lysosomal-endosomal proximity is necessary for the fusion between these organelles. However, these findings do not clarify whether elevated Ca2+is sufficient, whether it is specifically the lysosomal Ca2+ release that drives the fusion, or whether other sources of Ca2+, such as receptor-dependent Ca2+ release from the endoplasmic reticulum, can substitute for the lysosomal Ca2+release. In this respect, we have only rudimentary knowledge of lysosomal Ca2+ homeostasis, the molecular nature of the lysosomal Ca2+ loading mechanism(s) and the lysosomal Ca2+ release channel(s).

Figure 2.

Figure 2

Accumulation of storage bodies in cells treated with the intracellular Ca2+ chelator BAPTA-AM and with the H+ pump inhibitor Bafilomycin. HeLa cells were treated for 24 hours with 1 µM bafilomycin and 10 µM BAPTA. TRPML1 knockdown cells were analyzed shown 24 hours after transfection. Electron microscopic assays were performed as described before [19]. White arrows indicate dense storage bodies. Black arrows indicate large vacuolar structures that are present in BAPTA-treated but not in TRPML1 knockdown or Bafilomycin-treated cells.

Several candidates for the lysosomal Ca2+ release channels have recently emerged. Members of the mucolipin family of TRP channels have been implicated in the lysosomal-endosomal fusion shortly after their cloning and realization that they may function as Ca2+ permeable channels [29, 30]. The idea that the mucolipins, specifically TRPML1, are involved in Ca2+-dependent regulation of membrane flow in the endocytic pathway is based on two lines of evidence. The first is membrane traffic delays in cells lacking TRPML1 [29, 31] (critical analysis of these findings can be found in [28, 32]), and the second is the Ca2+ permeability of TRPML1 [33, 34]. Another member of the family, TRPML3, was shown to function as a Ca2+ channel [3336] that is expressed in multiple organelles to regulate membrane trafficking and autophagy [17, 18]. The findings with TRPML1 and TRPML3 suggest that these channels do participate in membrane trafficking and perhaps Ca2+-dependent fusion events (whether directly or indirectly). However, the role of the TRPMLs in membrane trafficking pertains to MLIV and not necessarily all LSDs.

Among other candidates for the lysosomal Ca2+ release channels are TRPM2, the NAADP sensitive two-pore channels (TPC1 and TPC2). At present, the strongest evidence support TPC2 as the lysosomal Ca2+ channel and TPC1 as the endosomal Ca2+ channel [37, 38]. TRPM2 appears to mediate Ca2+ release from an acidic compartment, but the identity of the compartment was not established with certainty and TRPM2 function is not ubiquitous but rather restricted to specific cells, like pancreatic β cells [37]. It was known for quite some time that the Ca2+ releasing second messenger NAADP releases Ca2+ from an acidic intracellular compartment that can be lysed using the lysosomal lytic agent Gly-Phy β-naphthylamide (GPN) [39]. Recently, two studies identified the NAADP receptors as the NAADP-activated Ca2+ channels TPC1 and TPC2 [37, 38]. The TPCs have been initially identified as the vacuolar Ca2+ release channels in plants [40], which is the equivalent of the animal cell lysosomes. The plant TPC1 appears to function as the main vacuolar Ca2+ release channels to mediate Ca2+ signaling in plants [41].In mammalian cells, TPC1 is localized mostly in endosomes, whereas TPC2 is found in lysosomes [37, 38]. TPC2 shows complete co-localization with TRPML1 (Yamaguchi et al., unpublished observation). Expression of TPC1 and TPC2 increased, while knockdown of the channels inhibited NAADP-mediated Ca2+ release from acidic compartments [37, 38].

Despite solid electrophysiological support for the role of the TPCs channels in the endosomal and lysosomal Ca2+ permeability, their role in endosomal-lysosomal function is not known yet. As of the time of writing this review, the effect of neither acute nor chronic down-regulation or knockdown of these channels on lysosomal function has been reported, making it difficult to discuss the significance of lysosomal Ca2+ permeability through these channels in the context of lysosomal deficiencies as a result of the loss of their permeability. However, establishing TPC2 as the lysosomal Ca2+ channel and determining its role in lysosomal function are likely to lead to dramatic enhancement of our knowledge of lysosomal function. These results will extend beyond degradation of endocytic matter, since the endocytic pathway is involved in many functions, such as antigen presentation and parasite defense. Therefore, identifying new ion channels that regulate lysosomal function will lead to novel pharmacological interventions in a variety of maladies.

Identification of the TRPMLs and the TPCs as lysosomal Ca2+ channels raise the questions of the source of the lysosomal Ca2+content and interaction of the lysosomal Ca2+ release channels with the activity of other transporters. Although it is well established that lysosomes contain high concentration of Ca2+, the mechanism and source of lysosomal Ca2+ accumulation are not known. The fact that the lysosomes can passively release Ca2+ to the cytosol indicates that their free Ca2+ concentration is higher than the cytoplasmic concentration of 0.1 µM and that the lysosomes may actively accumulate Ca2+. That the lysosomes can be depleted of Ca2+ by inhibition of their H+ pump suggests that they mediate H+-coupled Ca2+ uptake or that Ca2+ uptake is coupled to an uphill H+ transport mechanism that fuels Ca2+ uptake. It will be highly informative when such a mechanism is identified.

Functional interaction of the lysosomal Ca2+ release pathways with other electrogenic transporters deserves further investigation. Lysosomal H+ uptake and, therefore, acidification, which is perhaps the most important physiological ion transport function of the lysosome, is strictly modulated by the proton driving force. The proton driving force is a function of the steep proton gradient (pH 4.5–5 in the lysosome and 7.2–7.4 in the cytoplasm) that is established by the electrogenic V-type H+ pump [42], and of the lysosomal membrane potential that is set by the pump and by the ClC family of Cl channels [43], that balance the buildup of positive charge generated by the pump. ClC-7 has been established as the lysosomal Cl channel [44]. In fact, deletion of ClC-7 leads to LSD and neurodegeneration typical of LSD caused by mutation of lysosomal enzymes or of TRPML1 [45]. It has been convincingly established that modulation of the lysosomal membrane potential by the lysosomal Cl channel is the defining factor in lysosomal acidification [46]. In addition, the existence of a lysosomal cation channel whose function is to limit the lysosomal proton driving force was postulated [47]. It is clear that Ca2+ release through a lysosomal Ca2+ channel would strongly affect lysosomal membrane potential and, therefore, the proton driving force a well as the rates of lysosomal acidification and lysosomal pH. Exploring electrochemical interactions between the electrogenic lysosomal transporters is likely to yield new knowledge about the function of these key transporters in lysosomal function.

Anomalies in lysosomal Ca2+ regulation in lysosomal storage diseases

Identifying genetic determinants of lysosomal storage diseases has been an invaluable source of information on the components and the function of the endocytic pathway. It is, therefore, likely that exploring anomalies in lysosomal Ca2+ regulation in LSDs will facilitate our understanding of the endocytic machinery and may lead to novel pharmacological interventions into conditions caused by malfunction of the endocytic pathway. The most straightforward cause-and-effect relationship of Ca2+anomalies in LSDs has been suggested to explain the role of TRPML1 dysregulation in MLIV. The evidence that TRPML1 may function as a Ca2+channel [29, 30, 33] led to the suggestion that Ca2+ release through TRPML1 facilitates the fusion and/or fission in lysosomes to regulate the endocytic pathway [29, 31]. Although two independent reports did not confirm a direct role of TRPML1 in membrane traffic [28, 48], the role of TRPML1 in some Ca2+ dependent aspect of membrane flow in the endocytic pathway is supported by the recent observations of the Ca2+channel function of TRPML3 [3336], a relative of TRPML1, whose inactivation facilitates membrane trafficking, an opposite from what is predicted by the conventional model of Ca2+ release channels in membrane fusion.

The proposed role for TRPML1/TRPML3 in Ca2+ release from the endocytic compartments suggests that Ca2+ in endocytic organelles would be dysregulated as a result of altered function of these channels. At present, there is no evidence of such an effect: the only studies that assessed the total lysosomal Ca2+ as a function of TRPML1 status observed no difference in the total lysosomal Ca2+ in the absence or presence of TRPML1 or when TRPML1 is over-expressed [49]. Assaying lysosomal Ca2+ as a function of acute changes in the TRPML1/TRPML3 status probably holds one of the keys to identifying the role of these ion channels in lysosomal Ca2+ regulation.

The central problem in establishing a casual relationship between dysregulation of the endocytic pathway and lysosomal Ca2+homeostasis is the complex nature of lysosomal deficiencies caused by the lysosomal storage; hence, the buildup of undigested material in the lysosomes is likely to affect lysosomal Ca2+ content, either by changing the lysosomal Ca2+ buffering capacity or by affecting the activity and localization of the lysosomal ion transporters. Aberrant lysosomal Ca2+homeostasis in LSDs, whose causes are not directly related to Ca2+ handling, create an important precedent for such effects. Specifically, cells affected by NPC1, which is a defect in lysosomal cholesterol handling [50], show drastically suppressed lysosomal Ca2+ accumulation [27]. The cause of these diseases has not been directly linked to proteins responsible for lysosomal Ca2+ handling. Instead, facilitated Ca2+ release in the neuropathic Gaucher diseases has been attributed to potentiation of Ca2+ release through Ryanodine receptor by glucosylceramide (GlcCer) [51, 52]. This may imply that lysosomes express Ryanodine receptors. Although this has not been demonstrated directly, Ryanodine-sensitive Ca2+ release from an acidic pool has been postulated based on NAADP-mediated Ca2+ release and its inhibition by Ryanodine [53]. However, now we know that NAADP-evoked Ca2+ release is mediated by TPC1 and TPC2 [37, 38]. Thus, the GlcCer buildup in the Gaucher disease may affect aberrant activity of these channels. It is likely that lysosomal Ca2+ accumulation and release may be affected by the buildup of undigested endocytic products. A comprehensive account of proteins directly involved in the lysosomal Ca2+ regulation followed by analysis of their regulation by the products of endocytic digestion will help identify the effects of products of the lysosomal degradation on lysosomal Ca2+ homeostasis.

Anomalies of cellular Ca2+ regulation in lysosomal storage diseases

In addition to absorbing and degrading endocytosed material, lysosomes play a number of important housekeeping functions. They drive autophagy, a process of organelle renewal and regulation of the energy flow; they regulate surface expression of membrane proteins; and they are likely to play a role as a Ca2+ storage organelle. Therefore, similar to the role that LSDs played in the discovery of components of the endocytic pathway, documenting aberrations in Ca2+ signaling in LSDs will be an important tool in understanding the integrative function of cell, and more specifically, in better understanding how different organelles contribute to the response of the cell to a Ca2+ signal elicited by hormones and neurotransmitters.

Neurodegeneration is common in most forms of LSDs [12]. Classical forms of neurodegeneration, like Alzheimer's disease (AD), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), Huntington's disease (HD), have been firmly linked to anomalies in Ca2+ signaling [54], although the affected pathway remained somewhat controversial. In many cases, altered function of the IP3 receptors and of the endoplasmic reticulum (ER) Ca2+ ATPase pump (SERCA pump) have been implicated, although the most common finding is altered mitochondrial function (reviewed in [54, 55]). It is, therefore, not surprising that anomalous Ca2+ signaling has been also found in several forms of LSDs.

Several studies reported changes in several Ca2+ handling pathways that generate the receptor-evoked Ca2+ signal. Early studies examined the role of GlcCer, which accumulates in the lysosomes in Gaucher disease, on the function of the ER in cultured hippocampal neurons and reported increased tabulation of the ER, expansion of the ER Ca2+ pool, increased response to caffeine (indicative of increased Ca2+-induced Ca2+ release), an increased response to glutamate, and, thus, neurotoxicity [56]. Enhanced Ca2+ release was later reported in microsomes isolated from Gaucher type 1, type 2 and type 3 patient brains, and the enhanced Ca2+ release correlated with the increase in GlcCer in brain tissue [51]. The voltage-activated Ca2+ channels may also be affected in Gaucher disease. Treatment of patient fibroblasts with inhibitors of the L type Ca2+ channel partially restored glucocerebrosidase expression and activity [57]. This effect appeared to be related to the Ca2+ channel inhibitory function of the drugs that improved expression of cellular chaperones and folding of the mutated enzymes [57]. However, subsequent studies claimed that the effect of the blockers is not related to inhibition of Ca2+ channel activity but to their action as pharmacological chaperones [58]. Some support to the Ca2+ channel function effect of the L-type Ca2+ channel inhibitors appears to be provided by a global gene expression profiling of Niemann-Pick Type C disease, which found up-regulation of L-type Ca2+ channels in this diseases [59]. Nevertheless, the in vitro effect of the blockers in the patient fibroblasts appears to be reproducible; unfortunately, so far it failed to extend to the mouse model of the disease [60].

Studies with the mouse model of the lysosomal storage Sandhoff disease suggested reduced Ca2+ uptake into cortical microsomes that could be reversed by inhibition of glycolipid synthesis and GM2 storage [61]. Inhibition of Ca2+ uptake was attributed to reduction in SERCA pump activity by GM2 with no structural change in SERCA protein. However, only in the control microsomes about 50% of Ca2+ uptake activity was SERCA pump inhibitor thapsigargin-insensitive [61]. When this is corrected for, there is no apparent difference in Ca2+ uptake between the two groups. Hence, it remains to be determined whether Ca2+ homeostasis and signaling are altered in Sandhoff disease.

The lysosomal storage disease most characterized with respect to Ca2+ signaling is NPC1. It appears that generation and the properties of action potentials and depolarization mediated [Ca2+]i increase in neurons derived from the NPC1 mouse are completely normal [62], arguing that that Na+, K+ and voltage-gated Ca2+ channels are unaltered in these disease. Receptorstimulated Ca2+ signaling triggered by activation of G protein coupled receptors or tyrosine-kinase receptors in cells obtained from patients or the mouse model of NPC1 have not been reported. This deserves careful examination, especially in Purkinje neurons. Thus, early work examining expression of type 1 IP3 receptors (at the time known only as P400 protein) reported significant reduction in expression of the receptor and its elimination from NPC1 mice Purkinje neurons by 10–12 weeks of age [63]. A more recent and quite definitive study examined expression and IP3 receptors and SERCA pumps in cortical and cerebellar microsomes from Niemann-Pick type A mouse model and confirmed time-dependent reduction in Purkinje cells IP3 receptors and their elimination by 4 months and found a delayed reduction in SERCA pump expression and activity [64]. This is illustrated in Fig. 3 taken from [64]. Similar reduction in SERCA protein and activity was observed in brain microsomes of NPA patient [64].

Figure 3.

Figure 3

Changes in the expression levels of key Ca2+signaling proteins in the cerebellum of Niemann-Pick type A mice (modified from [64]). Immunostaining of cerebellar sliced obtained from control (ASM+/+) and Niemann-Pick mice (ASM−/−) mice. Note the severe loss of IP3 receptors, SERCA Ca2+ pumps and calbindin in Purkinje cells, all critical proteins in Ca2+signaling. The Ca2+ signaling proteins are lost before the structural protein Zebrin II, suggesting early deterioration of the Ca2+ signaling machinery and early onset of Ca2+ toxicity. Details can be found in [64].

At present, it is not clear whether alteration in SERCA pump and IP3 receptors is typical of all Niemann-Pick disease cells. A recent study analyzed Ca2+ signaling, storage, and release in the ER, mitochondria and lysosomes in human NPC1 B lymphocytes and fibroblasts [27]. All parameters examined: ryanodine receptor-mediated Ca2+ release, ER Ca2+ content and its release by thapsigargin or ionomycin, and mitochondrial Ca2+ release by the uncoupler FCCP, appear normal, except for Ca2+ content in acidic compartments that was found to be markedly reduced in NPC1 cells. The lysosomes express the NAADP-activated TPC2 [37, 38], and the response to NAADP is abolished in NPC1 cells, suggesting that the reduced Ca2+ storage in the acidic compartment reflects reduced lysosomal Ca2+ content [27]. The same Ca2+ phenotype could be reproduced by pharmacological induction of NPC1 in RAW microphages. Reduced lysosomal Ca2+ storage in NPC1 was attributed to accumulation of sphingosine in the lysosomes [27].

Lloyd-Evans E et al. [27] hypothesized that reduced lysosomal Ca2+ storage in NPC1 leads to impaired endocytosis and membrane trafficking and this is responsible for the disease phenotype, since [Ca2+]i elevation by activation of the SOC channels with thapsigargin partially corrected the phenotype. Based on this observation, they showed that treating NPC1 mice with the mild SERCA pump inhibitor curcumin [65] reduced the cellular NPC1 phenotype and significantly improved the weight and survival of the mice [27]. At present, it is not certain that the effects of treatment with thapsigargin and curcumin are due primarily to [Ca2+]i elevation and improved endocytosis and membrane trafficking. These drugs release Ca2+ from the ER to improve protein folding and have been used before to improve folding of misfolded mutant proteins, such as ΔF508-CFTR [66]. It is possible that this is also the case with the NPC1 mice, as was found with the L-type Ca2+ channel inhibitors in Gaucher [57] and NPC1 [60] diseases.

The combined findings in Niemann-Pick disease and models, of reduced IP3 receptors and SERCA pumps and reduced ER Ca2+ storage discussed above, predict that receptor-evoked Ca2+ signaling should be severely impaired in cells affected in NPC1 to alter all cellular functions mediated by Ca2+. The virtual lack of ER Ca2+ stores in NPC1 disease raises the question of the activity of the SOCs in these cells. The Orai and TRPC SOC channels are regulated by the Ca2+ binding protein STIM1 [67], which senses the Ca2+ stored in the ER and transmits it to the SOCs [68, 69]. Reduced or a lack of ER Ca2+ in NPC1 would predict that SOCs should be constitutively active in NPC1 cells. Aberrant SOC activity is highly toxic and is linked to numerous diseases, including neurodegeneration [70]. Such unregulated SOC activity can result in sustained [Ca2+]i increase and account in part for the general and neurotoxicity in NPC1.

Aberrant Ca2+ signaling that affects lysosomal function is also found in MLIV. MLIV is caused by mutations in the TRP channels TRPML1 [8, 32]. Our knowledge on the exact function of TRPML1 and how mutations in TRPML1 cause MLIV is very limited. The main reason is that so far none of the available clones is fully active. TRPML1 was shown to function as a Ca2+ selective channel [33, 34, 71] that can also transport Fe2+ [11]. However, the two functions were measured with TRPML1 mutant whose valine in the fifth transmembrane domain was replaced with proline. This mutation in the homologous TRPML3 causes the varitint-waddler phenotype [72] and results in gain-of-function [33, 34, 36, 73]. We already reported that the A419P mutation changes the channel properties of TRPML3 [74]. The same is very likely to be true for TRPML1 and thus the channel function of TRPML1 is not known with certainty. Nevertheless, our findings in mammalian cells [28, 49] and recent findings in Drosophila[75] indicate that TRPML1 regulates lysosomal pH by increasing H+ permeability of the lysosomes and reduces their acidity. The strong acidic lysosomal pH appears to be associated with inhibition of acidic lipase activity [49]. Indeed, pharmacological increase in lysosomal pH of fibroblasts from patients with MLIV reduced lysosomal storage [49] and changed the nature of the storage material [76].

Mitochondria and LSD

Common to all LSDs examined is aberrant mitochondrial function. This topic has been reviewed before [71] and only selective points are highlighted here. The mitochondria integrate several critical physiological functions and play central role in Ca2+ signaling. The mitochondria function to buffer cytoplasmic Ca2+ [77], control the activity of store operated channels (SOC) [70] and use Ca2+ to control cellular energetics and apoptosis [77]. Any change in mitochondrial function result in altered cellular Ca2+ response and cell toxicity, including neurotoxicity. This topic has been extensively covered in a recent special issue of Cell Calcium [55].

The Ca2+ signaling anomalies in LSDs can be caused or exacerbated by altered mitochondrial Ca2+ handling. This is illustrated in Figure 4, modified from [81], which shows mitochondrial function and its effect on cell signaling in cells obtained from patients with the LSD MLIV. The Figure shows that mitochondrial Ca2+ uptake is severely suppressed in MLIV cells, leading to the suggestion that cells affected by LSDs are extremely vulnerable to the pro-apoptotic effects of Ca2+.

Figure 4.

Figure 4

A model depicting energy-dependent excitotoxicity and the consequent cell death in LSDs. A. Dysregulation of mitochondrial Ca2+ uptake in LSDs (modified from [81]). Top panel: Human skin fibroblasts obtained from MLIV patients and matching controls were loaded with the fluorescent Ca2+ indicator Fura 2. Cytoplasmic Ca2+ spikes were induced by stimulating the cells with Bradykinin (Bk) and mitochondrial Ca2+ was released into the cytoplasm with the uncoupler FCCP. The magnitude of FCCP-induced Ca2+ spike is a measure of mitochondrial Ca2+content. Note that FCCP-induced Ca2+ release is significantly smaller in MLIV cells than in control cells. Bottom panel: Mitochondrial Ca2+uptake was measured directly with the mitochondrial Ca2+ dye Rhod 2 in control and MLIV cells during stimulation with Bk. Note the significantly suppressed mitochondrial Ca2+ uptake in MLV cells. B. A model of mitochondrial Ca2+ handling dysregulation and cell death in LSDs. In normal cells, a fraction of Ca2+ influx (1) induced by cell stimulation is absorbed by mitochondria (3) leading to an increase in ATP production (3). The increased ATP production stimulates Ca2+ extrusion by plasma membrane and ER Ca2+ pumps (4). In cells affected by LSDs, dysfunctional mitochondria that accumulate due to impaired autophagic recycling cannot absorb Ca2+ (5) and thus cannot respond to its buildup by increasing ATP production. The sustained increase in basal Ca2+ level leads to cell death (6).

Deficient Ca2+ signaling in cells affected by LSDs has been attributed to compromised mitochondrial function and resulting in energy–dependent excitotoxicity that causes cell death [78]. According to this model, defects in lysosomal function in LSDs negatively affect mitochondrial function, specificallyCa2+ uptake by mitochondria. Ca2+-dependent steps in the oxidative phosphorylation chain provide positive feedback mechanisms resulting in an increase in ATP production in response to flooding the cytoplasm (and mitochondria) with Ca2+. Such an increase in ATP production allows cells to boost the activity of Ca2+ pumps and to more efficiently regulate the Ca2+ signal. The loss of such a feedback loop, initially demonstrated in ovine model of ceroid lipofuscinosis [78], would make cells vulnerable to the pro-apoptotic effects of Ca2+ during neuronal activity. Hence, altered mitochondrial Ca2+ signaling and compromised mitochondrial Ca2+ buffering results in sustained [Ca2+]i increase during cellular activity, which triggers apoptosis [78] and necrosis [79].

How can LSDs lead to mitochondrial deficiencies? According to the “lysosomal-mitochondrial axis” model, lysosomal deficiencies in aged cells lead to reduced autophagy, and thus decreased mitochondria turnover and buildup of aged, fragmented mitochondria [10]. Accordingly, it is expected that cells affected by LSDs would demonstrate impaired autophagy [14, 20] and buildup of fragmented mitochondria, leading to imbalance Ca2+ handling by these cells [71, 80, 81]. The first part of this premise received significant experimental support in several LSDs: several recent reports demonstrate inhibition of autophagosomal formation in cells affected in LSDs [14, 17, 18, 20, 8183], although it is not clear at present whether the defective autophagy is due to up-regulated autophagic flow, or suppression of autophagy completion. Fragmentation of mitochondria was also observed in LSDs. This is illustrated in Figure 5 modified from [81], where fragmented mitochondria can be seen in cells from patients with MLII, MLIII, CLN2 and MLIV. Mitochondrial fragmentation similar to that observed in LSDs can be seen when autophagy in inhibited with 3MA or when membrane trafficking is inhibited by treatment with bafilomycin.

Figure 5.

Figure 5

Mitochondrial fragmentation in lysosomal storage diseases and under the conditions of suppressed autophagy or lysosomal function (modified from [81]). In order to visualize mitochondria, cells were stained with Rhodamine 123. Top two rows show control and LSDs affected cells; the bottom row shows control fibroblasts treated with the autophagy inhibitor 3-MA and with the lysosomal inhibitor Bafilomycin (Baf). Treatment and staining conditions can be found in [81].

According to the “lysosome-autophagy-mitochondria axis” model of cell death in Refs [71, 80, 81], suppressed autophagy in LSDs leads to buildup of aged mitochondria that cannot effectively buffer Ca2+, respond to flooding of the cytoplasm with Ca2+by increasing ATP production and inability to protect the cell from the pro-apoptotic effects of Ca2+ (illustrated in Figure 4). It should be noted that this model assumes only passive effect for the suppressed autophagy in LSDs on the mitochondrial status. It is, however, likely, that mitochondrial biogenesis also suffers from the block in autophagy.

In summary, it is becoming increasingly clear that LSDs are not simply due to cellular “indigestion”; rater they are complex cellular conditions that have a potential of improving our knowledge of cellular function and at the same time understanding affected cellular functions in LSDs should lead to better therapy. Studying the underlying causes of LSDs are likely to lead to identification of new lysosomal ion transporters and further illustrate the integrative function of the cell, the mechanisms that make cells work as whole. This is likely to lead to novel interventions in conditions in addition to LSDs that affect the status of the endocytic pathway, such as parasite invasions, and disease that affect the general cellular function, such as aging, dementia and cancer. It is also clear that anomalous Ca2+ signaling is playing a central role in the pathology of LSDs and perhaps controlled adjustment of the Ca2+ signal may prove a good site to interfere in progression of the disease.

Acknowledgements

The authors would like to extend their apologies to those authors whose work has not been cited here due to manuscript length consideration. Work in the authors’ labs is supported by the National Institutes of Health grants HD058577 and ES016782 to KK and the National Institutes of Health Grants DE12309 and DK38938 and the Ruth S. Harrell Professorship in Medical Research to S.M. S.Y. is supported by the Uehara Memorial Foundation, Tokyo, Japan.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Walkley SU. Pathogenic cascades in lysosomal disease-Why so complex? J Inherit Metab Dis. 2009;32(2):181–189. doi: 10.1007/s10545-008-1040-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Mach L. Biosynthesis of lysosomal proteinases in health and disease. Biol Chem. 2002;383(5):751–756. doi: 10.1515/BC.2002.078. [DOI] [PubMed] [Google Scholar]
  • 3.Mullins C, Bonifacino JS. The molecular machinery for lysosome biogenesis. Bioessays. 2001;23(4):333–343. doi: 10.1002/bies.1048. [DOI] [PubMed] [Google Scholar]
  • 4.Luzio JP, et al. Membrane traffic to and from lysosomes. Biochem Soc Symp. 2005 72;:77–86. doi: 10.1042/bss0720077. [DOI] [PubMed] [Google Scholar]
  • 5.Schilsky ML, Mistry P. Metabolic liver disease. Curr Opin Gastroenterol. 2001;17(3):221–231. doi: 10.1097/00001574-200105000-00005. [DOI] [PubMed] [Google Scholar]
  • 6.Anderson N, Borlak J. Drug-induced phospholipidosis. FEBS Lett. 2006;580(23):5533–5540. doi: 10.1016/j.febslet.2006.08.061. [DOI] [PubMed] [Google Scholar]
  • 7.Schneider P, Korolenko TA, Busch U. A review of drug-induced lysosomal disorders of the liver in man and laboratory animals. Microsc Res Tech. 1997;36(4):253–275. doi: 10.1002/(SICI)1097-0029(19970215)36:4<253::AID-JEMT4>3.0.CO;2-N. [DOI] [PubMed] [Google Scholar]
  • 8.Puertollano R, Kiselyov K. TRPMLs: in sickness and in health. Am J Physiol Renal Physiol. 2009;296(6):F1245–F1254. doi: 10.1152/ajprenal.90522.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Kurz T, et al. Lysosomes in iron metabolism, ageing and apoptosis. Histochem Cell Biol. 2008;129(4):389–406. doi: 10.1007/s00418-008-0394-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Stroikin Y, et al. Testing the "garbage" accumulation theory of ageing: mitotic activity protects cells from death induced by inhibition of autophagy. Biogerontology. 2005;6(1):39–47. doi: 10.1007/s10522-004-7382-y. [DOI] [PubMed] [Google Scholar]
  • 11.Dong XP, et al. The type IV mucolipidosis-associated protein TRPML1 is an endolysosomal iron release channel. Nature. 2008;455(7215):992–996. doi: 10.1038/nature07311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Nixon RA, Yang DS, Lee JH. Neurodegenerative lysosomal disorders: a continuum from development to late age. Autophagy. 2008;4(5):590–599. doi: 10.4161/auto.6259. [DOI] [PubMed] [Google Scholar]
  • 13.Weimer JM, et al. The neuronal ceroid lipofuscinoses: mutations in different proteins result in similar disease. Neuromolecular Med. 2002;1(2):111–124. doi: 10.1385/NMM:1:2:111. [DOI] [PubMed] [Google Scholar]
  • 14.Venugopal B, et al. Chaperone-mediated autophagy is defective in mucolipidosis type IV. J Cell Physiol. 2009;219(2):344–353. doi: 10.1002/jcp.21676. [DOI] [PubMed] [Google Scholar]
  • 15.Todde V, Veenhuis M, van der Klei IJ. Autophagy: principles and significance in health and disease. Biochim Biophys Acta. 2009;1792(1):3–13. doi: 10.1016/j.bbadis.2008.10.016. [DOI] [PubMed] [Google Scholar]
  • 16.Ruivo R, et al. Molecular and cellular basis of lysosomal transmembrane protein dysfunction. Biochim Biophys Acta. 2009;1793(4):636–649. doi: 10.1016/j.bbamcr.2008.12.008. [DOI] [PubMed] [Google Scholar]
  • 17.Martina JA, Lelouvier B, Puertollano R. The calcium channel mucolipin-3 is a novel regulator of trafficking along the endosomal pathway. Traffic. 2009;10(8):1143–1156. doi: 10.1111/j.1600-0854.2009.00935.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Kim HJ, et al. The Ca(2+) channel TRPML3 regulates membrane trafficking and autophagy. Traffic. 2009;10(8):1157–1167. doi: 10.1111/j.1600-0854.2009.00924.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Ballabio A, Gieselmann V. Lysosomal disorders: from storage to cellular damage. Biochim Biophys Acta. 2009;1793(4):684–696. doi: 10.1016/j.bbamcr.2008.12.001. [DOI] [PubMed] [Google Scholar]
  • 20.Vergarajauregui S, et al. Autophagic dysfunction in mucolipidosis type IV patients. Hum Mol Genet. 2008;17(17):2723–2737. doi: 10.1093/hmg/ddn174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Eskelinen EL, Saftig P. Autophagy: a lysosomal degradation pathway with a central role in health and disease. Biochim Biophys Acta. 2009;1793(4):664–673. doi: 10.1016/j.bbamcr.2008.07.014. [DOI] [PubMed] [Google Scholar]
  • 22.Terman A, et al. Lysosomal labilization. IUBMB Life. 2006;58(9):531–539. doi: 10.1080/15216540600904885. [DOI] [PubMed] [Google Scholar]
  • 23.Pryor PR, et al. The role of intraorganellar Ca(2+) in late endosome-lysosome heterotypic fusion and in the reformation of lysosomes from hybrid organelles. J Cell Biol. 2000;149(5):1053–1062. doi: 10.1083/jcb.149.5.1053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Luzio JP, Bright NA, Pryor PR. The role of calcium and other ions in sorting and delivery in the late endocytic pathway. Biochem Soc Trans. 2007;35(Pt 5):1088–1091. doi: 10.1042/BST0351088. [DOI] [PubMed] [Google Scholar]
  • 25.Sudhof TC. The synaptic vesicle cycle. Annu Rev Neurosci. 2004;27:509–547. doi: 10.1146/annurev.neuro.26.041002.131412. [DOI] [PubMed] [Google Scholar]
  • 26.Bakker AC, et al. Homotypic fusion between aggregated lysosomes triggered by elevated [Ca2+]i in fibroblasts. J Cell Sci. 1997;110(Pt 18):2227–2238. doi: 10.1242/jcs.110.18.2227. [DOI] [PubMed] [Google Scholar]
  • 27.Lloyd-Evans E, et al. Niemann-Pick disease type C1 is a sphingosine storage disease that causes deregulation of lysosomal calcium. Nat Med. 2008;14(11):1247–1255. doi: 10.1038/nm.1876. [DOI] [PubMed] [Google Scholar]
  • 28.Miedel MT, et al. Membrane traffic and turnover in TRP-ML1-deficient cells: a revised model for mucolipidosis type IV pathogenesis. J Exp Med. 2008;205(6):1477–1490. doi: 10.1084/jem.20072194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.LaPlante JM, et al. Functional links between mucolipin-1 and Ca2+-dependent membrane trafficking in mucolipidosis IV. Biochem Biophys Res Commun. 2004;322(4):1384–1391. doi: 10.1016/j.bbrc.2004.08.045. [DOI] [PubMed] [Google Scholar]
  • 30.LaPlante JM, et al. 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. 2002;532(1–2):183–187. doi: 10.1016/s0014-5793(02)03670-0. [DOI] [PubMed] [Google Scholar]
  • 31.Treusch S, et al. Caenorhabditis elegans functional orthologue of human protein h-mucolipin-1 is required for lysosome biogenesis. Proc Natl Acad Sci U S A. 2004;101(13):4483–4488. doi: 10.1073/pnas.0400709101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kiselyov K, Soyombo A, Muallem S. TRPpathies. J Physiol. 2007;578(Pt 3):641–653. doi: 10.1113/jphysiol.2006.119024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Xu H, et al. Activating mutation in a mucolipin transient receptor potential channel leads to melanocyte loss in varitint-waddler mice. Proc Natl Acad Sci U S A. 2007;104(46):18321–18326. doi: 10.1073/pnas.0709096104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Grimm C, et al. A helix-breaking mutation in TRPML3 leads to constitutive activity underlying deafness in the varitint-waddler mouse. Proc Natl Acad Sci U S A. 2007 doi: 10.1073/pnas.0709846104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Nagata K, et al. The varitint-waddler (Va) deafness mutation in TRPML3 generates constitutive, inward rectifying currents and causes cell degeneration. Proc Natl Acad Sci U S A. 2008;105(1):353–358. doi: 10.1073/pnas.0707963105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kim HJ, et al. Gain-of-function mutation in TRPML3 causes the mouse varitint-waddler phenotype. J Biol Chem. 2007 doi: 10.1074/jbc.C700190200. [DOI] [PubMed] [Google Scholar]
  • 37.Brailoiu E, et al. Essential requirement for two-pore channel 1 in NAADP-mediated calcium signaling. J Cell Biol. 2009;186(2):201–209. doi: 10.1083/jcb.200904073. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Calcraft PJ, et al. NAADP mobilizes calcium from acidic organelles through two-pore channels. Nature. 2009;459(7246):596–600. doi: 10.1038/nature08030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Churchill GC, et al. NAADP mobilizes Ca(2+) from reserve granules, lysosome-related organelles, in sea urchin eggs. Cell. 2002;111(5):703–708. doi: 10.1016/s0092-8674(02)01082-6. [DOI] [PubMed] [Google Scholar]
  • 40.Furuichi T, Cunningham KW, Muto S. A putative two pore channel AtTPC1 mediates Ca(2+) flux in Arabidopsis leaf cells. Plant Cell Physiol. 2001;42(9):900–905. doi: 10.1093/pcp/pce145. [DOI] [PubMed] [Google Scholar]
  • 41.Pottosin, Schonknecht G. Vacuolar calcium channels. J Exp Bot. 2007;58(7):1559–1569. doi: 10.1093/jxb/erm035. [DOI] [PubMed] [Google Scholar]
  • 42.Beyenbach KW, Wieczorek H. The V-type H+ ATPase: molecular structure and function, physiological roles and regulation. J Exp Biol. 2006;209(Pt 4):577–589. doi: 10.1242/jeb.02014. [DOI] [PubMed] [Google Scholar]
  • 43.Jentsch TJ. CLC chloride channels and transporters: from genes to protein structure, pathology and physiology. Crit Rev Biochem Mol Biol. 2008;43(1):3–36. doi: 10.1080/10409230701829110. [DOI] [PubMed] [Google Scholar]
  • 44.Jentsch TJ. Chloride and the endosomal-lysosomal pathway: emerging roles of CLC chloride transporters. J Physiol. 2007;578(Pt 3):633–640. doi: 10.1113/jphysiol.2006.124719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Kasper D, et al. Loss of the chloride channel ClC-7 leads to lysosomal storage disease and neurodegeneration. Embo J. 2005;24(5):1079–1091. doi: 10.1038/sj.emboj.7600576. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Paroutis P, Touret N, Grinstein S. The pH of the secretory pathway: measurement, determinants, and regulation. Physiology (Bethesda) 2004;19:207–215. doi: 10.1152/physiol.00005.2004. [DOI] [PubMed] [Google Scholar]
  • 47.Steinberg BE, et al. In situ measurement of the electrical potential across the phagosomal membrane using FRET and its contribution to the proton-motive force. Proc Natl Acad Sci U S A. 2007;104(22):9523–9528. doi: 10.1073/pnas.0700783104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Thompson EG, et al. Lysosomal trafficking functions of mucolipin-1 in murine macrophages. BMC Cell Biol. 2007;8:54. doi: 10.1186/1471-2121-8-54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Soyombo AA, et al. TRP-ML1 regulates lysosomal pH and acidic lysosomal lipid hydrolytic activity. J Biol Chem. 2006;281(11):7294–7301. doi: 10.1074/jbc.M508211200. [DOI] [PubMed] [Google Scholar]
  • 50.Tang Y, Li H, Liu JP. Niemann-Pick Disease Type C: From Molecule To Clinic. Clin Exp Pharmacol Physiol. 2009 doi: 10.1111/j.1440-1681.2009.05235.x. [DOI] [PubMed] [Google Scholar]
  • 51.Pelled D, et al. Enhanced calcium release in the acute neuronopathic form of Gaucher disease. Neurobiol Dis. 2005;18(1):83–88. doi: 10.1016/j.nbd.2004.09.004. [DOI] [PubMed] [Google Scholar]
  • 52.Wong K, et al. Neuropathology provides clues to the pathophysiology of Gaucher disease. Mol Genet Metab. 2004;82(3):192–207. doi: 10.1016/j.ymgme.2004.04.011. [DOI] [PubMed] [Google Scholar]
  • 53.Gerasimenko JV, et al. NAADP, cADPR and IP3 all release Ca2+ from the endoplasmic reticulum and an acidic store in the secretory granule area. J Cell Sci. 2006;119(Pt 2):226–238. doi: 10.1242/jcs.02721. [DOI] [PubMed] [Google Scholar]
  • 54.Bezprozvanny I. Calcium signaling and neurodegenerative diseases. Trends Mol Med. 2009;15(3):89–100. doi: 10.1016/j.molmed.2009.01.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Duchen MR, Verkhratsky A, Muallem S. Mitochondria and calcium in health and disease. Cell Calcium. 2008;44(1):1–5. doi: 10.1016/j.ceca.2008.02.001. [DOI] [PubMed] [Google Scholar]
  • 56.Korkotian E, et al. Elevation of intracellular glucosylceramide levels results in an increase in endoplasmic reticulum density and in functional calcium stores in cultured neurons. J Biol Chem. 1999;274(31):21673–21678. doi: 10.1074/jbc.274.31.21673. [DOI] [PubMed] [Google Scholar]
  • 57.Mu TW, Fowler DM, Kelly JW. Partial restoration of mutant enzyme homeostasis in three distinct lysosomal storage disease cell lines by altering calcium homeostasis. PLoS Biol. 2008;6(2):e26. doi: 10.1371/journal.pbio.0060026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Rigat B, Mahuran D. Diltiazem, a L-type Ca(2+) channel blocker, also acts as a pharmacological chaperone in Gaucher patient cells. Mol Genet Metab. 2009;96(4):225–232. doi: 10.1016/j.ymgme.2008.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Reddy JV, Ganley IG, Pfeffer SR. Clues to neuro-degeneration in Niemann-Pick Type C disease from global gene expression profiling. PLoS One. 2006;1:e19. doi: 10.1371/journal.pone.0000019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Sun Y, et al. In vivo and ex vivo evaluation of L-type calcium channel blockers on acid beta-glucosidase in Gaucher disease mouse models. PLoS One. 2009;4(10):e7320. doi: 10.1371/journal.pone.0007320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Pelled D, et al. 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. 2003;278(32):29496–29501. doi: 10.1074/jbc.M302964200. [DOI] [PubMed] [Google Scholar]
  • 62.Deisz RA, et al. Pathological cholesterol metabolism fails to modify electrophysiological properties of afflicted neurones in Niemann-Pick disease type C. Neuroscience. 2005;130(4):867–873. doi: 10.1016/j.neuroscience.2004.09.065. [DOI] [PubMed] [Google Scholar]
  • 63.Tanaka J, et al. Immunohistochemical expression of P400 protein in Purkinje cells of sphingomyelinosis mouse. Brain Dev. 1991;13(2):110–114. doi: 10.1016/s0387-7604(12)80117-3. [DOI] [PubMed] [Google Scholar]
  • 64.Ginzburg L, Futerman AH. Defective calcium homeostasis in the cerebellum in a mouse model of Niemann-Pick A disease. J Neurochem. 2005;95(6):1619–1628. doi: 10.1111/j.1471-4159.2005.03534.x. [DOI] [PubMed] [Google Scholar]
  • 65.Bilmen JG, et al. Inhibition of the SERCA Ca2+ pumps by curcumin. Curcumin putatively stabilizes the interaction between the nucleotide-binding and phosphorylation domains in the absence of ATP. Eur J Biochem. 2001;268(23):6318–6327. doi: 10.1046/j.0014-2956.2001.02589.x. [DOI] [PubMed] [Google Scholar]
  • 66.Egan ME, et al. Curcumin, a major constituent of turmeric, corrects cystic fibrosis defects. Science. 2004;304(5670):600–602. doi: 10.1126/science.1093941. [DOI] [PubMed] [Google Scholar]
  • 67.Yuan JP, et al. TRPC channels as STIM1-regulated SOCs. Channels (Austin) 2009;3(4):221–225. doi: 10.4161/chan.3.4.9198. [DOI] [PubMed] [Google Scholar]
  • 68.Roos J, et al. STIM1, an essential and conserved component of store-operated Ca2+ channel function. J Cell Biol. 2005;169(3):435–445. doi: 10.1083/jcb.200502019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Zhang SL, et al. STIM1 is a Ca2+ sensor that activates CRAC channels and migrates from the Ca2+ store to the plasma membrane. Nature. 2005;437(7060):902–905. doi: 10.1038/nature04147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Parekh AB, Putney JW., Jr. Store-operated calcium channels. Physiol Rev. 2005;85(2):757–810. doi: 10.1152/physrev.00057.2003. [DOI] [PubMed] [Google Scholar]
  • 71.Kiselyov K, Muallem S. Mitochondrial Ca(2+) homeostasis in lysosomal storage diseases. Cell Calcium. 2008;44(1):103–111. doi: 10.1016/j.ceca.2007.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Di Palma F, et al. Mutations in Mcoln3 associated with deafness and pigmentation defects in varitint-waddler (Va) mice. Proc Natl Acad Sci U S A. 2002;99(23):14994–14999. doi: 10.1073/pnas.222425399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Cuajungco MP, Samie MA. The varitint-waddler mouse phenotypes and the TRPML3 ion channel mutation: cause and consequence. Pflugers Arch. 2008 doi: 10.1007/s00424-008-0523-4. [DOI] [PubMed] [Google Scholar]
  • 74.Kim HJ, et al. A novel mode of TRPML3 regulation by extracytosolic pH absent in the varitint-waddler phenotype. Embo J. 2008;27(8):1197–1205. doi: 10.1038/emboj.2008.56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Venkatachalam K, et al. Motor deficit in a Drosophila model of mucolipidosis type IV due to defective clearance of apoptotic cells. Cell. 2008;135(5):838–851. doi: 10.1016/j.cell.2008.09.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kogot-Levin A, et al. Mucolipidosis type IV: the effect of increased lysosomal pH on the abnormal lysosomal storage. Pediatr Res. 2009;65(6):686–690. doi: 10.1203/PDR.0b013e3181a1681a. [DOI] [PubMed] [Google Scholar]
  • 77.Rizzuto R, Pozzan T. Microdomains of intracellular Ca2+: molecular determinants and functional consequences. Physiol Rev. 2006;86(1):369–408. doi: 10.1152/physrev.00004.2005. [DOI] [PubMed] [Google Scholar]
  • 78.Jolly RD, et al. Mitochondrial dysfunction in the neuronal ceroid-lipofuscinoses (Batten disease) Neurochem Int. 2002;40(6):565–571. doi: 10.1016/s0197-0186(01)00128-0. [DOI] [PubMed] [Google Scholar]
  • 79.Pinton P, et al. Calcium and apoptosis: ER-mitochondria Ca2+ transfer in the control of apoptosis. Oncogene. 2008;27(50):6407–6418. doi: 10.1038/onc.2008.308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kiselyov K, et al. Autophagy, mitochondria and cell death in lysosomal storage diseases. Autophagy. 2007;3(3):259–262. doi: 10.4161/auto.3906. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Jennings JJ, Jr., et al. Mitochondrial aberrations in mucolipidosis Type IV. J Biol Chem. 2006;281(51):39041–39050. doi: 10.1074/jbc.M607982200. [DOI] [PubMed] [Google Scholar]
  • 82.Tessitore A, Pirozzi M, Auricchio A. Abnormal autophagy, ubiquitination, inflammation and apoptosis are dependent upon lysosomal storage and are useful biomarkers of mucopolysaccharidosis VI. Pathogenetics. 2009;2(1):4. doi: 10.1186/1755-8417-2-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Takamura A, et al. Enhanced autophagy and mitochondrial aberrations in murine G(M1)-gangliosidosis. Biochem Biophys Res Commun. 2008;367(3):616–622. doi: 10.1016/j.bbrc.2007.12.187. [DOI] [PubMed] [Google Scholar]

RESOURCES