Next Article in Journal
Regenerating Skeletal Muscle Compensates for the Impaired Macrophage Functions Leading to Normal Muscle Repair in Retinol Saturase Null Mice
Next Article in Special Issue
Functional Role of STIM-1 and Orai1 in Human Microvascular Aging
Previous Article in Journal
RMR-Related DNAJC6 Expression Suppresses Adipogenesis in 3T3-L1 Cells
Previous Article in Special Issue
On the Connections between TRPM Channels and SOCE
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Store-Operated Calcium Entry and Its Implications in Cancer Stem Cells

Cellular Physiology Research Group, Department of Physiology, Institute of Molecular Pathology Biomarkers, University of Extremadura, 10003 Caceres, Spain
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cells 2022, 11(8), 1332; https://doi.org/10.3390/cells11081332
Submission received: 2 March 2022 / Revised: 4 April 2022 / Accepted: 12 April 2022 / Published: 13 April 2022
(This article belongs to the Collection STIM and Orai Communication in Health and Disease)

Abstract

:
Tumors are composed by a heterogeneous population of cells. Among them, a sub-population of cells, termed cancer stem cells, exhibit stemness features, such as self-renewal capabilities, disposition to differentiate to a more proliferative state, and chemotherapy resistance, processes that are all mediated by Ca2+. Ca2+ homeostasis is vital for several physiological processes, and alterations in the patterns of expressions of the proteins and molecules that modulate it have recently become a cancer hallmark. Store-operated Ca2+ entry is a major mechanism for Ca2+ entry from the extracellular medium in non-excitable cells that leads to increases in the cytosolic Ca2+ concentration required for several processes, including cancer stem cell properties. Here, we focus on the participation of STIM, Orai, and TRPC proteins, the store-operated Ca2+ entry key components, in cancer stem cell biology and tumorigenesis.

Graphical Abstract

1. Introduction

Normal stem cells are undifferentiated or partially differentiated cells that are characterized by their ability to self-renew, the process of bringing about indefinitely more cells of the same type, as well as to differentiate in more specialized mature cells. The term “stem cell” was coined by Ernst Haeckel in 1868 to describe the ancestor unicellular organism from which all multicellular organisms were supposed to evolve [1]. Normal stem cells can be found from the early embryos to the mature subject, where they can be present in different tissues, including the bone marrow, skin and hair follicles, muscle, brain, and epithelia, among others [2].
Cancer stem cells (CSC), also known as tumor-initiating cells, share features of both cancer and stem cells. These cells constitute a sub-population of tumor-resident malignant cells responsible for recurrence, metastasis formation, and chemoresistance. Experimental evidence indicates that CSC exhibit “stemness” properties, that is, the ability of cells to perpetuate their lineage, to bring about differentiated cells and to interact with their microenvironment to maintain a balance between quiescence, proliferation, and regeneration [3]. According to this, CSC exhibit low proliferative rates, self-renewing capacity, propensity to differentiate into proliferating tumor cells, resistance to apoptosis and senescence, as well as to chemo- and radio-therapy, evasion of immune attack, and are responsible for invasion and metastases [4,5].
CSC have been reported to derive from “normal” tissue resident stem cells or from differentiated cells undergoing transformation [6]. Furthermore, during cancer evolution, secondary self-renewing cell populations might arise, which supports the notion that the CSC phenotype might not be exclusively defined by the intrinsic characteristics of a cell but might also be determined by other phenomena, such as the microenvironment interaction [7]. The existence of CSC has been demonstrated in a variety of tumors, from leukemia [8] to solid tumors, such as colon [9], breast [10], brain [11], pancreatic [12], oral [13], esophageal [14], and liver [15] cancers as well as melanoma [16], among others. The functional role of CSC in tumor initiation is complex and not completely resolved. In contrast to the stochastic model that hypothesizes that most tumor cells can act as tumor initiating cells [17], the hierarchical or CSC hypothesis assumes that only a sub-population of cells, the CSC, have the ability for tumor initiation [18]. According to this hypothesis, CSC are responsible for the initiation, metastasis, chemotherapy resistance, and recurrence of the tumor [19]. Nevertheless, the CSC hypothesis applies to a limited number of cases. For instance, in certain tumors, such as testicular cancer, CSC are more sensitive to cisplatin than the differentiated tumor cells. Furthermore, in glioblastoma multiforme, a large number of differentiated tumor cells survive after anti-tumoral therapy, not just a small sub-population of CSC, and many of the surviving cells exhibit the ability of re-initiating the tumor. Therefore, the analysis of the complexity of the functional role of CSC in the context of neoplasia deserves further studies.
CSC show similar surface markers as normal stem cells in a given tissue, but a number of cell surface and intracellular biomarkers are commonly used to identify CSC among differentiated tumor cells and to isolate them. These markers include the clusters of differentiation (CD) CD44, CD24, or CD133, among others, the epithelial cell adhesion molecule (EpCAM) or the intracellular markers aldehyde dehydrogenase-1 (ALDH1) and the Notch, Wnt/β-catenin, Nanog, Sox2 pathways (for a more extensive review of CSC biomarkers in different tumors please see [20]). CD44 is expressed in CSC, as well as in a variety of normal stem cells, and plays an important role in CSC self-renewal and proliferation, leading to tumor growth, tumor metastasis, the activation of stemness transcription factors such as Nanog, Sox2, and Oct4, and chemotherapy resistance [21]. CD44 is a highly conserved surface glycoprotein encoded by the CD44 gene. The pre-mRNA contains 20 exons, where exons 1–5 and 16–20 are constant exons that lead to the standard form of 85 kDa while the remaining 10 exons (exons 6–15) are variant exons subjected to alternative splicing to produce the different CD44 variant forms [22]. The CD44v isoform is predominantly expressed in CSC over normal stem cells [23]. CD24 is a cell surface protein, heavily glycosylated, that plays an important role in cell-cell and cell-matrix interactions [24]. The expression of CD24 has commonly been investigated in combination with CD44 and other markers. For instance, high expression of CD44 and low expression of CD24 (CD44+/CD24−/low) together with expression of ALDH1 is a feature of breast cancer stem cells as compared to non-stem breast cancer cells [25]. Nevertheless, the expression of CD24 is variable among cancer cells [26]. ALDH1 has been reported as a CSC marker in adult tumors and, specially, is a bona fide marker of breast normal and cancer stem cells [27]. Interestingly, the ALDH1A3 isoform is predominantly expressed in CSC over normal stem cells [28]. The function of ALDH1 in CSC differentiation has been associated to its function in the oxidation of retinol to retinoic acid [29]; furthermore, positive ALDH1 expression has been reported to be correlated with chemotherapy resistance and poor prognosis [30].
Different signaling pathways and transcription factors have been reported to play an essential role in the state of cell stemness. Among them, developmental signaling pathways such as Notch, Wnt/β-catenin or Hedgehog play important roles in normal stem cell function. Notch signaling pathway is activated by the interaction of ligands of the DSL family with the receptor protein Notch, a single-pass transmembrane protein. The interaction of the ligand with the Notch extracellular domain (NECD) leads to the cleavage and release of the Notch intracellular domain (NICD) that acts as a transcription factor and interacts with the transcription factors of the CSL family and Mastermind [31]. The CSL-Notch-Mastermind transcription factor complex up-regulates transcription of Notch-responsive genes leading to cell proliferation and promoting the formation of CSC colonies in different cancer types, including glioma and colon and breast cancer [32] (Figure 1).
The Hedgehog (Hh) pathway also plays a major role in normal stem cell and CSC biology and tumorigenesis. Hh signaling begins with the interaction of Hh ligands (Sonic Hedgehog, Indian Hedgehog, and Desert Hedgehog) with Patched-1, and to a lesser extent Patched-2, a twelve-pass transmembrane protein receptor. This process internalizes the Patched receptor and relieves the constitutive repression of the G-protein-coupled receptor Smoothened, which, in turn, leads to the nuclear translocation or the transcription factor Glioma-associated oncogene (Gli) resulting in the transcription of Hh target genes [33] (Figure 1). The Hh pathway has been associated to chemotherapy resistance and disease relapse [33].
The Wnt/β-catenin signaling pathway promotes CSC self-renewal while reducing the differentiation of CSC to proliferating tumor cells [32]. The Wnt ligands, a large family of secreted glycoproteins, interact with a Frizzled receptor in the plasma membrane, which signaling through the protein Dishevelled (Dvl) leads to inhibition of phosphorylation and proteasomal degradation of the protein β-catenin. Then, β-catenin accumulates in the cytosol and translocates into the nucleus, thus promoting the transcript of Wnt target genes by a mechanism involving the T-cell factor and lymphoid enhancer factor-1 (TCF/LEF1) transcription factors [34] (Figure 1). In addition to the activation of the β-catenin/TCF/LEF1 transcriptional pathway, Wnt proteins can induce alternative or non-canonical signaling pathways. In this pathway, Dvl is linked through Daam1 (Dishevelled associated activator of morphogenesis 1) to allow activation of the small GTPases Rho and Rac, which, in turn, activate Rho-kinase and JNK, respectively. Another non-canonical Wnt process is the Wnt/Ca2+ pathway. The interaction of Wnts with Frizzled, a family of G-protein coupled receptors, leads to the activation of phospholipase C and, thus, IP3 (inositol 1,4,5-triphosphate)-dependent Ca2+ release from the intracellular stores and subsequent Ca2+ influx across the plasma membrane [35]. The intracellular calcium fluxes induce the activation of downstream effects, such as PKC, CaMKII or calcineurin, thus leading to the nuclear translocation and activation of NFAT (nuclear factor of activated T-cells; Figure 1) [36]. A reciprocal interaction between Wnt signaling and NF-κB has been reported to play a key role in the progression of inflammation and cancer [37].

2. Calcium Signaling in Cancer Stem Cells and Cancer Hallmarks

Calcium ion modulates a myriad of physiological processes, such as muscle contraction, secretion or gene transcription, through a sophisticated and well-orchestrated machinery that deftly tunes cytosolic Ca2+ concentration [38]. In addition, Ca2+ participates in several pathological conditions, including cancer. Resistance to apoptosis and chemotherapy, high proliferation rate or the ability to migrate and to invade different tissues, have been considered key features in all cancer types for years [39]. As stated above, CSCs share some of these properties, but they also exhibit unique abilities such as capability for self-renewal [4,5]. All those processes are modulated by Ca2+ [40], thus, in recent years, aberrant expression of the proteins that control Ca2+ homeostasis has been included as a cancer hallmark.
Increases in intracellular Ca2+ concentration are required to trigger several Ca2+-dependent downstream effectors that modulate cellular pathways, such as calmodulin, which is essential for cell cycle and proliferation [41], NFAT proteins, with a role in cell cycle, differentiation or tumorigenesis [42], or the mitogen-activated protein kinase/extracellular signal-regulated (MAPK/ERK) pathway, involved in cancer cell survival, metastasis and chemotherapy resistance [43,44]. Increments of cytosolic Ca2+ concentration is achieved by the cells either by releasing Ca2+ from intracellular reservoirs, or by the opening of Ca2+-permeable channels in the plasma membrane (PM), which ensures an unlimited source of Ca2+ influx from the extracellular medium. Recent reports have demonstrated that CSC exhibit altered function in those mechanism (Table 1). Furthermore, it has been demonstrated that different signaling pathways that contribute to CSC pluripotency, such as the Wnt, TGF-β or FGF2, actively tune cytoplasmic Ca2+ concentration [45,46,47].
Concerning the Ca2+ release from intracellular stores, two calcium channels located in the endoplasmic reticulum (ER) membrane, IP3- and ryanodine (Ry)- receptors, are vital for CSC stemness, proliferation, and metastasis in different cancer types, such as glioblastoma [48], melanoma [49], and breast cancer [50].
Regarding Ca2+ entry, CSC present channels in the plasma membrane that are permeable to Ca2+ and could be gated by a variety of stimuli, such as voltage, second messengers or depletion of intracellular Ca2+ stores. For instance, over-expression of L- and T-type voltage-dependent Ca2+ channels is involved in tumorigenesis, proliferation, migration, and resistance to drugs in ovarian and glioblastoma CSC [51,52,53,54]. Moreover, abnormal expression of the voltage-dependent Ca2+ channel α2δ1 subunit, which modulates Ca2+ oscillation amplitude and the expression of different genes by keeping transcription factors in the nucleus, has started to be considered a tumoral marker in many cancers, such as lung [55,56], breast [57], and liver cancer [58] or laryngeal squamous [59], with a major role in CSC expansion. Different members of the transient receptor potential (TRP) channels, which are activated by several stimuli, such as temperature, pressure, and second messengers, participate in Ca2+ entry and have a key role in the CSC physiopathology of different cancers. For instance, TRPC3, which is over-expressed in triple-negative breast cancer cells, is activated by lysophosphatidic acid, promoting the process of self-renewal in CSC [60]. Enhanced expression or activation of TRPM7 has also been characterized in lung [61], glioblastoma [62], or neuroblastoma [63] and linked with several features of CSC. Similar findings have been found for TRPV2 channels-gated, among others, by the lipid ligand lysophosphatidylcholine [64] in the stemness of esophageal CSC [65]. Conversely, TRPV2 activation and expression promotes loss of stemness and apoptotic cell death in glioma [64,66,67] and hepatocellular carcinoma CSC [68]. This inverse correlation has also been observed for TRPA1 and TRPV1 channels in glioblastoma CSC [69].
Ca2+ reuptake, and the proteins involved, such as Ca2+-ATPases, Ca2+ exchangers or mitochondrial uniporter [70,71,72] might also play a role in CSC biology. A recent study has demonstrated that the sarco/endoplasmic Ca2+ ATPase (SERCA) presents an important antiapoptotic function in breast CSC, by reducing Ca2+-dependent apoptosis during glucose deprivation. This process is mediated by CaMK2α, which triggers the activation of NF-κB, and, in turn, SERCA over-expression [73]. Another report has shown that the mitochondrial Ca2+ uniporter and the Na+/Ca2+ exchanger, located in mitochondria and in the plasma membrane, respectively, are highly expressed in glioblastoma CSCs [74]; however, the relevance of such an aberrant protein expression remains yet unclear.

3. Store-Operated Calcium Entry in Cancer Stem Cells and Cancer Hallmarks

Store-Operated Calcium Entry (SOCE), a major mechanism for Ca2+ influx from the extracellular medium into excitable and non-excitable cells, is physiologically triggered by the activation of phospholipase C (PLC) and the production of IP3, which subsequently leads to the release of Ca2+ from intracellular stores, mainly the ER, resulting in the activation of store-operated calcium channels in the plasma membrane and a rapid increase in cytosolic Ca2+ concentration [75,76]. SOCE is an extremely complex biological mechanism, with high dependency on the pattern of expression of its components-STIMs, Orai, and TRPC proteins- and its modulators in each cell type. Since the last decades of the 20th century, several studies, both in vivo and in vitro, have reported that an altered expression pattern of the proteins that mediate SOCE leads to unbalanced Ca2+ homeostasis, which might contribute to tumor development, poor prognosis, and chemotherapeutic drug resistance [77].
The proteins of the STromal Interaction Molecule (STIM) family, STIM1 and STIM2, and their splice variants, possess a single transmembrane domain, with the N-region located either in the ER lumen or the extracellular medium, and a long cytosolic C-region [78,79]. Both, N- and C-terminal regions, present several key domains that enact STIM proteins’ double function upon a diminishment of the luminal Ca2+ concentration in the intracellular stores: (1) as the Ca2+ sensors of intracellular organelles, mediated by EF-hand Ca2+-binding domains in the N-terminus; and (2) as the transmitters of the filling state of intracellular Ca2+ stores to, and the activators of, Ca2+ channels in the plasma membrane. The latter is achieved by direct interaction between different domains within the STIM cytosolic C-region and the store-operated Ca2+ channels (STIM proteins structure is reviewed in [80,81,82]).
SOCE could be mediated by two types of channels with different biophysical properties: (1) the Ca2+ Release-Activated Ca2+ (CRAC) channels that exhibit high Ca2+ selectivity and an inwardly rectifying current, termed ICRAC, which its exclusively conducted by members of the Orai family [83]; and (2) the Store-Operated Ca2+ (SOC) channels, responsible to mediate a non-selective cation current denominated ISOC, formed by both, Orai1 and TRPC1, the first identified member of the canonical Transient Receptor Potential (TRPC) channel subfamily [84,85].
Orai1 was initially characterized as the main component of CRAC channel during a RNAi screening in 2006, when it was found that the Orai1 R91W mutation was responsible for abrogated CRAC channel function, critical for T-cell activation, in immunodeficient patients [86]. Orai1 and its paralogues, Orai2 and Orai3, present a unique structure among other Ca2+ channels, with four transmembrane domains spanning the PM and both, N- and C-terminus, facing the cytoplasm [87]. Originally, it was thought that Orai channels were formed by a homo-tetramer [88]; however, the crystal structure from Drosophila melanogaster Orai1 (dOrai1) presented a hexamer configuration, with the ion pore formed by the first transmembrane domain of the Orai subunits and located in the center of the complex surrounded by the remaining Orai plasma membrane domains [89]. The three members of the Orai family are capable to mediate store dependent Ca2+ influx, each of them with different biophysical properties that are extensively discussed here [90,91]. Some years ago, a shorter splicing variant for Orai1, Orai1β, lacking 64 aa in the N-terminus but able to generate functional Orai1 channels, was identified. Orai1β can be fully activated by STIM1 in a store-dependent manner but exhibits differential inactivation patterns as compared with the long variant, Orai1α [85]. In addition, recent studies have shown that Orai proteins might have a role in non-capacitative Ca2+ influx forming heteromers, such as the arachidonate-regulated Ca2+ channels (ARC), where three Orai1 and two Orai3 subunits form a pentamer [92], or interacting with other proteins to mediate store-independent Ca2+ influx [93].
TRPC1 belongs to the TRP channel superfamily, whose members ubiquitously mediate ion fluxes across the whole animal kingdom in a cell type-dependent manner [94]. All TRPs possess a similar structure with six transmembrane domains and the pore located between the 5th and 6th transmembrane regions. TRPs exhibit N- and C-terminus of variable length, containing the TRP box and different functional domains, subfamily-dependent, which participate in the functions of TRP channels and their relationship with other molecules and proteins. A functional TRP channel is composed by four TRP subunits forming either a homo- or hetero-tetramer [95,96]. Prior to Orai1 characterization, TRPC1 was a suggested candidate as the channel responsible for SOCE as STIM1 is able to interact and activate TRPC1 channels [97,98]. The current hypothesis suggests that TRPC1, together with Orai1, is involved in the generation of ISOC currents [85,99,100,101]. TRPC1 channels, permeable to Na+, Ca2+, and Cs+ [102], are less selective for Ca2+ than Orai1 and allow a massive ion influx from the extracellular medium, required for the maintenance of SOCE and store replenishment [103].
Several stimuli might trigger intracellular Ca2+ stores depletion that will be sensed by STIM proteins (Figure 2). Minor reductions in luminal Ca2+ concentration will be detected by STIM2, which in turn, would momentarily trigger the opening of CRAC channels, allowing Ca2+ influx from the extracellular medium that will quickly be reintroduced into the stores by Ca2+-ATPase pumps to revert to resting conditions (Figure 2b) [80]. More extensive discharge of intracellular Ca2+ stores would trigger the activation of STIM1, in addition to STIM2, which will fully generate the opening of CRAC channels, subsequently followed by a rapid and transient Ca2+ entry [83,104,105,106]. Ca2+ entry conducted by Orai1 will be severely inhibited after few milliseconds by Ca2+ itself [107,108] as well as after a longer period of time by the interaction of Orai1 N- and C-terminus with different proteins, such as SARAF [109,110,111] or by Orai1 serine phosphorylation at the N-terminus by kinases such as PKC or PKA [112,113]. Ca2+ influx through Orai1 leads to the recruitment of TRPC1 at the plasma membrane, which conducts further Ca2+ influx to reach the critical cytosolic Ca2+ concentration required for the physiological response evoked by the stimulus (Figure 2c) [103,114]. Next, the excess of intracellular Ca2+ is speedily removed, either by reintroducing the ion into the ER or by its extrusion to the extracellular medium via Ca2+-ATPases [70,71]. When agonist stimulation ceases, replenishment of the Ca2+ stores leads to the incorporation of Ca2+ to STIM1/2 EF-hand domains, which return these proteins to their quiescent conformation, leading to the deactivation of SOCE [79,104].
The number of studies linking SOCE proteins with cancer stem cell properties is growing at an amazingly fast pace; however, our knowledge is still extremely limited. Regarding STIM proteins, it is known that STIM1 associates with the hypoxia-inducible factor-1 alpha (HIF-1α) modulating each other, in a reciprocal dependency, in hypoxic hepatocarcinoma cells (HCCs). HIF-1α up-regulates STIM1 transcription, which in turn, induces higher SOCE, activating the CaMKII and P300 pathways, which are required for the accumulation of HIF-1α in HCCs [115].
Even less is known about the role of TRPC1 in CSC, since some of the inhibitors used to block SOCE, act over both Orai1 and TRPC1 channels. For instance, treatment with SKF96365, a SOCE inhibitor, impairs CSC proliferation in the glioblastoma stem-like cell line, TG1, triggering these cells to adopt a quiescent state by up-regulation of CDKN1A and G0S2 and the down-regulation of CCNB1 genes [116]. Similarly, SOCE impairment by SKF96365 in liver cancer stem cells (LCSCs) resulted in a drastic reduction in their ability to form spheroids, suppressing at the same time the expression of stemness-related genes. SOCE is activated in LCSC via the fibroblast growth factor 19 (FGF19), promoting the nuclear translocation of NFATc2 and self-renewal [117]. Even when the expression of Orai and STIM proteins was checked in both studies, TRPC1 was not considered and might be a possible candidate for future approaches.

4. Functional Role of Orai in Cancer Stem Cells and Cancer Hallmarks

As described above, native CRAC channels are hexameric structures comprised by the heteromeric association of Orai1, Orai2, and Orai3. Although all Orai family members can conform the channel, Orai2 and Orai3 also act as Ca2+ current modulators due to their lower Ca2+ conductivity and greater fast Ca2+-dependent inactivation as compared to Orai1 [108,118]. Several studies have demonstrated that the three Orai proteins are overexpressed in tumor samples and different human cancer cell lines compared with their non-tumorigenic counterpart cell lines. Hence, Orai1 is overexpressed in oral/oropharyngeal squamous cell carcinoma cells (OSCC) [119,120], liver [121], and breast cancer cells [122,123], Orai2 expression is increased in gastric [124], breast [125], oral [120], and acute myeloid leukemia cancer cells [126], while Orai3 expression is enhanced in the luminal breast cancer subtype [123,127], as well as in lung [128], pancreatic [129], and prostate cancer cells [130] (for a more extensive review see [131,132,133,134,135]). Using pharmacological or gene silencing approaches, to inhibit protein function or to avoid protein expression, respectively, the mentioned studies showed that Orai proteins play a crucial role in both tumorigenesis and the development and maintenance of different cancer hallmarks, including resistance to apoptosis, proliferation, migration, invasion, and metastasis via SOCE. However, as mentioned above, Orai1 can also mediate cancer progression by regulating and driving different Ca2+ influx pathways that are independent of the filling state of intracellular Ca2+ stores [93]. These pathways include: (1) the arachidonic acid-regulated Ca2+ current mediated by a Orai1/3 channel [130,136,137], (2) the constitutive Ca2+ influx mediated by the physical interaction between Orai1 and secretory pathway Ca2+-ATPase-2 [138,139,140], and (3) the Ca2+ influx mediated by the physical and functional interaction of Orai1 with the small conductance Ca2+-activated K+ channel 3 [141,142] or with the voltage-dependent Kv10.1 channel in the plasma membrane [143,144]. In the latter, a reciprocal positive feedback loop promotes the activation of both K+ channels by Orai1-mediated Ca2+ entry, which in turn leads to plasma membrane hyperpolarization, thus maintaining the driving force for Ca2+ influx and Ca2+ entry through Orai1 channels [141,145,146].
The role of Orai family proteins has also been described in the induction of CSC phenotype in a variety of cancers, such as glioblastoma, lung, and OSCC cancer cells. This CSC phenotype includes self-renewal capacity, tumor spheres formation, drug resistance, increased migration ability, and enhanced expression of stemness-related transcription factors and CSC-related markers [119,147,148]. Lee et al. demonstrated that Orai1, the predominant Orai family member in OSCC, is overexpressed in OSCC-derived CSC and its function is required for the maintenance of stemness and CSC phenotype through NFAT signaling pathway. Hence, Orai1 mediates the enhanced expression of stemness-related transcription factors, such as Nanog, Oct4 or Sox2, and promotes some CSC-related markers, including an increased ALDH1 activity and a higher CSC-related gene expression (Ezh2, Gli1, Hes1, Zeb2, FGF4, and IL4). The inhibition of Orai1 function in human tongue squamous carcinoma cell lines SCC4 and HOK-16B BapT by a pharmacological approach, using the Orai1 specific small molecular blocker compound 5D, impaired self-renewal capacity and reduced migration and invasion abilities in these cancer cells. Comparable results were also obtained by two different genetic approaches, using a specific siRNA to reduce Orai1 gene expression and inducing the overexpression of an Orai1 dominant negative mutant. Furthermore, Orai1 overexpression using viral vectors promoted CSC phenotype in non-tumorigenic immortalized oral epithelial cells HOK-16B [119]. Using related approaches, Singh et al. demonstrated that Orai1 and Orai2 overexpression is required for cell proliferation, migration, and colonization in SAS human tongue carcinoma cell line, processes that were found to be dependent on Akt/mTOR/NF-κB signaling pathway activation [120]. Analogous results were reported in glioblastoma stem cells derived from different human glioblastoma surgical samples. In these cells, the treatment with YM-58483, a CRAC current inhibitor, or with GSK-7975A, a more specific inhibitor of Orai1-mediated Ca2+ current, promoted a decrease in Sox2 expression, effect that was associated with reduced spheres formation and with the inhibition of their proliferation and self-renewal capacities [148]. Orai1 has been also related with chemoresistance, event that has been widely associated with CSC phenotype in cancer cells as previously mentioned. Hence, it has been demonstrated that ectopic overexpression of Orai1, using a plasmid vector, inhibited 5-fluorouracil-induced cell death in HepG2 hepatocarcinoma cells; meanwhile, Orai1 gene expression knockdown promoted the autophagic cell death induced by this pharmacological compound [121]. Similar findings were observed in cisplatin-resistant A2780 ovary carcinoma cells, in which Orai1 expression and SOCE are increased compared to therapy-sensitive parental cells. Pharmacological inhibition of Orai1 in cisplatin-resistant A2780 cells, using 2-aminoethoxydiphenyl borate (2-APB), promoted cisplatin-induced apoptotic cell death similarly to those observed in therapy-sensitive A2780 cells [149]. Conversely, an opposite effect has been reported in prostate cancer cells since the downregulation of Orai1 expression, caused by steroid-deprived conditions or by using specific siRNA against Orai1, and the impairment of Orai1 function by the overexpression of two Orai1 mutants, Orai1 R91W and Orai1 L273S, prevented the apoptotic cell death induced by different pharmacological compounds, including thapsigargin, TNFα, cisplatin, and oxaliplatin. Furthermore, the restoration of Orai1 expression in steroid-deprived cells by transfection with a Orai1 plasmid vector promoted the loss of chemoresistance in these cells [150].
Regarding the role of Orai3 in the CSC phenotype acquisition in cancer cells, it has been demonstrated that Orai3 overexpression is correlated with tumoral aggressiveness and chemoresistance acquisition in breast cancer cells [127,147]. Orai3 stable overexpressing T47D and MCF7 clones exhibited resistance to apoptotic cell death induced by thapsigargin, cisplatin, 5-fluorouracil, and paclitaxel compared with their parental cells transfected with the empty vector. This Orai3-dependent chemoresistance is acquired by ubiquitin ligase Nedd4-2-mediated p53 ubiquitination via the PI3K/Sgk-1 signaling pathway [127]. Previously, the same group demonstrated that Orai3 expression is also positively correlated with the oncogene c-myc expression in the ER-positive (luminal-like) breast cancer cell line MCF7 [151]. Daya et al. revealed that chemotherapy treatment increased Orai3 expression in primary human lung adenocarcinoma cells derived from bronchial biopsy specimens. Similar findings were reported in lung adenocarcinoma cell lines A549 and NCI-H23 after treatment with cisplatin. Interestingly, cisplatin treatment increased SOCE without affecting the expression of other proteins involved in CRAC current activation, such as STIM1, STIM2, and Orai1, even a slight decrease in the expression of Orai1 was observed in A549 cells. Orai3 gene expression knockdown using a specific siRNA enhanced cisplatin-induced apoptotic cell death in both lung adenocarcinoma cell lines, while Orai3 overexpression drastically reduced cisplatin-induced cell death and enhanced stemness in non-small cell lung cancer cells, as demonstrated by the enhanced expression of the stemness-related transcription factors Nanog and Sox2 via PI3K/AKT pathway, which resulted to be dependent on the increase in Orai3 expression [147].

5. Conclusions

Altogether, the presented data support an essential role of SOCE mediators-STIMs, Orais, and TRPC proteins in the induction of CSC phenotype. However, our current understanding about the role of these proteins in cancer stemness is incomplete since the existing studies do not take in consideration their participation in other pathways, such as the store independent function of STIM1, Orai1, and Orai3, while the possible implications of Orai2 or TRPC1 in the stemness properties of CSC remain unclear.

Author Contributions

Conceptualization, I.J. and J.A.R.; funding acquisition, J.A.R. and G.M.S.; writing—original draft preparation, J.A.R., I.J., J.J.L., J.S.-C. and L.J.G. writing—review and editing, I.J., J.A.R., G.M.S., J.J.L. and L.J.G. All authors have read and agreed to the published version of the manuscript.

Funding

Grants PID2019-104084GB-C21 funded by MCIN/AEI/ 10.13039/501100011033 and ERDF A way of making Europe, and Junta de Extremadura-Fondo Europeo de Desarrollo Regional (FEDER; Grants IB20007 and GR21008) to J.A.R., J.J.L. and I.J. are supported by contracts from Junta de Extremadura (TA18011 and TA18054, respectively). J.S.-C. is supported by a contract from Ministry of Science, Innovation, and Universities, Spain.

Institutional Review Board Statement

The study was conducted in accordance with the guidelines of the Declaration of Helsinki and approved by the Institutional Ethical Committee of the University of Extremadura (protocol code 195/2019 and date of approval 24 July 2019).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ramalho-Santos, M.; Willenbring, H. On the origin of the term “stem cell”. Cell Stem Cell 2007, 1, 35–38. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Soldner, F.; Jaenisch, R. Stem Cells, Genome Editing, and the Path to Translational Medicine. Cell 2018, 175, 615–632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Aponte, P.M.; Caicedo, A. Stemness in Cancer: Stem Cells, Cancer Stem Cells, and Their Microenvironment. Stem Cells Int. 2017, 2017, 5619472. [Google Scholar] [CrossRef] [PubMed]
  4. Neuzil, J.; Stantic, M.; Zobalova, R.; Chladova, J.; Wang, X.; Prochazka, L.; Dong, L.; Andera, L.; Ralph, S.J. Tumour-initiating cells vs. ‘cancer stem’ cells and CD133: What’s in the name? Biochem. Biophys. Res. Commun. 2007, 355, 855–859. [Google Scholar] [CrossRef] [PubMed]
  5. Kabakov, A.; Yakimova, A.; Matchuk, O. Molecular Chaperones in Cancer Stem Cells: Determinants of Stemness and Potential Targets for Antitumor Therapy. Cells 2020, 9, 892. [Google Scholar] [CrossRef] [Green Version]
  6. Li, L.; Neaves, W.B. Normal stem cells and cancer stem cells: The niche matters. Cancer Res. 2006, 66, 4553–4557. [Google Scholar] [CrossRef] [Green Version]
  7. Rahman, M.; Deleyrolle, L.; Vedam-Mai, V.; Azari, H.; Abd-El-Barr, M.; Reynolds, B.A. The cancer stem cell hypothesis: Failures and pitfalls. Neurosurgery 2011, 68, 531–545. [Google Scholar] [CrossRef] [Green Version]
  8. Bonnet, D.; Dick, J.E. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat. Med. 1997, 3, 730–737. [Google Scholar] [CrossRef]
  9. O’Brien, C.A.; Pollett, A.; Gallinger, S.; Dick, J.E. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature 2007, 445, 106–110. [Google Scholar] [CrossRef]
  10. Al-Hajj, M.; Wicha, M.S.; Benito-Hernandez, A.; Morrison, S.J.; Clarke, M.F. Prospective identification of tumorigenic breast cancer cells. Proc. Natl. Acad. Sci. USA 2003, 100, 3983–3988. [Google Scholar] [CrossRef] [Green Version]
  11. Singh, S.K.; Hawkins, C.; Clarke, I.D.; Squire, J.A.; Bayani, J.; Hide, T.; Henkelman, R.M.; Cusimano, M.D.; Dirks, P.B. Identification of human brain tumour initiating cells. Nature 2004, 432, 396–401. [Google Scholar] [CrossRef] [PubMed]
  12. Hollande, E.; de St-Front, V.T.; Louet-Hermitte, P.; Bara, J.; Pequignot, J.; Estival, A.; Clemente, F. Differentiation features of human pancreatic tumor cells maintained in nude mice and in culture: Immunocytochemical and ultrastructural studies. Int. J. Cancer 1984, 34, 177–185. [Google Scholar] [CrossRef] [PubMed]
  13. Sato, S.; Miyauchi, M.; Takekoshi, T.; Zhao, M.; Kudo, Y.; Ogawa, I.; Kitagawa, S.; Fujita, M.; Takata, T. Reduced expression of CD44 variant 9 is related to lymph node metastasis and poor survival in squamous cell carcinoma of tongue. Oral Oncol. 2000, 36, 545–549. [Google Scholar] [CrossRef]
  14. Grotenhuis, B.A.; Dinjens, W.N.; Wijnhoven, B.P.; Sonneveld, P.; Sacchetti, A.; Franken, P.F.; van Dekken, H.; Tilanus, H.W.; van Lanschot, J.J.; Fodde, R. Barrett’s oesophageal adenocarcinoma encompasses tumour-initiating cells that do not express common cancer stem cell markers. J. Pathol. 2010, 221, 379–389. [Google Scholar] [CrossRef]
  15. Yang, Z.F.; Ngai, P.; Ho, D.W.; Yu, W.C.; Ng, M.N.; Lau, C.K.; Li, M.L.; Tam, K.H.; Lam, C.T.; Poon, R.T.; et al. Identification of local and circulating cancer stem cells in human liver cancer. Hepatology 2008, 47, 919–928. [Google Scholar] [CrossRef]
  16. Schatton, T.; Murphy, G.F.; Frank, N.Y.; Yamaura, K.; Waaga-Gasser, A.M.; Gasser, M.; Zhan, Q.; Jordan, S.; Duncan, L.M.; Weishaupt, C.; et al. Identification of cells initiating human melanomas. Nature 2008, 451, 345–349. [Google Scholar] [CrossRef] [Green Version]
  17. Reya, T.; Morrison, S.J.; Clarke, M.F.; Weissman, I.L. Stem cells, cancer, and cancer stem cells. Nature 2001, 414, 105–111. [Google Scholar] [CrossRef] [Green Version]
  18. Wicha, M.S.; Liu, S.; Dontu, G. Cancer stem cells: An old idea—A paradigm shift. Cancer Res. 2006, 66, 1883–1890. [Google Scholar] [CrossRef] [Green Version]
  19. Dalerba, P.; Cho, R.W.; Clarke, M.F. Cancer stem cells: Models and concepts. Annu. Rev. Med. 2007, 58, 267–284. [Google Scholar] [CrossRef] [Green Version]
  20. Walcher, L.; Kistenmacher, A.K.; Suo, H.; Kitte, R.; Dluczek, S.; Strauss, A.; Blaudszun, A.R.; Yevsa, T.; Fricke, S.; Kossatz-Boehlert, U. Cancer Stem Cells-Origins and Biomarkers: Perspectives for Targeted Personalized Therapies. Front. Immunol. 2020, 11, 1280. [Google Scholar] [CrossRef]
  21. Cho, Y.; Lee, H.W.; Kang, H.G.; Kim, H.Y.; Kim, S.J.; Chun, K.H. Cleaved CD44 intracellular domain supports activation of stemness factors and promotes tumorigenesis of breast cancer. Oncotarget 2015, 6, 8709–8721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Screaton, G.R.; Bell, M.V.; Jackson, D.G.; Cornelis, F.B.; Gerth, U.; Bell, J.I. Genomic structure of DNA encoding the lymphocyte homing receptor CD44 reveals at least 12 alternatively spliced exons. Proc. Natl. Acad. Sci. USA 1992, 89, 12160–12164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Gunthert, U.; Hofmann, M.; Rudy, W.; Reber, S.; Zoller, M.; Haussmann, I.; Matzku, S.; Wenzel, A.; Ponta, H.; Herrlich, P. A new variant of glycoprotein CD44 confers metastatic potential to rat carcinoma cells. Cell 1991, 65, 13–24. [Google Scholar] [CrossRef]
  24. Pierres, M.; Naquet, P.; Barbet, J.; Marchetto, S.; Marics, I.; Devaux, C.; Barad, M.; Hyman, R.; Rougon, G. Evidence that murine hematopoietic cell subset marker J11d is attached to a glycosyl-phosphatidylinositol membrane anchor. Eur. J. Immunol. 1987, 17, 1781–1785. [Google Scholar] [CrossRef] [PubMed]
  25. Ricardo, S.; Vieira, A.F.; Gerhard, R.; Leitao, D.; Pinto, R.; Cameselle-Teijeiro, J.F.; Milanezi, F.; Schmitt, F.; Paredes, J. Breast cancer stem cell markers CD44, CD24 and ALDH1: Expression distribution within intrinsic molecular subtype. J. Clin. Pathol. 2011, 64, 937–946. [Google Scholar] [CrossRef]
  26. Li, W.; Ma, H.; Zhang, J.; Zhu, L.; Wang, C.; Yang, Y. Unraveling the roles of CD44/CD24 and ALDH1 as cancer stem cell markers in tumorigenesis and metastasis. Sci. Rep. 2017, 7, 13856. [Google Scholar] [CrossRef] [Green Version]
  27. Ginestier, C.; Hur, M.H.; Charafe-Jauffret, E.; Monville, F.; Dutcher, J.; Brown, M.; Jacquemier, J.; Viens, P.; Kleer, C.G.; Liu, S.; et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell 2007, 1, 555–567. [Google Scholar] [CrossRef] [Green Version]
  28. Marcato, P.; Dean, C.A.; Pan, D.; Araslanova, R.; Gillis, M.; Joshi, M.; Helyer, L.; Pan, L.; Leidal, A.; Gujar, S.; et al. Aldehyde dehydrogenase activity of breast cancer stem cells is primarily due to isoform ALDH1A3 and its expression is predictive of metastasis. Stem Cells 2011, 29, 32–45. [Google Scholar] [CrossRef]
  29. Ohi, Y.; Umekita, Y.; Yoshioka, T.; Souda, M.; Rai, Y.; Sagara, Y.; Tanimoto, A. Aldehyde dehydrogenase 1 expression predicts poor prognosis in triple-negative breast cancer. Histopathology 2011, 59, 776–780. [Google Scholar] [CrossRef]
  30. Zhou, L.; Li, K.; Luo, Y.; Tian, L.; Wang, M.; Li, C.; Huang, Q. Novel prognostic markers for patients with triple-negative breast cancer. Hum. Pathol. 2013, 44, 2180–2187. [Google Scholar] [CrossRef]
  31. Wilson, J.J.; Kovall, R.A. Crystal structure of the CSL-Notch-Mastermind ternary complex bound to DNA. Cell 2006, 124, 985–996. [Google Scholar] [CrossRef] [Green Version]
  32. Wang, R.; Sun, Q.; Wang, P.; Liu, M.; Xiong, S.; Luo, J.; Huang, H.; Du, Q.; Geller, D.A.; Cheng, B. Notch and Wnt/beta-catenin signaling pathway play important roles in activating liver cancer stem cells. Oncotarget 2016, 7, 5754–5768. [Google Scholar] [CrossRef] [Green Version]
  33. Cochrane, C.R.; Szczepny, A.; Watkins, D.N.; Cain, J.E. Hedgehog Signaling in the Maintenance of Cancer Stem Cells. Cancers 2015, 7, 1554–1585. [Google Scholar] [CrossRef] [PubMed]
  34. Liu, X.; Huang, Y.; Zhang, Y.; Li, X.; Liu, C.; Huang, S.; Xu, D.; Wu, Y. T-cell factor (TCF/LEF1) binding elements (TBEs) of FasL (Fas ligand or CD95 ligand) bind and cluster Fas (CD95) and form complexes with the TCF-4 and b-catenin transcription factors in vitro and in vivo which result in triggering cell death and/or cell activation. Cell. Mol. Neurobiol. 2016, 36, 1001–1013. [Google Scholar] [CrossRef] [PubMed]
  35. Hooper, R.; Zaidi, M.R.; Soboloff, J. The heterogeneity of store-operated calcium entry in melanoma. Sci. China Life Sci. 2016, 59, 764–769. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. van Amerongen, R. Alternative Wnt pathways and receptors. Cold Spring Harb. Perspect. Biol. 2012, 4, a007914. [Google Scholar] [CrossRef] [Green Version]
  37. Ma, B.; Hottiger, M.O. Crosstalk between Wnt/beta-Catenin and NF-kappaB Signaling Pathway during Inflammation. Front. Immunol. 2016, 7, 378. [Google Scholar] [CrossRef]
  38. Berridge, M.J.; Bootman, M.D.; Roderick, H.L. Calcium signalling: Dynamics, homeostasis and remodelling. Nat. Rev. Mol. Cell Biol. 2003, 4, 517–529. [Google Scholar] [CrossRef] [Green Version]
  39. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  40. Monteith, G.R.; McAndrew, D.; Faddy, H.M.; Roberts-Thomson, S.J. Calcium and cancer: Targeting Ca2+ transport. Nat. Rev. Cancer 2007, 7, 519–530. [Google Scholar] [CrossRef]
  41. Hodeify, R.; Yu, F.; Courjaret, R.; Nader, N.; Dib, M.; Sun, L.; Adap, E.; Hubrack, S.; Machaca, K. Regulation and Role of Store-Operated Ca2+ Entry in Cellular Proliferation. Calcium Entry Channels Non-Excit. Cells 2018, 2017, 215–240. [Google Scholar] [CrossRef]
  42. Qin, J.J.; Nag, S.; Wang, W.; Zhou, J.; Zhang, W.D.; Wang, H.; Zhang, R. NFAT as cancer target: Mission possible? Biochim. Biophys. Acta 2014, 1846, 297–311. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Burotto, M.; Chiou, V.L.; Lee, J.M.; Kohn, E.C. The MAPK pathway across different malignancies: A new perspective. Cancer 2014, 120, 3446–3456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. De Luca, A.; Maiello, M.R.; D’Alessio, A.; Pergameno, M.; Normanno, N. The RAS/RAF/MEK/ERK and the PI3K/AKT signalling pathways: Role in cancer pathogenesis and implications for therapeutic approaches. Expert Opin. Ther. Targets 2012, 16, S17–S27. [Google Scholar] [CrossRef]
  45. Lin, R.J.; Kuo, M.W.; Yang, B.C.; Tsai, H.H.; Chen, K.; Huang, J.R.; Lee, Y.S.; Yu, A.L.; Yu, J. B3GALT5 knockout alters gycosphingolipid profile and facilitates transition to human naive pluripotency. Proc. Natl. Acad. Sci. USA 2020, 117, 27435–27444. [Google Scholar] [CrossRef]
  46. Zubeldia, I.G.; Bleau, A.M.; Redrado, M.; Serrano, D.; Agliano, A.; Gil-Puig, C.; Vidal-Vanaclocha, F.; Lecanda, J.; Calvo, A. Epithelial to mesenchymal transition and cancer stem cell phenotypes leading to liver metastasis are abrogated by the novel TGFbeta1-targeting peptides P17 and P144. Exp. Cell Res. 2013, 319, 12–22. [Google Scholar] [CrossRef]
  47. Cocola, C.; Molgora, S.; Piscitelli, E.; Veronesi, M.C.; Greco, M.; Bragato, C.; Moro, M.; Crosti, M.; Gray, B.; Milanesi, L.; et al. FGF2 and EGF Are Required for Self-Renewal and Organoid Formation of Canine Normal and Tumor Breast Stem Cells. J. Cell Biochem. 2017, 118, 570–584. [Google Scholar] [CrossRef]
  48. Dong, J.; Aulestia, F.J.; Assad Kahn, S.; Zeniou, M.; Dubois, L.G.; El-Habr, E.A.; Daubeuf, F.; Tounsi, N.; Cheshier, S.H.; Frossard, N.; et al. Bisacodyl and its cytotoxic activity on human glioblastoma stem-like cells. Implication of inositol 1,4,5-triphosphate receptor dependent calcium signaling. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 1018–1027. [Google Scholar] [CrossRef]
  49. Marciel, M.P.; Khadka, V.S.; Deng, Y.; Kilicaslan, P.; Pham, A.; Bertino, P.; Lee, K.; Chen, S.; Glibetic, N.; Hoffmann, F.W.; et al. Selenoprotein K deficiency inhibits melanoma by reducing calcium flux required for tumor growth and metastasis. Oncotarget 2018, 9, 13407–13422. [Google Scholar] [CrossRef] [Green Version]
  50. Lu, H.; Chen, I.; Shimoda, L.A.; Park, Y.; Zhang, C.; Tran, L.; Zhang, H.; Semenza, G.L. Chemotherapy-Induced Ca2+ Release Stimulates Breast Cancer Stem Cell Enrichment. Cell Rep. 2017, 18, 1946–1957. [Google Scholar] [CrossRef]
  51. Prevarskaya, N.; Skryma, R.; Shuba, Y. Ion Channels in Cancer: Are Cancer Hallmarks Oncochannelopathies? Physiol. Rev. 2018, 98, 559–621. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Zhang, Y.; Cruickshanks, N.; Yuan, F.; Wang, B.; Pahuski, M.; Wulfkuhle, J.; Gallagher, I.; Koeppel, A.F.; Hatef, S.; Papanicolas, C.; et al. Targetable T-type Calcium Channels Drive Glioblastoma. Cancer Res. 2017, 77, 3479–3490. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Lee, H.; Kim, J.W.; Kim, D.K.; Choi, D.K.; Lee, S.; Yu, J.H.; Kwon, O.B.; Lee, J.; Lee, D.S.; Kim, J.H.; et al. Calcium Channels as Novel Therapeutic Targets for Ovarian Cancer Stem Cells. Int. J. Mol. Sci. 2020, 21, 2327. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Niklasson, M.; Maddalo, G.; Sramkova, Z.; Mutlu, E.; Wee, S.; Sekyrova, P.; Schmidt, L.; Fritz, N.; Dehnisch, I.; Kyriatzis, G.; et al. Membrane-Depolarizing Channel Blockers Induce Selective Glioma Cell Death by Impairing Nutrient Transport and Unfolded Protein/Amino Acid Responses. Cancer Res. 2017, 77, 1741–1752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Yu, J.; Wang, S.; Zhao, W.; Duan, J.; Wang, Z.; Chen, H.; Tian, Y.; Wang, D.; Zhao, J.; An, T.; et al. Mechanistic Exploration of Cancer Stem Cell Marker Voltage-Dependent Calcium Channel alpha2delta1 Subunit-mediated Chemotherapy Resistance in Small-Cell Lung Cancer. Clin. Cancer Res. 2018, 24, 2148–2158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Ma, Y.; Yang, X.; Zhao, W.; Yang, Y.; Zhang, Z. Calcium channel alpha2delta1 subunit is a functional marker and therapeutic target for tumor-initiating cells in non-small cell lung cancer. Cell Death Dis. 2021, 12, 257. [Google Scholar] [CrossRef] [PubMed]
  57. Li, M.; Zhang, W.; Yang, X.; An, G.; Zhao, W. The alpha2delta1 subunit of the voltage-gated calcium channel acts as a potential candidate for breast cancer tumor initial cells biomarker. Cancer Biomark. 2021, 31, 295–305. [Google Scholar] [CrossRef]
  58. Zhao, W.; Wang, L.; Han, H.; Jin, K.; Lin, N.; Guo, T.; Chen, Y.; Cheng, H.; Lu, F.; Fang, W.; et al. 1B50-1, a mAb raised against recurrent tumor cells, targets liver tumor-initiating cells by binding to the calcium channel alpha2delta1 subunit. Cancer Cell 2013, 23, 541–556. [Google Scholar] [CrossRef] [Green Version]
  59. Huang, C.; Li, Y.; Zhao, W.; Zhang, A.; Lu, C.; Wang, Z.; Liu, L. alpha2delta1 may be a potential marker for cancer stem cell in laryngeal squamous cell carcinoma. Cancer Biomark. 2019, 24, 97–107. [Google Scholar] [CrossRef] [Green Version]
  60. Hirata, N.; Yamada, S.; Yanagida, S.; Ono, A.; Yasuhiko, Y.; Nishida, M.; Kanda, Y. Lysophosphatidic Acid Promotes the Expansion of Cancer Stem Cells via TRPC3 Channels in Triple-Negative Breast Cancer. Int. J. Mol. Sci. 2022, 23, 1967. [Google Scholar] [CrossRef]
  61. Liu, K.; Xu, S.H.; Chen, Z.; Zeng, Q.X.; Li, Z.J.; Chen, Z.M. TRPM7 overexpression enhances the cancer stem cell-like and metastatic phenotypes of lung cancer through modulation of the Hsp90alpha/uPA/MMP2 signaling pathway. BMC Cancer 2018, 18, 1167. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, M.; Inoue, K.; Leng, T.; Guo, S.; Xiong, Z.G. TRPM7 channels regulate glioma stem cell through STAT3 and Notch signaling pathways. Cell. Signal. 2014, 26, 2773–2781. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Middelbeek, J.; Visser, D.; Henneman, L.; Kamermans, A.; Kuipers, A.J.; Hoogerbrugge, P.M.; Jalink, K.; van Leeuwen, F.N. TRPM7 maintains progenitor-like features of neuroblastoma cells: Implications for metastasis formation. Oncotarget 2015, 6, 8760–8776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Santoni, G.; Amantini, C. The Transient Receptor Potential Vanilloid Type-2(TRPV2) Ion Channels in Neurogenesis andGliomagenesis: Cross-Talk between TranscriptionFactors and Signaling Molecules. Cancers 2019, 11, 322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Shiozaki, A.; Kudou, M.; Ichikawa, D.; Fujiwara, H.; Shimizu, H.; Ishimoto, T.; Arita, T.; Kosuga, T.; Konishi, H.; Komatsu, S.; et al. Esophageal cancer stem cells are suppressed by tranilast, a TRPV2 channel inhibitor. J. Gastroenterol. 2018, 53, 197–207. [Google Scholar] [CrossRef] [PubMed]
  66. Morelli, M.B.; Nabissi, M.; Amantini, C.; Farfariello, V.; Ricci-Vitiani, L.; di Martino, S.; Pallini, R.; Larocca, L.M.; Caprodossi, S.; Santoni, M.; et al. The transient receptor potential vanilloid-2 cation channel impairs glioblastoma stem-like cell proliferation and promotes differentiation. Int. J. Cancer 2012, 131, E1067–E1077. [Google Scholar] [CrossRef] [Green Version]
  67. Nabissi, M.; Morelli, M.B.; Amantini, C.; Liberati, S.; Santoni, M.; Ricci-Vitiani, L.; Pallini, R.; Santoni, G. Cannabidiol stimulates Aml-1a-dependent glial differentiation and inhibits glioma stem-like cells proliferation by inducing autophagy in a TRPV2-dependent manner. Int. J. Cancer 2015, 137, 1855–1869. [Google Scholar] [CrossRef] [Green Version]
  68. Hu, Z.; Cao, X.; Fang, Y.; Liu, G.; Xie, C.; Qian, K.; Lei, X.; Cao, Z.; Du, H.; Cheng, X.; et al. Transient receptor potential vanilloid-type 2 targeting on stemness in liver cancer. Biomed. Pharmacother. 2018, 105, 697–706. [Google Scholar] [CrossRef]
  69. Santoni, G.; Nabissi, M.; Amantini, C.; Santoni, M.; Ricci-Vitiani, L.; Pallini, R.; Maggi, F.; Morelli, M.B. ERK Phosphorylation Regulates the Aml1/Runx1 Splice Variants and the TRP Channels Expression during the Differentiation of Glioma Stem Cell Lines. Cells 2021, 10, 2052. [Google Scholar] [CrossRef]
  70. Redondo, P.C.; Rosado, J.A.; Pariente, J.A.; Salido, G.M. Collaborative effect of SERCA and PMCA in cytosolic calcium homeostasis in human platelets. J. Physiol. Biochem. 2005, 61, 507–516. [Google Scholar] [CrossRef]
  71. Elaib, Z.; Saller, F.; Bobe, R. The Calcium Entry-Calcium Refilling Coupling. Adv. Exp. Med. Biol. 2016, 898, 333–352. [Google Scholar] [CrossRef] [PubMed]
  72. Lariccia, V.; Piccirillo, S.; Preziuso, A.; Amoroso, S.; Magi, S. Cracking the code of sodium/calcium exchanger (NCX) gating: Old and new complexities surfacing from the deep web of secondary regulations. Cell Calcium 2020, 87, 102169. [Google Scholar] [CrossRef] [PubMed]
  73. Park, K.C.; Kim, S.W.; Jeon, J.Y.; Jo, A.R.; Choi, H.J.; Kim, J.; Lee, H.G.; Kim, Y.; Mills, G.B.; Noh, S.H.; et al. Survival of Cancer Stem-Like Cells Under Metabolic Stress via CaMK2alpha-mediated Upregulation of Sarco/Endoplasmic Reticulum Calcium ATPase Expression. Clin. Cancer Res. 2018, 24, 1677–1690. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Robil, N.; Petel, F.; Kilhoffer, M.C.; Haiech, J. Glioblastoma and calcium signaling—Analysis of calcium toolbox expression. Int. J. Dev. Biol. 2015, 59, 407–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Putney, J.W.; Bird, G.S. Cytoplasmic calcium oscillations and store-operated calcium influx. J. Physiol. 2008, 586, 3055–3059. [Google Scholar] [CrossRef]
  76. Putney, J.W., Jr. A model for receptor-regulated calcium entry. Cell Calcium 1986, 7, 1–12. [Google Scholar] [CrossRef]
  77. Jardin, I.; Rosado, J.A. STIM and calcium channel complexes in cancer. Biochim. Biophys. Acta 2016, 1863, 1418–1426. [Google Scholar] [CrossRef]
  78. Roos, J.; DiGregorio, P.J.; Yeromin, A.V.; Ohlsen, K.; Lioudyno, M.; Zhang, S.; Safrina, O.; Kozak, J.A.; Wagner, S.L.; Cahalan, M.D.; et al. STIM1, an essential and conserved component of store-operated Ca2+ channel function. J. Cell Biol. 2005, 169, 435–445. [Google Scholar] [CrossRef] [Green Version]
  79. Zhang, S.L.; Yu, Y.; Roos, J.; Kozak, J.A.; Deerinck, T.J.; Ellisman, M.H.; Stauderman, K.A.; Cahalan, M.D. STIM1 is a Ca2+ sensor that activates CRAC channels and migrates from the Ca2+ store to the plasma membrane. Nature 2005, 437, 902–905. [Google Scholar] [CrossRef]
  80. Berna-Erro, A.; Jardin, I.; Salido, G.M.; Rosado, J.A. Role of STIM2 in cell function and physiopathology. J. Physiol. 2017, 595, 3111–3128. [Google Scholar] [CrossRef] [Green Version]
  81. Grabmayr, H.; Romanin, C.; Fahrner, M. STIM Proteins: An Ever-Expanding Family. Int. J. Mol. Sci. 2020, 22, 378. [Google Scholar] [CrossRef] [PubMed]
  82. Fahrner, M.; Romanin, C. The many states of STIM1. Elife 2021, 10, e75174. [Google Scholar] [CrossRef] [PubMed]
  83. Yuan, J.P.; Zeng, W.; Dorwart, M.R.; Choi, Y.J.; Worley, P.F.; Muallem, S. SOAR and the polybasic STIM1 domains gate and regulate Orai channels. Nat. Cell Biol. 2009, 11, 337–343. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, Z.Y.; Pan, L.J.; Zhang, Z.M. Functional interactions among STIM1, Orai1 and TRPC1 on the activation of SOCs in HL-7702 cells. Amino Acids 2009, 39, 195–204. [Google Scholar] [CrossRef]
  85. Desai, P.N.; Zhang, X.; Wu, S.; Janoshazi, A.; Bolimuntha, S.; Putney, J.W.; Trebak, M. Multiple types of calcium channels arising from alternative translation initiation of the Orai1 message. Sci. Signal. 2015, 8, ra74. [Google Scholar] [CrossRef] [Green Version]
  86. Feske, S.; Gwack, Y.; Prakriya, M.; Srikanth, S.; Puppel, S.H.; Tanasa, B.; Hogan, P.G.; Lewis, R.S.; Daly, M.; Rao, A. A mutation in Orai1 causes immune deficiency by abrogating CRAC channel function. Nature 2006, 441, 179–185. [Google Scholar] [CrossRef]
  87. Zhou, Y.; Ramachandran, S.; Oh-Hora, M.; Rao, A.; Hogan, P.G. Pore architecture of the ORAI1 store-operated calcium channel. Proc. Natl. Acad. Sci. USA 2010, 107, 4896–4901. [Google Scholar] [CrossRef] [Green Version]
  88. Mignen, O.; Thompson, J.L.; Shuttleworth, T.J. Orai1 subunit stoichiometry of the mammalian CRAC channel pore. J. Physiol. 2008, 586, 419–425. [Google Scholar] [CrossRef]
  89. Hou, X.; Pedi, L.; Diver, M.M.; Long, S.B. Crystal structure of the calcium release-activated calcium channel Orai. Science 2012, 338, 1308–1313. [Google Scholar] [CrossRef] [Green Version]
  90. Butorac, C.; Krizova, A.; Derler, I. Review: Structure and Activation Mechanisms of CRAC Channels. Adv. Exp. Med. Biol. 2020, 1131, 547–604. [Google Scholar] [CrossRef]
  91. Qiu, R.; Lewis, R.S. Structural features of STIM and Orai underlying store-operated calcium entry. Curr. Opin. Cell Biol. 2019, 57, 90–98. [Google Scholar] [CrossRef] [PubMed]
  92. Shuttleworth, T.J. Arachidonic acid, ARC channels, and Orai proteins. Cell Calcium 2009, 45, 602–610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Cantonero, C.; Sanchez-Collado, J.; Gonzalez-Nunez, M.A.; Salido, G.M.; Lopez, J.J.; Jardin, I.; Rosado, J.A. Store-independent Orai1-mediated Ca2+ entry and cancer. Cell Calcium 2019, 80, 1–7. [Google Scholar] [CrossRef] [PubMed]
  94. Clapham, D.E. TRP channels as cellular sensors. Nature 2003, 426, 517–524. [Google Scholar] [CrossRef] [PubMed]
  95. Guo, W.; Chen, L. Recent progress in structural studies on canonical TRP ion channels. Cell Calcium 2019, 83, 102075. [Google Scholar] [CrossRef]
  96. Lopez, J.J.; Jardin, I.; Albarran, L.; Sanchez-Collado, J.; Cantonero, C.; Salido, G.M.; Smani, T.; Rosado, J.A. Molecular Basis and Regulation of Store-Operated Calcium Entry. Adv. Exp. Med. Biol. 2020, 1131, 445–469. [Google Scholar] [CrossRef]
  97. Huang, G.N.; Zeng, W.; Kim, J.Y.; Yuan, J.P.; Han, L.; Muallem, S.; Worley, P.F. STIM1 carboxyl-terminus activates native SOC, I(crac) and TRPC1 channels. Nat. Cell Biol. 2006, 8, 1003–1010. [Google Scholar] [CrossRef]
  98. Yuan, J.P.; Zeng, W.; Huang, G.N.; Worley, P.F.; Muallem, S. STIM1 heteromultimerizes TRPC channels to determine their function as store-operated channels. Nat. Cell Biol. 2007, 9, 636–645. [Google Scholar] [CrossRef]
  99. Cheng, K.T.; Liu, X.; Ong, H.L.; Ambudkar, I.S. Functional requirement for Orai1 in store-operated TRPC1-STIM1 channels. J. Biol. Chem. 2008, 283, 12935–12940. [Google Scholar] [CrossRef] [Green Version]
  100. Lee, K.P.; Choi, S.; Hong, J.H.; Ahuja, M.; Graham, S.; Ma, R.; So, I.; Shin, D.M.; Muallem, S.; Yuan, J.P. Molecular determinants mediating gating of Transient Receptor Potential Canonical (TRPC) channels by stromal interaction molecule 1 (STIM1). J. Biol. Chem. 2014, 289, 6372–6382. [Google Scholar] [CrossRef] [Green Version]
  101. Jardin, I.; Lopez, J.J.; Salido, G.M.; Rosado, J.A. Orai1 mediates the interaction between STIM1 and hTRPC1 and regulates the mode of activation of hTRPC1-forming Ca2+ channels. J. Biol. Chem. 2008, 283, 25296–25304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Zitt, C.; Zobel, A.; Obukhov, A.G.; Harteneck, C.; Kalkbrenner, F.; Luckhoff, A.; Schultz, G. Cloning and functional expression of a human Ca2+-permeable cation channel activated by calcium store depletion. Neuron 1996, 16, 1189–1196. [Google Scholar] [CrossRef] [Green Version]
  103. Cheng, K.T.; Liu, X.; Ong, H.L.; Swaim, W.; Ambudkar, I.S. Local Ca2+ entry via Orai1 regulates plasma membrane recruitment of TRPC1 and controls cytosolic Ca2+ signals required for specific cell functions. PLoS Biol. 2011, 9, e1001025. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Stathopulos, P.B.; Zheng, L.; Li, G.Y.; Plevin, M.J.; Ikura, M. Structural and mechanistic insights into STIM1-mediated initiation of store-operated calcium entry. Cell 2008, 135, 110–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Muik, M.; Frischauf, I.; Derler, I.; Fahrner, M.; Bergsmann, J.; Eder, P.; Schindl, R.; Hesch, C.; Polzinger, B.; Fritsch, R.; et al. Dynamic coupling of the putative coiled-coil domain of ORAI1 with STIM1 mediates ORAI1 channel activation. J. Biol. Chem. 2008, 283, 8014–8022. [Google Scholar] [CrossRef] [Green Version]
  106. Park, C.Y.; Hoover, P.J.; Mullins, F.M.; Bachhawat, P.; Covington, E.D.; Raunser, S.; Walz, T.; Garcia, K.C.; Dolmetsch, R.E.; Lewis, R.S. STIM1 clusters and activates CRAC channels via direct binding of a cytosolic domain to Orai1. Cell 2009, 136, 876–890. [Google Scholar] [CrossRef] [Green Version]
  107. Derler, I.; Fahrner, M.; Muik, M.; Lackner, B.; Schindl, R.; Groschner, K.; Romanin, C. A CRAC modulatory domain (CMD) within STIM1 mediates fast Ca2+-dependent inactivation of ORAI1 channels. J. Biol. Chem. 2009, 284, 24933–24938. [Google Scholar] [CrossRef] [Green Version]
  108. Lee, K.P.; Yuan, J.P.; Zeng, W.; So, I.; Worley, P.F.; Muallem, S. Molecular determinants of fast Ca2+-dependent inactivation and gating of the Orai channels. Proc. Natl. Acad. Sci. USA 2009, 106, 14687–14692. [Google Scholar] [CrossRef] [Green Version]
  109. Albarran, L.; Lopez, J.J.; Jardin, I.; Sanchez-Collado, J.; Berna-Erro, A.; Smani, T.; Camello, P.J.; Salido, G.M.; Rosado, J.A. EFHB is a Novel Cytosolic Ca2+ Sensor That Modulates STIM1-SARAF Interaction. Cell Physiol. Biochem. 2018, 51, 1164–1178. [Google Scholar] [CrossRef]
  110. Jha, A.; Ahuja, M.; Maleth, J.; Moreno, C.M.; Yuan, J.P.; Kim, M.S.; Muallem, S. The STIM1 CTID domain determines access of SARAF to SOAR to regulate Orai1 channel function. J. Cell Biol. 2013, 202, 71–79. [Google Scholar] [CrossRef] [Green Version]
  111. Palty, R.; Raveh, A.; Kaminsky, I.; Meller, R.; Reuveny, E. SARAF inactivates the store operated calcium entry machinery to prevent excess calcium refilling. Cell 2012, 149, 425–438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Kawasaki, T.; Ueyama, T.; Lange, I.; Feske, S.; Saito, N. Protein kinase C-induced phosphorylation of Orai1 regulates the intracellular Ca2+ level via the store-operated Ca2+ channel. J. Biol. Chem. 2010, 285, 25720–25730. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Zhang, X.; Pathak, T.; Yoast, R.; Emrich, S.; Xin, P.; Nwokonko, R.M.; Johnson, M.; Wu, S.; Delierneux, C.; Gueguinou, M.; et al. A calcium/cAMP signaling loop at the ORAI1 mouth drives channel inactivation to shape NFAT induction. Nat. Commun. 2019, 10, 1971. [Google Scholar] [CrossRef] [PubMed]
  114. Sanchez-Collado, J.; Lopez, J.J.; Jardin, I.; Berna-Erro, A.; Camello, P.J.; Cantonero, C.; Smani, T.; Salido, G.M.; Rosado, J.A. Orai1alpha, but not Orai1beta, co-localizes with TRPC1 and is required for its plasma membrane location and activation in HeLa cells. Cell Mol. Life Sci. 2022, 79, 33. [Google Scholar] [CrossRef]
  115. Li, Y.; Guo, B.; Xie, Q.; Ye, D.; Zhang, D.; Zhu, Y.; Chen, H.; Zhu, B. STIM1 Mediates Hypoxia-Driven Hepatocarcinogenesis via Interaction with HIF-1. Cell Rep. 2015, 12, 388–395. [Google Scholar] [CrossRef] [Green Version]
  116. Aulestia, F.J.; Neant, I.; Dong, J.; Haiech, J.; Kilhoffer, M.C.; Moreau, M.; Leclerc, C. Quiescence status of glioblastoma stem-like cells involves remodelling of Ca2+ signalling and mitochondrial shape. Sci. Rep. 2018, 8, 9731. [Google Scholar] [CrossRef]
  117. Wang, J.; Zhao, H.; Zheng, L.; Zhou, Y.; Wu, L.; Xu, Y.; Zhang, X.; Yan, G.; Sheng, H.; Xin, R.; et al. FGF19/SOCE/NFATc2 signaling circuit facilitates the self-renewal of liver cancer stem cells. Theranostics 2021, 11, 5045–5060. [Google Scholar] [CrossRef]
  118. Yoast, R.E.; Emrich, S.M.; Zhang, X.; Xin, P.; Johnson, M.T.; Fike, A.J.; Walter, V.; Hempel, N.; Yule, D.I.; Sneyd, J.; et al. The native ORAI channel trio underlies the diversity of Ca2+ signaling events. Nat. Commun. 2020, 11, 2444. [Google Scholar] [CrossRef]
  119. Lee, S.H.; Rigas, N.K.; Lee, C.R.; Bang, A.; Srikanth, S.; Gwack, Y.; Kang, M.K.; Kim, R.H.; Park, N.H.; Shin, K.H. Orai1 promotes tumor progression by enhancing cancer stemness via NFAT signaling in oral/oropharyngeal squamous cell carcinoma. Oncotarget 2016, 7, 43239–43255. [Google Scholar] [CrossRef] [Green Version]
  120. Singh, A.K.; Roy, N.K.; Bordoloi, D.; Padmavathi, G.; Banik, K.; Khwairakpam, A.D.; Kunnumakkara, A.B.; Sukumar, P. Orai-1 and Orai-2 regulate oral cancer cell migration and colonisation by suppressing Akt/mTOR/NF-kappaB signalling. Life Sci. 2020, 261, 118372. [Google Scholar] [CrossRef]
  121. Tang, B.D.; Xia, X.; Lv, X.F.; Yu, B.X.; Yuan, J.N.; Mai, X.Y.; Shang, J.Y.; Zhou, J.G.; Liang, S.J.; Pang, R.P. Inhibition of Orai1-mediated Ca2+ entry enhances chemosensitivity of HepG2 hepatocarcinoma cells to 5-fluorouracil. J. Cell. Mol. Med. 2017, 21, 904–915. [Google Scholar] [CrossRef] [PubMed]
  122. Jardin, I.; Diez-Bello, R.; Lopez, J.J.; Redondo, P.C.; Salido, G.M.; Smani, T.; Rosado, J.A. TRPC6 Channels Are Required for Proliferation, Migration and Invasion of Breast Cancer Cell Lines by Modulation of Orai1 and Orai3 Surface Exposure. Cancers 2018, 10, 331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Motiani, R.K.; Abdullaev, I.F.; Trebak, M. A novel native store-operated calcium channel encoded by Orai3: Selective requirement of Orai3 versus Orai1 in estrogen receptor-positive versus estrogen receptor-negative breast cancer cells. J. Biol. Chem. 2010, 285, 19173–19183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Wu, S.; Chen, M.; Huang, J.; Zhang, F.; Lv, Z.; Jia, Y.; Cui, Y.Z.; Sun, L.Z.; Wang, Y.; Tang, Y.; et al. ORAI2 Promotes Gastric Cancer Tumorigenicity and Metastasis through PI3K/Akt Signaling and MAPK-Dependent Focal Adhesion Disassembly. Cancer Res. 2021, 81, 986–1000. [Google Scholar] [CrossRef] [PubMed]
  125. Sanchez-Collado, J.; Lopez, J.J.; Cantonero, C.; Jardin, I.; Regodon, S.; Redondo, P.C.; Gordillo, J.; Smani, T.; Salido, G.M.; Rosado, J.A. Orai2 Modulates Store-Operated Ca(2+) Entry and Cell Cycle Progression in Breast Cancer Cells. Cancers 2021, 14, 114. [Google Scholar] [CrossRef] [PubMed]
  126. Diez-Bello, R.; Jardin, I.; Salido, G.M.; Rosado, J.A. Orai1 and Orai2 mediate store-operated calcium entry that regulates HL60 cell migration and FAK phosphorylation. Biochim. Biophys. Acta Mol. Cell Res. 2017, 1864, 1064–1070. [Google Scholar] [CrossRef]
  127. Hasna, J.; Hague, F.; Rodat-Despoix, L.; Geerts, D.; Leroy, C.; Tulasne, D.; Ouadid-Ahidouch, H.; Kischel, P. Orai3 calcium channel and resistance to chemotherapy in breast cancer cells: The p53 connection. Cell Death Differ. 2018, 25, 693–707. [Google Scholar] [CrossRef]
  128. Benzerdjeb, N.; Sevestre, H.; Ahidouch, A.; Ouadid-Ahidouch, H. Orai3 is a predictive marker of metastasis and survival in resectable lung adenocarcinoma. Oncotarget 2016, 7, 81588–81597. [Google Scholar] [CrossRef] [Green Version]
  129. Arora, S.; Tanwar, J.; Sharma, N.; Saurav, S.; Motiani, R.K. Orai3 Regulates Pancreatic Cancer Metastasis by Encoding a Functional Store Operated Calcium Entry Channel. Cancers 2021, 13, 5937. [Google Scholar] [CrossRef]
  130. Dubois, C.; Vanden Abeele, F.; Lehen’kyi, V.; Gkika, D.; Guarmit, B.; Lepage, G.; Slomianny, C.; Borowiec, A.S.; Bidaux, G.; Benahmed, M.; et al. Remodeling of channel-forming ORAI proteins determines an oncogenic switch in prostate cancer. Cancer Cell 2014, 26, 19–32. [Google Scholar] [CrossRef] [Green Version]
  131. Tanwar, J.; Arora, S.; Motiani, R.K. Orai3: Oncochannel with therapeutic potential. Cell Calcium 2020, 90, 102247. [Google Scholar] [CrossRef] [PubMed]
  132. Chalmers, S.B.; Monteith, G.R. ORAI channels and cancer. Cell Calcium 2018, 74, 160–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Jardin, I.; Lopez, J.J.; Salido, G.M.; Rosado, J.A. Store-Operated Ca2+ Entry in Breast Cancer Cells: Remodeling and Functional Role. Int. J. Mol. Sci. 2018, 19, 4053. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Sanchez-Collado, J.; Jardin, I.; Lopez, J.J.; Ronco, V.; Salido, G.M.; Dubois, C.; Prevarskaya, N.; Rosado, J.A. Role of Orai3 in the Pathophysiology of Cancer. Int. J. Mol. Sci. 2021, 22, 11426. [Google Scholar] [CrossRef]
  135. Vashisht, A.; Trebak, M.; Motiani, R.K. STIM and Orai proteins as novel targets for cancer therapy. A Review in the Theme: Cell and Molecular Processes in Cancer Metastasis. Am. J. Physiol. Cell Physiol. 2015, 309, C457–C469. [Google Scholar] [CrossRef] [Green Version]
  136. Goswamee, P.; Pounardjian, T.; Giovannucci, D.R. Arachidonic acid-induced Ca2+ entry and migration in a neuroendocrine cancer cell line. Cancer Cell Int. 2018, 18, 30. [Google Scholar] [CrossRef] [Green Version]
  137. Fiorio Pla, A.; Grange, C.; Antoniotti, S.; Tomatis, C.; Merlino, A.; Bussolati, B.; Munaron, L. Arachidonic acid-induced Ca2+ entry is involved in early steps of tumor angiogenesis. Mol. Cancer Res. 2008, 6, 535–545. [Google Scholar] [CrossRef] [Green Version]
  138. Baron, S.; Vangheluwe, P.; Sepulveda, M.R.; Wuytack, F.; Raeymaekers, L.; Vanoevelen, J. The secretory pathway Ca2+-ATPase 1 is associated with cholesterol-rich microdomains of human colon adenocarcinoma cells. Biochim. Biophys. Acta 2010, 1798, 1512–1521. [Google Scholar] [CrossRef] [Green Version]
  139. Feng, M.; Grice, D.M.; Faddy, H.M.; Nguyen, N.; Leitch, S.; Wang, Y.; Muend, S.; Kenny, P.A.; Sukumar, S.; Roberts-Thomson, S.J.; et al. Store-independent activation of Orai1 by SPCA2 in mammary tumors. Cell 2010, 143, 84–98. [Google Scholar] [CrossRef] [Green Version]
  140. Feng, M.Y.; Rao, R. New insights into store-independent Ca2+ entry: Secretory pathway calcium ATPase 2 in normal physiology and cancer. Int. J. Oral. Sci. 2013, 5, 71–74. [Google Scholar] [CrossRef]
  141. Chantome, A.; Potier-Cartereau, M.; Clarysse, L.; Fromont, G.; Marionneau-Lambot, S.; Gueguinou, M.; Pages, J.C.; Collin, C.; Oullier, T.; Girault, A.; et al. Pivotal role of the lipid Raft SK3-Orai1 complex in human cancer cell migration and bone metastases. Cancer Res. 2013, 73, 4852–4861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Gueguinou, M.; Harnois, T.; Crottes, D.; Uguen, A.; Deliot, N.; Gambade, A.; Chantome, A.; Haelters, J.P.; Jaffres, P.A.; Jourdan, M.L.; et al. SK3/TRPC1/Orai1 complex regulates SOCE-dependent colon cancer cell migration: A novel opportunity to modulate anti-EGFR mAb action by the alkyl-lipid Ohmline. Oncotarget 2016, 7, 36168–36184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Badaoui, M.; Mimsy-Julienne, C.; Saby, C.; Van Gulick, L.; Peretti, M.; Jeannesson, P.; Morjani, H.; Ouadid-Ahidouch, H. Collagen type 1 promotes survival of human breast cancer cells by overexpressing Kv10.1 potassium and Orai1 calcium channels through DDR1-dependent pathway. Oncotarget 2018, 9, 24653–24671. [Google Scholar] [CrossRef] [PubMed]
  144. Hammadi, M.; Chopin, V.; Matifat, F.; Dhennin-Duthille, I.; Chasseraud, M.; Sevestre, H.; Ouadid-Ahidouch, H. Human ether a-gogo K(+) channel 1 (hEag1) regulates MDA-MB-231 breast cancer cell migration through Orai1-dependent calcium entry. J. Cell Physiol. 2012, 227, 3837–3846. [Google Scholar] [CrossRef]
  145. Tiffner, A.; Hopl, V.; Schober, R.; Sallinger, M.; Grabmayr, H.; Hoglinger, C.; Fahrner, M.; Lunz, V.; Maltan, L.; Frischauf, I.; et al. Orai1 Boosts SK3 Channel Activation. Cancers 2021, 13, 6357. [Google Scholar] [CrossRef]
  146. Peretti, M.; Badaoui, M.; Girault, A.; Van Gulick, L.; Mabille, M.P.; Tebbakha, R.; Sevestre, H.; Morjani, H.; Ouadid-Ahidouch, H. Original association of ion transporters mediates the ECM-induced breast cancer cell survival: Kv10.1-Orai1-SPCA2 partnership. Sci. Rep. 2019, 9, 1175. [Google Scholar] [CrossRef]
  147. Daya, H.A.; Kouba, S.; Ouled-Haddou, H.; Benzerdjeb, N.; Telliez, M.S.; Dayen, C.; Sevestre, H.; Garcon, L.; Hague, F.; Ouadid-Ahidouch, H. Orai3-Mediates Cisplatin-Resistance in Non-Small Cell Lung Cancer Cells by Enriching Cancer Stem Cell Population through PI3K/AKT Pathway. Cancers 2021, 13, 2314. [Google Scholar] [CrossRef]
  148. Terrie, E.; Deliot, N.; Benzidane, Y.; Harnois, T.; Cousin, L.; Bois, P.; Oliver, L.; Arnault, P.; Vallette, F.; Constantin, B.; et al. Store-Operated Calcium Channels Control Proliferation and Self-Renewal of Cancer Stem Cells from Glioblastoma. Cancers 2021, 13, 3428. [Google Scholar] [CrossRef]
  149. Schmidt, S.; Liu, G.; Liu, G.; Yang, W.; Honisch, S.; Pantelakos, S.; Stournaras, C.; Honig, A.; Lang, F. Enhanced Orai1 and STIM1 expression as well as store operated Ca2+ entry in therapy resistant ovary carcinoma cells. Oncotarget 2014, 5, 4799–4810. [Google Scholar] [CrossRef] [Green Version]
  150. Flourakis, M.; Lehen’kyi, V.; Beck, B.; Raphael, M.; Vandenberghe, M.; Abeele, F.V.; Roudbaraki, M.; Lepage, G.; Mauroy, B.; Romanin, C.; et al. Orai1 contributes to the establishment of an apoptosis-resistant phenotype in prostate cancer cells. Cell Death Dis. 2010, 1, e75. [Google Scholar] [CrossRef] [Green Version]
  151. Faouzi, M.; Kischel, P.; Hague, F.; Ahidouch, A.; Benzerdjeb, N.; Sevestre, H.; Penner, R.; Ouadid-Ahidouch, H. ORAI3 silencing alters cell proliferation and cell cycle progression via c-myc pathway in breast cancer cells. Biochim. Biophys. Acta 2013, 1833, 752–760. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Notch, Wnt, and Hedgehog pathways in CSC. NECD, Notch extracellular domain; NICD, Notch intracellular domain; Mam, Mastermind; Dvl, Dishevelled; TCF/LEF, T-cell factor/lymphoid enhancer factor; Hh, hedgehog; Smo, Smoothened; Gli, glioma-associated oncogene; Daam, Dishevelled associated activator of morphogenesis; RhoK, Rho-kinase; CN, calcineurin; NFAT, nuclear factor of activated T-cells.
Figure 1. Notch, Wnt, and Hedgehog pathways in CSC. NECD, Notch extracellular domain; NICD, Notch intracellular domain; Mam, Mastermind; Dvl, Dishevelled; TCF/LEF, T-cell factor/lymphoid enhancer factor; Hh, hedgehog; Smo, Smoothened; Gli, glioma-associated oncogene; Daam, Dishevelled associated activator of morphogenesis; RhoK, Rho-kinase; CN, calcineurin; NFAT, nuclear factor of activated T-cells.
Cells 11 01332 g001
Figure 2. Store-operated Ca2+ in cancer stem cells. Cartoon depicting a hypothetical mechanism of SOCE in CSC, based on our current knowledge in non-tumoral cells: (a) when intracellular Ca2+ stores are filled, STIM proteins remain in their quiescent conformation; (b) small changes in luminal Ca2+ concentration are controlled by STIM2, briefly activating CRAC channels to replenish the reservoirs; (c) massive Ca2+ store depletion triggers the activation of STIM proteins and the channels in plasma membrane, resulting in the critical increase in cytosolic Ca2+ required for tumorigenic responses.
Figure 2. Store-operated Ca2+ in cancer stem cells. Cartoon depicting a hypothetical mechanism of SOCE in CSC, based on our current knowledge in non-tumoral cells: (a) when intracellular Ca2+ stores are filled, STIM proteins remain in their quiescent conformation; (b) small changes in luminal Ca2+ concentration are controlled by STIM2, briefly activating CRAC channels to replenish the reservoirs; (c) massive Ca2+ store depletion triggers the activation of STIM proteins and the channels in plasma membrane, resulting in the critical increase in cytosolic Ca2+ required for tumorigenic responses.
Cells 11 01332 g002
Table 1. Implications of calcium-related proteins in cancer stem cells. ND: not determined. VGCC: voltage-gated calcium channels.
Table 1. Implications of calcium-related proteins in cancer stem cells. ND: not determined. VGCC: voltage-gated calcium channels.
Ca2+ Pumps and Exchangers
ProteinExpression/Functional Change in CSCCSC TypeRole in CSCSignaling Pathway ActivatedRef.
SERCAOverexpressionBreast cancer stem cellsCell survival in glucose-deprived conditionsDecrease [Ca2+]c and avoid Ca2+-dependent apoptosis during glucose deprivation[73]
ER Ca2+ channels
ProteinExpression/Functional Change in CSCCSC TypeRole in CSCSignaling Pathway ActivatedRef.
RyR1HIF-depended activationBreast cancer stem cellsChemoresistancePYK2/SRC/STAT3 signaling pathway[50]
IP3RChannel activationMelanoma stem cellsStemness maintenanceND[49]
Channel activationGlioblastoma stem cellCell self-renewal
Chemoresistance
ND[48]
Non-SOCE channels
ProteinExpression/Functional Change in CSCCSC TypeRole in CSCSignaling Pathway ActivatedRef.
VGCC
L- and T-typeOverexpressionOvarian cancer stem cellsTumor spheres formation
Apoptosis resistance
Stemness maintenance
Increase the transcription of Oct, Nanog, and Sox2 via ERK1/2 and AKT signaling pathways[53]
T-type calcium channelOverexpression
(Cav3.2)
Glioblastoma stem cellsApoptosis resistance
Chemoresistance
Stemness maintenance
Increase cell survival via AKT/mTOR pathways[52]
OverexpressionGlioblastoma stem cellsApoptosis resistanceStimulate Na+-dependent nutrient transport[54]
α2δ1
subunit
OverexpressionSmall cell lung cancer stem cellsChemoresistanceMEK/ERK signal pathway??[55]
α2δ1
subunit
OverexpressionNon-small cell lung cancer stem cellsChemoresistance
Cell Survival
Stemness maintenance
Notch3 activation via Ca2+-Calcineurin/NFATc2 signaling pathway[56]
OverexpressionBreast cancer stem cellsStemness maintenance
Cell self-renewal
ND[57]
OverexpressionHepatocellular cancer stem cellsCell self-renewal
Cell survival
Stemness maintenance
ERK1/2 MAPK signaling pathway[58]
OverexpressionLaryngeal squamous cancer stem cellsTumor spheres formation
Chemoresistance
Tumorigenesis
Stemness maintenance
ND[59]
ProteinExpression/Functional Change in CSCCSC TypeRole in CSCSignaling Pathway ActivatedRef.
TRP Channels
TRPC3OverexpressionBreast cancer stem cellsCell self-renewalIncrease IL-8 secretion via LPA/LPAR3/TRPC3 pathway[60]
TRPM7OverexpressionLung cancer stem cellsTumor spheres formation
Stemness maintenance
Hsp90α/uPA/MMP2 signaling pathway[61]
Channel activationGlioblastoma stem cellsStemness maintenance
Cell proliferation, migration and invasion
STAT3 and Notch signaling pathways[62]
OverexpressionNeuroblastoma stem cellsStemness maintenanceND[63]
TRPA1Channel activation
Overexpression
Glioma stem cellsCell differentiation
Apoptotic cell death
Stemness loss
ND[69]
TRPV1Channel activation
Overexpression
Glioma stem cellsCell differentiation
Apoptotic cell death
Stemness loss
ND[69]
TRPV2OverexpressionEsophageal squamous cancer stem cellsStemness maintenance
Cell proliferation
ND[65]
Channel activation
Overexpression
Glioblastoma stem cellsStem cell
differentiation
Reduce self-renewal capacity
Apoptotic cell death
AKT-PI3K/RPS6KBI/PTEN signaling pathway[64,66,67]
Channel activation
Overexpression
Liver cancer stem cellsImpair tumor spheres formation and self-renewal capacity
Stemness loss
ND[68]
SOCE channels
ProteinExpression/Functional Change in CSCCSC TypeRole in CSCSignaling Pathway ActivatedRef.
Orai1Overexpressionoral/oropharyngeal squamous cancer stem cellsTumor spheres formation
Cell self-renewal
Stemness maintenance
NFAT signaling pathway[119]
OverexpressionGlioblastoma stem cellTumor spheres formation
Cell self-renewal
Stemness maintenance
ND[148]
Orai3OverexpressionNon-small cell lung cancer stem cellsChemoresistance
Stemness maintenance
PI3K/AKT signaling pathway[147]
SOC ChannelsChannel activationGlioblastoma stem cellCell proliferationUp-regulation of CDKN1A and G0S2 and the down-regulation of CCNB1 genes[116]
Channel activationLiver cancer stem cellsStemness maintenance
Tumor spheres formation
Cell self-renewal
FGF19/SOCE/NFATc2 signaling pathway[117]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Jardin, I.; Lopez, J.J.; Sanchez-Collado, J.; Gomez, L.J.; Salido, G.M.; Rosado, J.A. Store-Operated Calcium Entry and Its Implications in Cancer Stem Cells. Cells 2022, 11, 1332. https://doi.org/10.3390/cells11081332

AMA Style

Jardin I, Lopez JJ, Sanchez-Collado J, Gomez LJ, Salido GM, Rosado JA. Store-Operated Calcium Entry and Its Implications in Cancer Stem Cells. Cells. 2022; 11(8):1332. https://doi.org/10.3390/cells11081332

Chicago/Turabian Style

Jardin, Isaac, Jose J. Lopez, Jose Sanchez-Collado, Luis J. Gomez, Gines M. Salido, and Juan A. Rosado. 2022. "Store-Operated Calcium Entry and Its Implications in Cancer Stem Cells" Cells 11, no. 8: 1332. https://doi.org/10.3390/cells11081332

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop