Next Article in Journal
Myelopoiesis during Solid Cancers and Strategies for Immunotherapy
Next Article in Special Issue
Targeting HDACs in Pancreatic Neuroendocrine Tumor Models
Previous Article in Journal
Selection, Expansion, and Unique Pretreatment of Allogeneic Human Natural Killer Cells with Anti-CD38 Monoclonal Antibody for Efficient Multiple Myeloma Treatment
Previous Article in Special Issue
RNA Extraction from Endoscopic Ultrasound-Acquired Tissue of Pancreatic Cancer Is Feasible and Allows Investigation of Molecular Features
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Store-Operated Calcium Entry: Shaping the Transcriptional and Epigenetic Landscape in Pancreatic Cancer

1
Clinic for General, Visceral and Pediatric Surgery, University Medical Center Göttingen, 37075 Göttingen, Germany
2
Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
*
Author to whom correspondence should be addressed.
Cells 2021, 10(5), 966; https://doi.org/10.3390/cells10050966
Submission received: 26 March 2021 / Revised: 13 April 2021 / Accepted: 19 April 2021 / Published: 21 April 2021
(This article belongs to the Special Issue Genome Dynamics in Pancreatic Cancer Biology and Therapy)

Abstract

:
Pancreatic ductal adenocarcinoma (PDAC) displays a particularly poor prognosis and low survival rate, mainly due to late diagnosis and high incidence of chemotherapy resistance. Genomic aberrations, together with changes in the epigenomic profile, elicit a shift in cellular signaling response and a transcriptional reprograming in pancreatic tumors. This endows them with malignant attributes that enable them to not only overcome chemotherapeutic challenges, but to also attain diverse oncogenic properties. In fact, certain genetic amplifications elicit a rewiring of calcium signaling, which can confer ER stress resistance to tumors while also aberrantly activating known drivers of oncogenic programs such as NFAT. While calcium is a well-known second messenger, the transcriptional programs driven by aberrant calcium signaling remain largely undescribed in pancreatic cancer. In this review, we focus on calcium-dependent signaling and its role in epigenetic programs and transcriptional regulation. We also briefly discuss genetic aberration events, exemplifying how genetic alterations can rewire cellular signaling cascades, including calcium-dependent ones.

1. Introduction

Chemoresistance is a main contributor to the low survival rates faced by pancreatic ductal adenocarcinoma (PDAC) patients [1]. Marked by high genomic instability, pancreatic tumors readily adapt to selective pressures, showing high epigenomic and transcriptomic plasticity [2,3,4]. In fact, genomic amplifications of drug-metabolizing enzymes have been observed in pancreatic cancer as well as in other types of cancer as being the main drivers of acquired chemoresistance [5,6]. Interestingly, several other genes are commonly co-amplified during these genomic aberration events, possibly conferring tumors with additional oncogenic properties. For example, upon gemcitabine resistance acquisition in pancreatic cancer as well as in non-small-cell lung carcinoma, an amplification of one of the main targets of gemcitabine, ribonucleotide reductase catalytic subunit M1 (RRM1), was observed. This amplification of RRM1 was accompanied by a gain in copy number and overexpression of other genes, such as stromal interaction molecule 1 (STIM1) [5,7]. Interestingly, the co-amplification of STIM1 induced a shift in calcium signaling, leading to aberrant de-regulation of various transcriptional programs [5]. In fact, calcium signaling has been described to drive key oncogenic processes such as proliferation, migration, invasion and angiogenesis [8,9], but the effects of its de-regulation in PDAC remain largely unknown. Thus, it is plausible that several other genetic aberrations, which would be considered as passenger effects, actually endow the tumor with additional properties. It is therefore of utmost importance to understand and characterize the underlying consequences that various genomic aberrations have in shaping cellular signaling response and transcriptional programs to leverage these dependencies as novel therapeutic targets. Here, we review how selective pressures imposed on pancreatic tumors promote genomic aberrations, which in turn divert the cellular signaling response to stimuli, eliciting transcriptional reprogramming with a primary emphasis on calcium signaling in pancreatic cancer.

2. Genetic Amplifications in Pancreatic Cancer

Tumor cells commonly display genomic aberrations such as the amplification or deletion of entire segments of their chromosomes. Such genomic rearrangements are mainly due to failures to conventionally resolve double stranded breaks, while also being a consequence of major catastrophic events [10,11,12]. Thus, copy number variations take place randomly and may be selected for as they provide tumors with additional characteristics. In fact, the development of pancreatic cancer is marked by several genetic aberrations such as KRAS mutation and amplification, cyclin-dependent kinase inhibitor 2A (CDKN2A) silencing or loss of heterozygosity as well as mutation or loss of tumor protein 53 (TP53) and deletion of SMAD family member 4 (SMAD4) [13,14,15]. This suggests that many early pro-tumorigenic events take place in the form of genomic aberrations. They shape the cellular transcriptome, while also affecting several processes such as DNA damage response, cell cycle arrest and cellular signaling response.
In addition to its crucial role in the development of cancer, genomic amplifications may affect tumor response to chemotherapy or take place after chemotherapeutic treatment. This may be due to the high genomic instability and plasticity associated with cancer cells. Several studies have described a correlation between the genetic background of a tumor and its responsiveness to chemotherapy. For example, PDAC response to platinum-based agents has been associated with genomic instability and mutation frequency of DNA damage responsive genes, such as BRCA1 and BRCA2 [16,17]. Furthermore, major KRAS genomic imbalances have been correlated with higher chemotherapy resistance, worse prognosis and more aggressive metastatic subtypes [4]. Key mitogen-activated protein kinase (MAPK) signaling genes, such as epidermal growth factor receptor (EGFR), Erb-b2 receptor tyrosine kinase 2 (ERBB2) or AKT serine/threonine kinase 1 (AKT1), are also commonly amplified in PDAC. Tumors with such alterations displayed higher sensitivity to receptor tyrosine kinase inhibitors, suggesting that these tumors show a greater dependence on MAPK signaling [17,18]. This suggests that the genetic background of cells may highly influence their sensitivity to chemotherapy and shape their cellular dependencies. It further implies that tumors are extremely plastic, favoring certain genetic mutations and/or undergoing major genomic rearrangements as a consequence of selective pressure.
Co-amplifications may reinforce each other or confer independent properties to tumors. Interestingly, some genomic aberration events have been reported to present a high co-occurrence rate, while others have been shown to be mutually exclusive, revealing the importance of the selection applied on tumors. Mutual exclusivity can be a result of genomic aberration redundancy or synthetic lethality, where cells cannot handle both alterations at the same time. On the other hand, co-occurrence of genomic aberrations suggests that the alterations complement each other [19,20]. Thus, characterizing gene amplifications is not only valuable to dissect cancer development, but also to better understand how tumors may adapt to stresses, such as chemotherapeutic agents. Even though specific gene amplifications are regarded as drivers of a certain cancer type or chemotherapeutic resistance, co-amplified genes may also contribute to tumorigenesis. Therefore, considering the genetic background is crucial, while dissecting the interplay between genomic aberrations may widen our understanding of cancer and cancer cell plasticity.
One such example is the amplification of ERBB2 in breast cancer. The amplified region extends over 100 KB on chr17 and encompasses not only ERBB2, but also several other genes including TCAP, PNMT, PGAP3, MIEN1 and GRB7 [21,22]. Later studies revealed that the co-amplification of MIEN1 facilitates increased migratory potential in breast cancer in a HER2-independent manner. In this case, MIEN1 was shown to promote migration by triggering downstream Syk-dependent signaling and interacting with Annexin A2 [23,24]. On the other hand, the overexpression of GRB7 further augments the effects of HER2 overexpression. GRB7 interacts with HER2 directly via its SH2-domain, and activates Ras triggering the initiation of the MAPK signaling cascade upon EGF signaling [25,26]. This reinforces the dependency of HER2 amplified tumors on MAPK signaling, making them especially vulnerable to HER2 and MAPK inhibitors [27]. Thus, the overexpression of co-amplified genes may synergistically promote tumorigenesis or endow cells with different oncogenic properties. Furthermore, by co-amplifying several components of a signaling cascade, cancer cells rewire their response to stimuli and present a shift in their dependencies.
Another example of such an occurrence is the co-amplification of WEE1 and ILK on chromosome 11 upon acquired gemcitabine resistance [5]. The overexpression of WEE1 may augment gemcitabine resistance, as gemcitabine targets DNA polymerase triggering replication stress, while WEE1 is known to protect cells from DNA damage by inducing cell cycle arrest [28,29]. This way, the upregulation of WEE1 could play a cytoprotective role upon gemcitabine treatment. In fact, WEE1 inhibition in combination with gemcitabine and radiation therapy has shown promising results in pancreatic cancer patients [30,31]. On the other hand, ILK has been shown to promote cell proliferation and migration by activating the Akt/mTOR and glycogen synthase kinase 3β (GSK-3β) pathways, while also fostering the expression of Snail family transcriptional repressor 1 (SNAI1) [32,33]. Furthermore, ILK inhibition in pancreatic cancer has been proven to prevent tumor growth and to synergize with acute gemcitabine treatment [34]. Thus, while the overexpression of WEE1 and ILK may both contribute to gemcitabine resistance, they likely elicit different dependencies on cell cycle and Akt/mTOR signaling, respectively. Still, the exact cellular properties conferred by WEE1 and ILK amplification upon gemcitabine resistance in PDAC have not been addressed.
Similarly, other genes amplified upon gemcitabine resistance in pancreatic cancer are RRM1 and STIM1 [5]. The amplification and overexpression of RRM1 was shown to drive gemcitabine resistance, while the upregulation of STIM1 elicits a shift in calcium signaling, promoting ER stress resistance and aberrant transcriptional activation [5]. This deregulation elicits a crucial shift in pancreatic cancer, as calcium is a major second messenger of several intracellular signaling cascades, influencing various cellular processes, such as transcription, metabolism, stress response [8,9,35]. Consistently, the de-regulation of calcium signaling and homeostasis is tightly associated with pancreatitis, which is a major risk factor for pancreatic cancer [36,37,38,39,40], and has been suggested to take place early in PDAC development [41]. In fact, several genes associated with calcium signaling have been shown to be differentially methylated upon pancreatic cancer development [41]. Thus, there is a need to better elucidate the role of calcium signaling deregulation in pancreatic cancer.

3. The Role of Calcium in the Cell

The importance of calcium in mediating cellular responses to environmental stimuli was first reported in 1883, where electrolytes were described to be essential for muscle contraction [42]. Since then, great advances in calcium signaling have been made and its importance in a variety of cellular processes recognized. The intensity of calcium uptake by the cell and its organelles, together with the amount of calcium released by cellular stores, determines the kinetics and intensity of the response to a stimulus. Furthermore, changes in calcium levels in different cellular compartments can have drastic consequences. Increases in cytoplasmic calcium levels can trigger the activation of several calmodulin-dependent proteins culminating in the activation of a diverse range of transcription factors [35,43,44,45].

3.1. Calcium-Dependent Signaling

In non-excitable cell systems, G protein-coupled receptors (GPCRs) and receptor tyrosine kinases (RTKs) relay external events to the cell often by activating calcium-dependent signaling cascades in the cell. Upon substrate binding, GPCRs and RTKs are activated and lead to phosphorylation of phospholipase C (PLC). Once activated, PLC hydrolyzes phosphatidylinositol 4,5-bisphosphate (PIP2) into inositol 1,4,5-triphosphate (IP3) and diacylglycerol (DAG) [46,47]. IP3 then binds to its receptor (IP3R) in the ER, promoting the release of calcium from the ER into the cytoplasm. As cytoplasmic calcium levels increase, protein kinase C (PKC) binds calcium and DAG, thereby, becoming activated. PKC in turn phosphorylates several targets such as components of the MAPK pathway, amplifying the stimulus sensed by GPCRs and RTKs [48,49]. In this way, PKC activity and the overall signaling intensity are directly modulated by cytoplasmic calcium levels. Consequently, several calcium channels, pumps and exchangers modulate PKC activity (Figure 1).
STIM and calcium release-activated calcium channel (ORAI) positively regulate PKC by sensing calcium depletion from the ER. STIMs are ER transmembrane proteins which oligomerize upon ER calcium store depletion, activating ORAI at ER-cytoplasmic membrane junctions [50,51]. ORAI channel opening promotes the increase in cytoplasmic calcium levels in a process called store-operated calcium entry (SOCE) [51]. Alternatively, STIMs can activate canonical transient receptor protein (TRPC) together with ORAI, promoting the store-operated influx of calcium as well as other cations from the extracellular matrix into the cytoplasm [52]. In turn, ER calcium levels are restored by sarco/endoplasmic reticulum calcium-ATPase (SERCA), which hydrolyze ATP in order to transport calcium ions against the concentration gradient back into the ER [53] and thereby negatively regulate PKC activity [49]. Thus, GPCRs and RTKs rely on an increase in cytosolic calcium levels to activate PKC and the MAPK pathway. Consequently, the kinetics of the response are tightly regulated by calcium pumps, channels, and exchangers in all cellular compartments where their activities are closely interconnected.

3.2. Calcium-Dependent Transcription Factors

Increased cytoplasmic calcium levels lead to the activation of calmodulin-dependent proteins such as kinases and phosphatases. These, in turn, promote the activation or inactivation of several other transcription factors [54,55] (Figure 1). One of these important kinases is calcium/calmodulin-dependent protein kinase II (CAMKII) which translocates to the nucleus and phosphorylates calcium/calmodulin-dependent protein kinase IV (CAMKIV) upon activation. CAMKII and CAMKIV further phosphorylate and activate several transcription factors in the cytoplasm and the nucleus [56]. In the cytoplasm, CAMKII releases the inflammation-associated transcription factor nuclear factor-kappa B (NFκB) from an inhibitory complex by activating inhibitor of nuclear-factor kappa B kinase subunit beta (IKK2) [57,58,59]. Once in the nucleus, cAMP response element-binding protein (CREB) is activated upon phosphorylation of Ser133 by CAMKIV, while CAMKII can phosphorylate Ser133 or Ser142 of CREB, thereby activating or inhibiting it, respectively [60,61]. CREB hyperactivation has been tightly associated with cancer initiation, development and metastasis [62,63]. Another common target of CAMKII and CAMKIV is the serum response factor (SRF), which is activated upon phosphorylation by both enzymes [64,65], and is best characterized for driving key cell migration genes [66,67]. CAMKII activity is also linked to the cellular stress response, as it phosphorylates and activates the stress responsive transcription factor heat shock factor 1 (HSF1) at Ser230 [68,69]. HSF1 has been shown to not only enable tumor cells to thrive under stress, but to also foster cell proliferation, invasion and migration, while dampening inflammation and apoptotic programs [70,71]. Protein c-ets-1 (ETS1) is also inhibited upon phosphorylation by CAMKII [72,73]. ETS1 is best known for fostering cell invasion and playing an important role in angiogenesis, while also promoting epithelial-to-mesenchymal transition (EMT) in several cancers including PDAC [74]. Interestingly, CAMKII and CAMKIV can also phosphorylate histone deacetylase 4 (HDAC4) promoting its nuclear export. HDAC4 not only deacetylates histones but can also inhibit SRF and myocyte enhancer factor 2 (MEF2). This way, CAMKII and CAMKIV indirectly activate SRF, while also promoting the activity of MEF2, whose oncogenic or tumor suppressive roles remain to be further investigated [54,75].
The phosphatase calcineurin (CaN) also becomes active upon SOCE stimulation, dephosphorylating transcription factors which are usually kept inactive in the cytoplasm by phosphorylation. CaN can also be inhibited upon phosphorylation by CAMKII in a negative feedback loop [76,77]. CaN has been suggested to foster MEF2 activity when active, although the exact mechanism is still under investigation [78]. The main substrate of CaN is the nuclear factor of activated T cells (NFAT) family, which is crucial for T cell activation, differentiation and development, while also promoting cancer development [9,79]. Upon dephosphorylation by CaN, the nuclear localization signal of NFAT protein is unmasked, allowing NFATs to translocate into the nucleus and activate target genes [79]. Interestingly, NFATs have been described to dimerize with several members of the activator protein 1 (AP1) family of transcription factors including FOS and JUN, converging in a transcriptional program that is highly dependent on both MAPK and calcium signaling [80,81,82]. This may be of particular relevance to pancreatic tumors that highly depend on both mutant KRAS signaling and heightened SOCE.
In PDAC, NFATs have been extensively characterized as drivers of pancreatic cancer development and growth. In fact, NFATc1 activation, together with aberrant KRAS signaling, has been shown to drive acinar to ductal metaplasia [83]. The promoter of NFATc1 itself has been shown to be methylated by Enhancer of Zeste Homolog 2 (EZH2) and consequently silenced in pancreatic acinar cells. This was reversed upon KRAS activation during PDAC development, leading to the de-repression and concomitant activation of NFATc1 [84]. Importantly, NFATs then drive the expression of the ductal determining factor SRY-box transcription factor 9 (SOX9) fostering acinar to ductal metaplasia [83,85]. Furthermore, NFATs and signal transducer and activator of transcription 3 (STAT3) have been shown to be activated upon inflammation promoting inflammation-driven pancreatic cancer development. NFATc1-STAT3 dimers were shown to act on enhancer regions, promoting the transcription of Wnt family members, EGFR, and several other genes associated with stemness, migration, invasion and growth [86]. NFATc2 has also been shown to promote the silencing of the tumor suppressor CDKN2B, further fostering tumorigenesis [87].
In pancreatic cancer, NFATs have been described to promote cell proliferation and EMT. For example, transforming growth factor β (TGFβ) has been reported to activate NFAT, which in turn displaces SMAD family member 3 (SMAD3) repressor complexes allowing for the upregulation of MYC and ultimately promoting tumor growth [88,89]. Interestingly, NFAT binding to the MYC promoter region, following serum treatment, also fosters p300-depedent acetylation of nearby histones. This, in turn, facilitates the recruitment of ETS like-1 (ELK1) and allows for the upregulation of MYC [90]. Finally, NFATc1 has been shown to drive SRY-box transcription factor 2 (SOX2) expression, while also dimerizing with it and promoting metastasis by regulating EMT genes, such as zinc finger E-box-binding homeobox 1 (ZEB1) and SNAI1 [91]. Taken together, NFATs play a determining role in pancreatic cancer development, activating key proliferation as well as EMT genes.
In summary, calcium affects various transcription factor programs that are implicated in different cellular processes. Additionally, calcium was reported to play an important role in stress response, which frequently occurs in tumors.

3.3. Calcium and the ER Stress Response

ER stress results from protein chaperones in the ER responding to the accumulation of unfolded proteins triggered by internal as well as external cues. These stimuli encompass, among others, disturbances in the oxidizing environment and calcium levels in the ER as well as overall cellular ATP levels and culminate in the accumulation of unfolded proteins. Proteins being folded in the ER contain several disulfide bonds, requiring an oxidizing environment. Thus, failure to recycle protein disulfide isomerases upon oxidative stress leads to an accumulation of misfolded proteins which lack disulfide bonds. Furthermore, protein chaperones need ATP to bind and release proteins, coupling protein folding to overall energy levels in the cell. Calcium is a second messenger, which is stored in the ER and released upon distinct signals, including ER stress. It is buffered and used as a co-factor by protein chaperones, linking ER stress to several cellular processes such as apoptosis, oxidative phosphorylation and transcriptional activation [35,92,93].
In fact, ER stress is commonly characterized by aberrant ER calcium store depletion resulting from increased ER calcium leakage or IP3R activity [35]. This drop in ER calcium levels triggers the opening of ORAIs and TRPCs replenishing the ER with calcium and alleviating ER stress in macrophages [94]. Similarly, increased SOCE in STIM1-overexpressing cancer cells renders them resistant to ER stress [5], while decreased STIM1 and SOCE levels promote ER stress in pancreatic β cells [95]. Interestingly, ORAI1 and SOCE levels have been associated with increased ER stress-driven apoptosis in prostate cancer [96]. Thus, calcium signaling in the cytoplasm and the ER are tightly coupled to ER stress in a context-specific manner.
Moreover, the interface between the mitochondria and the ER, namely mitochondria associated membranes (MAMs), allows calcium to easily flow from the ER to the mitochondria, influencing energy production, reactive oxygen species (ROS) formation and cell death initiation. Upon several stimuli, ER calcium is released through the IP3R channel and taken up by the mitochondrial calcium uniporter (MCU) [9,43]. High frequency calcium oscillations result in the activation of calcium-dependent enzymes, boosting oxidative phosphorylation and thereby, ATP production. Still, a continuous overflow of calcium in the mitochondria can trigger apoptosis [35]. Thus, calcium flux in MAMs modulates oxidative phosphorylation as it is tightly linked to the maintenance of the oxidizing environment in the ER and to the production of ROS, a known trigger of ER stress [97]. Notably, decreased levels of the ER protein disulfide isomerases, thioredoxin related transmembrane protein 1 and 3 (TMX1 and TMX3), in melanoma cells have been shown to lead to less MAMs, lowered ER–mitochondria calcium flux and increased ROS levels [98]. This underscores the role of calcium and MAMs in redox homeostasis and its coupling to ER protein folding. Furthermore, ER stress mediators have been tightly associated with MAMs and have been suggested to modulate ER–mitochondria communication [35]. Thus, calcium signaling in MAMs fine tunes ER homeostasis, directly affecting ER protein folding and consequently triggering and shaping ER stress response.
In the ER, protein chaperones buffer almost all calcium in the ER, while also using it as a co-factor. One such example is Heat Shock Protein Family A (Hsp70) Member 5 (GRP78), which alone buffers about 25% of ER calcium [99]. Furthermore, the glycoprotein chaperones Calnexin and Calreticulin also bind calcium whereby the structure of the C-terminal domain of Calreticulin is highly dependent on how many calcium ions it binds and overall calcium levels in the ER [100]. Calcium buffering in the ER and in the cell is essential as it controls the amount of calcium which can be released upon a stimulus, fine tuning the cellular response to stimuli [101]. Furthermore, the chaperoning activity of GRP78, Calnexin and Calreticulin depends on calcium and is reduced in response to low calcium levels in the ER. Thus, calcium levels directly influence protein chaperoning activity and ER stress activation [102,103,104].
GRP78 is the main sensor of unfolded proteins and direct regulator of ER stress mediators, linking the cellular energy and ER calcium levels to ER stress. Under resting conditions, GRP78 inhibits ER stress mediators by directly interacting with their luminal domains, which in turn hinder the ATPase activity of GRP78. GRP78 binds unfolded proteins and hydrolyzes ATP to ADP upon stress, trapping the unfolded protein and dissociating from ER stress mediators triggering ER stress. Only after successful folding of the protein and exchanging ADP for ATP is GRP78 able to inhibit ER stress mediators again [105,106]. Thus, GRP78 is the main activator of the stress response, being the major sensor of accumulated unfolded proteins in the ER, and whose activity is directly modulated by cellular ATP as well as ER calcium levels (Figure 2).

3.4. ER Stress Responsive Transcription Factors

ER stress mediators are the bridge between ER homeostasis and the stress response, activating the bZIP transcription factors: Activating Transcription Factor 6 (ATF6), X-Box-Binding Protein 1 (XBP1) and ATF4. The ER stress mediators are comprised of three transmembrane ER proteins: ATF6, inositol requiring enzyme 1 (IRE1), and PKR-like ER kinase (PERK). While ATF6 activates UPR-responsive genes including mainly protein chaperone genes, IRE1 leads to the activation of c-Jun N-terminal Kinases (JNK) as well as of XBP1, promoting apoptotic programs as well as the upregulation of protein chaperone, lipid synthesis and ER-associated protein degradation (ERAD) genes [107,108,109]. PERK, in turn, activates the transcription factor Nuclear Factor Erythroid 2-related Factor 2 (NRF2), while triggering ATF4 accumulation [110,111].

3.4.1. ATF6

ATF6 is an ER-transmembrane protein that is vesicle transported to the Golgi apparatus upon ER stress. Site-1 protease (S1P) cleaves off the luminal domain of ATF6 in the ER and site-2 protease (S2P) removes the transmembrane anchor of ATF6. The N-terminus of ATF6 is then released into the cytosol translocating into the nucleus and activating several ER protein chaperone genes such as GRP78 and protein disulfide isomerase family A member 6 (PDIA6) [108,112,113,114]. Thus, ATF6 not only senses ER stress, but also activates stress-responsive genes to promote ER protein folding.

3.4.2. XBP1 and JNK

IRE1 is a kinase and endoribonuclease, which targets several mRNAs and miRNAs in a process called regulated IRE1-dependent decay (RIDD), while also excising an intron from the XBP1 mRNA [115,116]. IRE1 autophosphorylates in trans upon ER stress, leading to its oligomerization and stabilizing its RNase active site. Oligomerized IRE1 binds TNF receptor-associated factor 2 (TRAF2), leading to the activation of JNK, and the promotion of apoptosis by stimulating pro-apoptotic protein Bcl-2-like protein 11 (BIM) and inhibiting anti-apoptotic protein B-cell lymphoma 2 (BCL2) [117,118]. IRE1 further fosters pro-survival pathways by promoting the accumulation of XBP1. IRE1 splices out an intron of the XBP1 mRNA, leading to a shift in the open reading frame, such that the mRNA can be properly translated [119,120,121]. XBP1 then translocates to the nucleus where it activates many pro-survival genes including protein chaperones, ERAD subunits and lipid synthesis protein coding genes. Consequently, XBP1 promotes the proper folding of newly synthesized proteins and the degradation of unfolded proteins while also stimulating the production of phospholipids for ER membrane expansion [109,122]. Thus, IRE1 triggers pro-survival as well as apoptotic transcriptional programs in response to ER stress.

3.4.3. ATF4 and NRF2

The third ER stress mediator is PERK, which senses unfolded proteins in the ER and undergoes oligomerization and autophosphorylation. PERK phosphorylates itself as well as NRF2 and eukaryotic translation initiation factor 2 alpha subunit (eIF2α) [123,124,125,126,127]. Phosphorylation of NRF2 promotes its translocation to the nucleus and activates metabolic enzymes and antioxidant protein coding genes [123,124,128]. The phosphorylation of eIF2α by PERK leads to an inhibition of global cap-dependent translation and a cap-independent translation of ATF4, triggering the transcription of pro-survival and apoptotic genes.
ATF4 has been described to upregulate the expression of several autophagy genes such as microtubule-associated proteins 1A/1B light chain 3B (MAP1LC3), autophagy-related 5 (ATG5) and sequestosome 1 (SQSTM1) [129,130]. This helps catabolize proteins, replenishing the pool of amino acids, while also lowering the amount of unfolded proteins, thereby further alleviating stress [131,132]. Upon prolonged stress, ATF4 upregulates DNA damage-inducible transcript 3 (DDIT3), which encodes the transcription factor CHOP. In response to ER stress, ATF4 and CHOP have been shown to promote the transcription of several pro-apoptotic genes such as the Bcl-2 family members Bcl-2-binding component 3 (PUMA) and BIM [133,134]. Another important target gene of ATF4 and CHOP is tribbles pseudokinase 3 (TRIB3), which was shown to repress tumorigenesis and promote apoptosis by inhibiting AKT activation [135,136]. Furthermore, ATF4 and CHOP cooperatively promote the transcription of protein phosphatase 1, regulatory subunit 15A (GADD34), whose main substrate is peIF2α and which negatively regulates PERK-dependent ER stress response [126,137]. Thus, PERK activates NRF2 and leads to an antioxidant response and ATF4 accumulation upon ER stress, thereby promoting the transcription of pro-survival and apoptotic genes.
Taken together, ER stress is triggered by several stimuli including changes in ER calcium levels, which impart protein chaperoning activity eliciting an accumulation of unfolded proteins. This, in turn, leads to the activation of different ER stress mediators and a wide range of transcription factors which can then fine-tune the stress response and trigger the transcription of pro-survival genes or, if the cell fails to respond, of apoptotic genes.

3.5. ER Stress Response: Essential or Dispensable for the Tumor?

Several studies have pointed at the fact that tumors may take advantage of environmentally induced stress by hijacking the pro-survival branch of the stress response. In fact, the pro-survival ATF4-triggered response has been associated with several tumorigenic processes such as angiogenesis, metastasis, genomic instability, and therapeutic resistance [115,138,139,140,141]. One of the best characterized examples is hypoxia-triggered ER stress response and the subsequent adaptation to oxygen-deprivation. In this case, ATF4 activation has been proposed to be crucial for the transcription of angiogenic genes. Both ATF4 and XBP1 have been reported to bind the promoter region of vascular endothelial growth factor A (VEGFA) and ATF4 has been shown to be a driver of VEGFA expression upon stress [142,143]. Thus, it is possible that highly hypoxic tumors such as PDAC rather profit from the activation of ATF4.
Following this hypothesis, it is possible that PDAC tumors displaying genetic amplifications, which confer a dampened ER stress response, may be more vulnerable to hypoxia. While genetic amplifications, such as the overexpression of STIM1, may protect pancreatic cancer cells from the pro-apoptotic branch of the ER stress response [5], they may potentially also impede them from hijacking the pro-survival branch upon hypoxia and other ER stresses. One could speculate that these tumors would fail to activate ATF4, thus failing to upregulate VEGFA and to stabilize HIF1a upon hypoxia.
On the other hand, STIM1 and SOCE have been shown to promote HIF1a expression and stabilization during hepatocarcinogenesis. In this case, upon hypoxia, elevated SOCE leads to the activation of CAMKII and subsequently of the p300 acetyltransferase [144]. p300, in turn, interacts with the transactivation domain of HIF1a, stabilizing and preventing its degradation [145]. Interestingly, in a feedback mechanism, HIF1a has been shown to bind the promoter region of STIM1 increasing its expression upon hypoxia. Furthermore, several different tumor types have shown a positive correlation between STIM1 and HIF1a levels [144,146]. Thus, it is also plausible that some tumors rely on the activation of SOCE by STIM1 to cope with hypoxia.
For this reason, tumor cells harboring an amplification of STIM1 may be able to thrive under hypoxic conditions by activating HIF1a through SOCE, circumventing the dampened ER stress response and ATF4-dependent HIF1a activation. In this case, the benefits of hijacking the pro-survival pathway of the ER stress response upon hypoxia may become dispensable for the cell. Instead, tumors may employ SOCE-dependent alternative mechanisms and pathways to cope with oxygen-deprivation. Thus, redundant signaling pathways may aid tumors with very different genetic and epigenetic backgrounds to adapt to the same source of stress. This may apply not only to hypoxia-induced stress, but also to other external stimuli.

4. The Benefits and Drawbacks of Targeting Calcium Signaling in Pancreatic Cancer

As described in the sections above, co-amplifications acquired by tumors may divert the cellular signaling response and therefore the transcriptional dependencies of cancer cells. Calcium is an important second messenger involved in relaying diverse signals in the cell, ultimately leading to the activation or inhibition of a vast array of transcription factors. Thus, genomic aberrations involving calcium regulators may result in an altered calcium signaling response and an aberrant activation or repression of transcription. One such example is that of the co-amplification of STIM1 in pancreatic cancer cells upon the acquisition of gemcitabine resistance, which elicits a shift in calcium signaling and transcription factor dependencies [5]. The consequent increased SOCE and NFAT activity may confer the cells with several additional oncogenic properties, while the dampened response to ER stress protects the cells from apoptosis. Thus, genetic amplifications modulating calcium signaling elicit novel dependencies in tumors, which might be exploited for targeted therapy. In fact, the inhibition of SOCE or NFAT may prevent the activation of pro-tumorigenic processes and transcription factors, while targeting SOCE may also re-establish ER stress sensitivity (Figure 1).
NFAT activation can be targeted by inhibiting calcineurin activity with cyclosporine A (CSA) or FK506 (tacrolimus). These compounds are routinely used in the clinic as immunosuppressants, preventing organ transplantation rejection, and their potential in cancer treatment is still under investigation [147,148]. Studies have shown the benefits of using these FDA-approved drugs in the treatment of different cancer entities. In bladder cancer, both CSA and tacrolimus led to decreased migration and invasion in vitro as well as decreased tumor volume in vivo [149]. In breast cancer, tacrolimus treatment reduced cancer cell proliferation and migration, while inhibiting angiogenesis [150]. Still, the use of these compounds for cancer treatment has to be carefully assessed and treatment schedules wisely planned. Prolonged exposure to CSA or tacrolimus has been associated with increased cancer incidence [151,152]. However, both compounds elicit severe side effects [147,153]. Still, CSA or tacrolimus treatment would solely inhibit potential pro-oncogenic pathways that arise due to aberrant NFAT activation without affecting the aberrant activation or repression of other calcium-dependent transcription factors, while also not influencing other calcium-dependent cellular processes such as the ER stress response.
Thus, SOCE inhibitors could be employed in order to target the potential pro-oncogenic properties elicited by aberrant NFAT activation, while also blocking the aberrant activation or repression of other calcium-dependent transcription factors and sensitizing tumor cells to ER stress. In fact, treatment with the SOCE inhibitor RP4010 in pancreatic cancer led to reduced NFAT translocation and increased expression of eukaryotic translation initiation factor 4E-binding protein 1 (EIF4EBP1) [154]. Interestingly, ATF4 and MYC have been shown to upregulate EIF4EBP1 in order to cope with proteotoxic stress [116,155], suggesting increased activation of ATF4 upon RP4010 treatment. Furthermore, SOCE inhibition in PDAC has been shown to synergize with the standard chemotherapeutic agents, gemcitabine and nab-paclitaxel, in vitro as well as in patient-derived xenografts [154]. Even though no SOCE inhibitor has been approved by the FDA yet, several have been developed and some are being tested in clinical trials for the treatment of acute pancreatitis and pneumonia [156,157,158,159,160,161]. During preclinical studies, the SOCE inhibitors GSK7975A and CM128 have shown very promising results in treating acute pancreatitis while also preventing pancreatic acinar cell injury [162,163]. As clinical trials with CM4620 (also known as Auxora) and other SOCE inhibitors evolve, scientists may better gauge the impact of SOCE inhibition in the body. It is highly possible that SOCE inhibitors lead to the immunosuppression of patients, as SOCE is upstream of NFAT activation, and as the immunosuppressants CSA and tacrolimus are known repressors of NFAT activity. Furthermore, several important SOCE-dependent physiological processes such as the release of insulin in the pancreas, the glomerular hemodynamics in the kidney, the formation of osteoclasts in the bone and the differentiation of myoblasts in skeletal muscle may be transiently impaired during SOCE inhibitor treatment [164]. Still, the occurrence and severity of these potential side effects will largely depend on the concentration and duration of the inhibitor treatment.
Assuming CM4620 and/or other SOCE inhibitors are approved by the FDA and little to mild side effects are detected, SOCE inhibitors may be suitable candidates to treat pancreatic tumors in which STIM1 is overexpressed. Thus, assessing the expression of STIM1 in patient biopsies before and after chemotherapy may help stratify tumors and gauge their sensitivity to stress as well as their suitability for SOCE inhibitor treatments. Furthermore, these tumors may be more sensitive to alternative chemotherapies which trigger apoptosis via ER stress by restoring the cellular sensitivity to ER stress through SOCE inhibitor treatment. Taken together, SOCE inhibition may pose an alternative treatment option for pancreatic cancer patients presenting increased STIM1 expression and a priori or acquired gemcitabine resistance.

5. Conclusions

Taken together, certain cellular features, such as genetic aberrations, are selected for upon external or internal cues. These features shape the cellular response to stimuli, promoting a transcriptional program which enables the cells to thrive under such adverse conditions. One such example, described and discussed extensively in this review is that of the genetic alteration of calcium regulatory genes. Calcium is a key second messenger that fine tunes the cellular response to various internal and external cues, ultimately shaping the cellular transcriptome upon stimuli. Thus, in order to identify changes in cellular transcriptional program dependencies, the characterization of genomic aberration events and their consequences in calcium and overall cellular signaling response is of utmost importance. This may aid in the identification of novel druggable targets in precision oncology.

Author Contributions

A.P.K., S.A.J. and F.H.H. have written, edited and revised the manuscript. The majority of this manuscript, including slightly modified figures, were included in the dissertation of A.P.K. All authors have read and agreed to the published version of the manuscript.

Funding

This review did not receive external funding.

Acknowledgments

The authors would like to thank Xin Wang, Zeynab Najafova, Evangelos Prokakis, Florian Wegwitz, Alex Q. Wixom, Josephine Choo, Matthias Dobbelstein, Christine S. Gibhardt, Ivan Bogeski, Elisabeth Hessmann and Volker Ellenrieder for the fruitful discussions.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Viale, P.H. The American Cancer Society’s facts & figures. J. Adv. Pract. Oncol. 2020, 11, 135. [Google Scholar]
  2. Notta, F.; Chan-Seng-Yue, M.; Lemire, M.; Li, Y.; Wilson, G.W.; Connor, A.A.; Denroche, R.E.; Liang, S.-B.; Brown, A.M.K.; Kim, J.C.; et al. A renewed model of pancreatic cancer evolution based on genomic rearrangement patterns. Nature 2016, 538, 378–382. [Google Scholar] [CrossRef]
  3. Cortés-Ciriano, I.; Lee, J.J.-K.; Xi, R.; Jain, D.; Jung, Y.L.; Yang, L.; Gordenin, D.; Klimczak, L.J.; Zhang, C.-Z.; Pellman, D.S.; et al. Comprehensive analysis of chromothripsis in 2658 human cancers using whole-genome sequencing. Nat. Genet. 2020, 52, 331–341. [Google Scholar] [CrossRef] [Green Version]
  4. Chan-Seng-Yue, M.; Kim, J.C.; Wilson, G.W.; Ng, K.; Figueroa, E.F.; O’Kane, G.M.; Connor, A.A.; Denroche, R.E.; Grant, R.C.; McLeod, J.; et al. Transcription phenotypes of pancreatic cancer are driven by genomic events during tumor evolution. Nat. Genet. 2020, 52, 231–240. [Google Scholar] [CrossRef] [PubMed]
  5. Kutschat, A.P.; Hamdan, F.H.; Wang, X.; Wixom, A.Q.; Najafova, Z.; Gibhardt, C.S.; Kopp, W.; Gaedcke, J.; Ströbel, P.; Ellenrieder, V.; et al. STIM1 Mediates Calcium-dependent Epigenetic Reprogramming in Pancreatic Cancer. Cancer Res. 2021. [Google Scholar] [CrossRef]
  6. Tooker, P.; Yen, W.-C.; Ng, S.-C.; Negro-Vilar, A.; Hermann, T.W. Bexarotene (LGD1069, Targretin), a Selective Retinoid X Receptor Agonist, Prevents and Reverses Gemcitabine Resistance in NSCLC Cells by Modulating Gene Amplification. Cancer Res. 2007, 67, 4425–4433. [Google Scholar] [CrossRef] [Green Version]
  7. Zhou, J.; Zhang, L.; Zheng, H.; Ge, W.; Huang, Y.; Yan, Y.; Zhou, X.; Zhu, W.; Kong, Y.; Ding, Y.; et al. Identification of chemoresistance-related mRNAs based on gemcitabine-resistant pancreatic cancer cell lines. Cancer Med. 2019, 9, 1115–1130. [Google Scholar] [CrossRef]
  8. Marchi, S.; Giorgi, C.; Galluzzi, L.; Pinton, P. Ca2+ Fluxes and Cancer. Mol. Cell 2020, 78, 1055–1069. [Google Scholar] [CrossRef]
  9. Monteith, G.R.; Prevarskaya, N.; Roberts-Thomson, S.J. The calcium–cancer signalling nexus. Nat. Rev. Cancer 2017, 17, 373–380. [Google Scholar] [CrossRef] [Green Version]
  10. Willis, N.A.; Rass, E.; Scully, R. Deciphering the Code of the Cancer Genome: Mechanisms of Chromosome Rearrangement. Trends Cancer 2015, 1, 217–230. [Google Scholar] [CrossRef] [Green Version]
  11. Korbel, J.O.; Campbell, P.J. Criteria for Inference of Chromothripsis in Cancer Genomes. Cell 2013, 152, 1226–1236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Li, Y.; Roberts, N.D.; Wala, J.A.; Shapira, O.; Schumacher, S.E.; Kumar, K.; Khurana, E.; Waszak, S.; Korbel, J.O.; Haber, J.E.; et al. Patterns of somatic structural variation in human cancer genomes. Nature 2020, 578, 112–121. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Vogelstein, B.; Fearon, E.R.; Hamilton, S.R.; Kern, S.E.; Preisinger, A.C.; Leppert, M.; Smits, A.M.; Bos, J.L. Genetic Alterations during Colorectal-Tumor Development. N. Engl. J. Med. 1988, 319, 525–532. [Google Scholar] [CrossRef] [Green Version]
  14. Rozenblum, E.; Schutte, M.; Goggins, M.; Hahn, S.A.; Panzer, S.; Zahurak, M.; Goodman, S.N.; Sohn, T.A.; Hruban, R.H.; Yeo, C.J.; et al. Tumor-suppressive pathways in pancreatic carcinoma. Cancer Res. 1997, 57, 1731–1734. [Google Scholar]
  15. Distler, M.; Aust, D.; Weitz, J.; Pilarsky, C.; Grützmann, R. Precursor Lesions for Sporadic Pancreatic Cancer: PanIN, IPMN, and MCN. BioMed Res. Int. 2014, 2014, 1–11. [Google Scholar] [CrossRef] [PubMed]
  16. Waddell, N.; Pajic, M.; Patch, A.-M.; Chang, D.K.; Kassahn, K.S.; Bailey, P.; Johns, A.L.; Miller, D.; Nones, K.; Quek, K.; et al. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015, 518, 495–501. [Google Scholar] [CrossRef] [Green Version]
  17. Singhi, A.D.; George, B.; Greenbowe, J.R.; Chung, J.; Suh, J.; Maitra, A.; Klempner, S.J.; Hendifar, A.; Milind, J.M.; Golan, T.; et al. Real-Time Targeted Genome Profile Analysis of Pancreatic Ductal Adenocarcinomas Identifies Genetic Alterations That Might Be Targeted With Existing Drugs or Used as Biomarkers. Gastroenterology 2019, 156, 2242–2253.e4. [Google Scholar] [CrossRef] [Green Version]
  18. Tiriac, H.; Belleau, P.; Engle, D.D.; Plenker, D.; Deschênes, A.; Somerville, T.D.D.; Froeling, F.E.M.; Burkhart, R.A.; Denroche, R.E.; Jang, G.H.; et al. Organoid Profiling Identifies Common Responders to Chemotherapy in Pancreatic Cancer. Cancer Discov. 2018, 8, 1112–1129. [Google Scholar] [CrossRef] [Green Version]
  19. Deng, N.; Goh, L.K.; Wang, H.; Das, K.; Tao, J.; Tan, I.B.; Zhang, S.; Lee, M.; Wu, J.; Lim, K.H.; et al. A comprehensive survey of genomic alterations in gastric cancer reveals systematic patterns of molecular exclusivity and co-occurrence among distinct therapeutic targets. Gut 2012, 61, 673–684. [Google Scholar] [CrossRef]
  20. Sanchez-Vega, F.; Mina, M.; Armenia, J.; Chatila, W.K.; Luna, A.; La, K.C.; Dimitriadoy, S.; Liu, D.L.; Kantheti, H.S.; Saghafinia, S.; et al. Oncogenic Signaling Pathways in The Cancer Genome Atlas. Cell 2018, 173, 321–337.e10. [Google Scholar] [CrossRef] [Green Version]
  21. Staaf, J.; Jönsson, G.; Ringnér, M.; Vallon-Christersson, J.; Grabau, D.; Arason, A.; Gunnarsson, H.; Agnarsson, B.A.; Malmström, P.-O.; Johannsson, O.T.; et al. High-resolution genomic and expression analyses of copy number alterations in HER2-amplified breast cancer. Breast Cancer Res. 2010, 12, 1–18. [Google Scholar] [CrossRef] [Green Version]
  22. Ferrari, A.; Vincent-Salomon, A.; Pivot, X.; Sertier, A.-S.; Thomas, E.; Tonon, L.; Boyault, S.; Mulugeta, E.; Treilleux, I.; MacGrogan, G.; et al. A whole-genome sequence and transcriptome perspective on HER2-positive breast cancers. Nat. Commun. 2016, 7, 12222. [Google Scholar] [CrossRef]
  23. Kpetemey, M.; Dasgupta, S.; Rajendiran, S.; Das, S.; Gibbs, L.D.; Shetty, P.; Gryczynski, Z.; Vishwanatha, J.K. MIEN1, a novel interactor of Annexin A2, promotes tumor cell migration by enhancing AnxA2 cell surface expression. Mol. Cancer 2015, 14, 156. [Google Scholar] [CrossRef] [Green Version]
  24. Katz, E.; Dubois-Marshall, S.; Sims, A.; Faratian, D.; Li, J.; Smith, E.S.; Quinn, J.A.; Edward, M.; Meehan, R.R.; Evans, E.E.; et al. A gene on the HER2 amplicon, C35, is an oncogene in breast cancer whose actions are prevented by inhibition of Syk. Br. J. Cancer 2010, 103, 401–410. [Google Scholar] [CrossRef] [Green Version]
  25. Janes, P.W.; Lackmann, M.; Church, W.B.; Sanderson, G.M.; Sutherland, R.L.; Daly, R.J. Structural Determinants of the Interaction between the erbB2 Receptor and the Src Homology 2 Domain of Grb7. J. Biol. Chem. 1997, 272, 8490–8497. [Google Scholar] [CrossRef] [Green Version]
  26. Chu, P.-Y.; Li, T.-K.; Ding, S.-T.; Lai, I.-R.; Shen, T.-L. EGF-induced Grb7 Recruits and Promotes Ras Activity Essential for the Tumorigenicity of Sk-Br3 Breast Cancer Cells. J. Biol. Chem. 2010, 285, 29279–29285. [Google Scholar] [CrossRef] [Green Version]
  27. Chu, P.-Y.; Tai, Y.-L.; Shen, T.-L. Grb7, a Critical Mediator of EGFR/ErbB Signaling, in Cancer Development and as a Potential Therapeutic Target. Cells 2019, 8, 435. [Google Scholar] [CrossRef] [Green Version]
  28. Beck, H.; Nähse-Kumpf, V.; Larsen, M.S.Y.; O’Hanlon, K.A.; Patzke, S.; Holmberg, C.; Mejlvang, J.; Groth, A.; Nielsen, O.; Syljuåsen, R.G.; et al. Cyclin-Dependent Kinase Suppression by WEE1 Kinase Protects the Genome through Control of Replication Initiation and Nucleotide Consumption. Mol. Cell. Biol. 2012, 32, 4226–4236. [Google Scholar] [CrossRef] [Green Version]
  29. Sørensen, C.S.; Syljuåsen, R.G. Safeguarding genome integrity: The checkpoint kinases ATR, CHK1 and WEE1 restrain CDK activity during normal DNA replication. Nucleic Acids Res. 2011, 40, 477–486. [Google Scholar] [CrossRef] [Green Version]
  30. Koh, S.-B.; Wallez, Y.; Dunlop, C.R.; Fernández, S.B.D.Q.; Bapiro, T.E.; Richards, F.M.; Jodrell, D.I. Mechanistic Distinctions between CHK1 and WEE1 Inhibition Guide the Scheduling of Triple Therapy with Gemcitabine. Cancer Res. 2018, 78, 3054–3066. [Google Scholar] [CrossRef] [Green Version]
  31. Cuneo, K.C.; Morgan, M.A.; Sahai, V.; Schipper, M.J.; Parsels, L.A.; Parsels, J.D.; Devasia, T.; Al-Hawaray, M.; Cho, C.S.; Nathan, H.; et al. Dose Escalation Trial of the Wee1 Inhibitor Adavosertib (AZD1775) in Combination With Gemcitabine and Radiation for Patients With Locally Advanced Pancreatic Cancer. J. Clin. Oncol. 2019, 37, 2643–2650. [Google Scholar] [CrossRef]
  32. Hannigan, G.E.; Troussard, A.A.; Dedhar, S. Integrin-linked kinase: A cancer therapeutic target unique among its ILK. Nat. Rev. Cancer 2005, 5, 51–63. [Google Scholar] [CrossRef]
  33. Sawai, H.; Okada, Y.; Funahashi, H.; Matsuo, Y.; Takahashi, H.; Takeyama, H.; Manabe, T. Integrin-linked kinase activity is associated with interleukin-1α-induced progressive behavior of pancreatic cancer and poor patient survival. Oncogene 2006, 25, 3237–3246. [Google Scholar] [CrossRef] [Green Version]
  34. Yau, C.Y. Inhibition of Integrin-Linked Kinase by a Selective Small Molecule Inhibitor, QLT0254, Inhibits the PI3K/PKB/mTOR, Stat3, and FKHR Pathways and Tumor Growth, and Enhances Gemcitabine-Induced Apoptosis in Human Orthotopic Primary Pancreatic Cancer Xenografts. Cancer Res. 2005, 65, 1497–1504. [Google Scholar] [CrossRef] [Green Version]
  35. Carreras-Sureda, A.; Pihán, P.; Hetz, C. Calcium signaling at the endoplasmic reticulum: Fine-tuning stress responses. Cell Calcium 2018, 70, 24–31. [Google Scholar] [CrossRef]
  36. Lee, P.J.; Papachristou, G.I. New insights into acute pancreatitis. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 479–496. [Google Scholar] [CrossRef]
  37. Gukovskaya, A.S.; Gorelick, F.S.; Groblewski, G.E.; Mareninova, O.A.; Lugea, A.; Antonucci, L.; Waldron, R.T.; Habtezion, A.; Karin, M.; Pandol, S.J.; et al. Recent Insights Into the Pathogenic Mechanism of Pancreatitis. Pancreas 2019, 48, 459–470. [Google Scholar] [CrossRef]
  38. Mayerle, J.; Sendler, M.; Hegyi, E.; Beyer, G.; Lerch, M.M.; Sahin-Tóth, M. Genetics, Cell Biology, and Pathophysiology of Pancreatitis. Gastroenterology 2019, 156, 1951–1968.e1. [Google Scholar] [CrossRef] [Green Version]
  39. Kirkegård, J.; Cronin-Fenton, D.; Heide-Jørgensen, U.; Mortensen, F.V. Acute Pancreatitis and Pancreatic Cancer Risk: A Nationwide Matched-Cohort Study in Denmark. Gastroenterology 2018, 154, 1729–1736. [Google Scholar] [CrossRef] [Green Version]
  40. Kirkegård, J.; Mortensen, F.V.; Cronin-Fenton, D. Chronic Pancreatitis and Pancreatic Cancer Risk: A Systematic Review and Meta-analysis. Am. J. Gastroenterol. 2017, 112, 1366–1372. [Google Scholar] [CrossRef] [Green Version]
  41. Gregório, C.; Soares-Lima, S.C.; Alemar, B.; Recamonde-Mendoza, M.; Camuzi, D.; De Souza-Santos, P.T.; Rivero, R.; Machado, S.; Osvaldt, A.; Ashton-Prolla, P.; et al. Calcium Signaling Alterations Caused by Epigenetic Mechanisms in Pancreatic Cancer: From Early Markers to Prognostic Impact. Cancers 2020, 12, 1735. [Google Scholar] [CrossRef]
  42. Ringer, S. A further Contribution regarding the influence of the different Constituents of the Blood on the Contraction of the Heart. J. Physiol. 1883, 4, 29–42. [Google Scholar] [CrossRef]
  43. Giorgi, C.; Marchi, S.; Pinton, P. The machineries, regulation and cellular functions of mitochondrial calcium. Nat. Rev. Mol. Cell Biol. 2018, 19, 713–730. [Google Scholar] [CrossRef]
  44. 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]
  45. Berridge, M.J.; Lipp, P.; Bootman, M.D. The versatility and universality of calcium signalling. Nat. Rev. Mol. Cell Biol. 2000, 1, 11–21. [Google Scholar] [CrossRef]
  46. Hanlon, C.D.; Andrew, D.J. Outside-in signaling—A brief review of GPCR signaling with a focus on the Drosophila GPCR family. J. Cell Sci. 2015, 128, 3533–3542. [Google Scholar] [CrossRef] [Green Version]
  47. Lemmon, M.A.; Schlessinger, J. Cell Signaling by Receptor Tyrosine Kinases. Cell 2010, 141, 1117–1134. [Google Scholar] [CrossRef] [Green Version]
  48. VanRenterghem, B.; Browning, M.; Maller, J. Regulation of mitogen-activated protein kinase activation by protein kinases A and C in a cell-free system. J. Biol. Chem. 1994, 269, 24666–24672. [Google Scholar] [CrossRef]
  49. Lipp, P.; Reither, G. Protein Kinase C: The “Masters” of Calcium and Lipid. Cold Spring Harb. Perspect. Biol. 2011, 3, a004556. [Google Scholar] [CrossRef]
  50. Ma, G.; Wei, M.; He, L.; Liu, C.; Wu, B.; Zhang, S.L.; Jing, J.; Liang, X.; Senes, A.; Tan, P.; et al. Inside-out Ca2+ signalling prompted by STIM1 conformational switch. Nat. Commun. 2015, 6, 7826. [Google Scholar] [CrossRef] [Green Version]
  51. Prakriya, M.; Lewis, R.S. Store-operated calcium channels. Physiol. Rev. 2015, 95, 1383–1436. [Google Scholar] [CrossRef] [Green Version]
  52. Lopez, J.J.; Jardin, I.; Sanchez-Collado, J.; Salido, G.M.; Smani, T.; Rosado, J.A. TRPC Channels in the SOCE Scenario. Cells 2020, 9, 126. [Google Scholar] [CrossRef] [Green Version]
  53. Vandecaetsbeek, I.; Vangheluwe, P.; Raeymaekers, L.; Wuytack, F.; Vanoevelen, J. The Ca2+ Pumps of the Endoplasmic Reticulum and Golgi Apparatus. Cold Spring Harb. Perspect. Biol. 2011, 3, a004184. [Google Scholar] [CrossRef] [Green Version]
  54. Dewenter, M.; Von Der Lieth, A.; Katus, H.A.; Backs, J. Calcium Signaling and Transcriptional Regulation in Cardiomyocytes. Circ. Res. 2017, 121, 1000–1020. [Google Scholar] [CrossRef]
  55. Feske, S. Calcium signalling in lymphocyte activation and disease. Nat. Rev. Immunol. 2007, 7, 690–702. [Google Scholar] [CrossRef]
  56. Ma, H.; Groth, R.D.; Cohen, S.M.; Emery, J.F.; Li, B.; Hoedt, E.; Zhang, G.; Neubert, T.A.; Tsien, R.W. γCaMKII Shuttles Ca2+/CaM to the Nucleus to Trigger CREB Phosphorylation and Gene Expression. Cell 2014, 159, 281–294. [Google Scholar] [CrossRef] [Green Version]
  57. Ling, H.; Gray, C.B.; Zambon, A.C.; Grimm, M.; Gu, Y.; Dalton, N.D.; Purcell, N.H.; Peterson, K.L.; Brown, J.H. Ca2+/Calmodulin-Dependent Protein Kinase II δ Mediates Myocardial Ischemia/Reperfusion Injury Through Nuclear Factor-κB. Circ. Res. 2013, 112, 935–944. [Google Scholar] [CrossRef] [Green Version]
  58. Kashiwase, K.; Higuchi, Y.; Hirotani, S.; Yamaguchi, O.; Hikoso, S.; Takeda, T.; Watanabe, T.; Taniike, M.; Nakai, A.; Tsujimoto, I.; et al. CaMKII activates ASK1 and NF-κB to induce cardiomyocyte hypertrophy. Biochem. Biophys. Res. Commun. 2005, 327, 136–142. [Google Scholar] [CrossRef] [PubMed]
  59. Gray, C.B.; Suetomi, T.; Xiang, S.; Mishra, S.; Blackwood, E.A.; Glembotski, C.C.; Miyamoto, S.; Westenbrink, B.D.; Brown, J.H. CaMKIIδ subtypes differentially regulate infarct formation following ex vivo myocardial ischemia/reperfusion through NF-κB and TNF-α. J. Mol. Cell. Cardiol. 2017, 103, 48–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Sun, P.; Enslen, H.; Myung, P.S.; Maurer, R.A. Differential activation of CREB by Ca2+/calmodulin-dependent protein kinases type II and type IV involves phosphorylation of a site that negatively regulates activity. Genes Dev. 1994, 8, 2527–2539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Cruzalegui, F.H.; Means, A.R. Biochemical characterization of the multifunctional Ca2+/calmodulin-dependent protein kinase type IV expressed in insect cells. J. Biol. Chem. 1993, 268, 26171–26178. [Google Scholar] [CrossRef]
  62. Zhang, H.; Kong, Q.; Wang, J.; Jiang, Y.; Hua, H. Complex roles of cAMP–PKA–CREB signaling in cancer. Exp. Hematol. Oncol. 2020, 9, 1–13. [Google Scholar] [CrossRef]
  63. Sapio, L.; Salzillo, A.; Ragone, A.; Illiano, M.; Spina, A.; Naviglio, S. Targeting CREB in Cancer Therapy: A Key Candidate or One of Many? An Update. Cancers 2020, 12, 3166. [Google Scholar] [CrossRef] [PubMed]
  64. Miranti, C.K.; Ginty, D.D.; Huang, G.; Chatila, T.; Greenberg, M.E. Calcium activates serum response factor-dependent transcription by a Ras- and Elk-1-independent mechanism that involves a Ca2+/calmodulin-dependent kinase. Mol. Cell. Biol. 1995, 15, 3672–3684. [Google Scholar] [CrossRef] [Green Version]
  65. Flück, M.; Booth, F.W.; Waxhamb, M.N. Skeletal Muscle CaMKII Enriches in Nuclei and Phosphorylates Myogenic Factor SRF at Multiple Sites. Biochem. Biophys. Res. Commun. 2000, 270, 488–494. [Google Scholar] [CrossRef]
  66. Gau, D.; Roy, P. SRF’ing and SAP’ing—The role of MRTF proteins in cell migration. J. Cell Sci. 2018, 131, jcs218222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Pipes, G.C.T.; Creemers, E.E.; Olson, E.N. The myocardin family of transcriptional coactivators: Versatile regulators of cell growth, migration, and myogenesis. Genes Dev. 2006, 20, 1545–1556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Peng, W.; Zhang, Y.; Zheng, M.; Cheng, H.; Zhu, W.; Cao, C.-M.; Xiao, R.-P. Cardioprotection by CaMKII-δB Is Mediated by Phosphorylation of Heat Shock Factor 1 and Subsequent Expression of Inducible Heat Shock Protein 70. Circ. Res. 2010, 106, 102–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Holmberg, C.I.; Hietakangas, V.; Mikhailov, A.; Rantanen, J.O.; Kallio, M.; Meinander, A.; Hellman, J.; Morrice, N.; Mackintosh, C.; Morimoto, R.I.; et al. Phosphorylation of serine 230 promotes inducible transcriptional activity of heat shock factor 1. EMBO J. 2001, 20, 3800–3810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Dong, B.; Jaeger, A.M.; Thiele, D.J. Inhibiting Heat Shock Factor 1 in Cancer: A Unique Therapeutic Opportunity. Trends Pharmacol. Sci. 2019, 40, 986–1005. [Google Scholar] [CrossRef] [PubMed]
  71. Puustinen, M.C.; Sistonen, L. Molecular Mechanisms of Heat Shock Factors in Cancer. Cells 2020, 9, 1202. [Google Scholar] [CrossRef] [PubMed]
  72. Pognonec, P.; Boulukos, K.E.; Gesquière, J.C.; Stéhelin, D.; Ghysdael, J. Mitogenic stimulation of thymocytes results in the calcium-dependent phosphorylation of c-ets-1 proteins. EMBO J. 1988, 7, 977–983. [Google Scholar] [CrossRef]
  73. Cowley, D.O.; Graves, B.J. Phosphorylation represses Ets-1 DNA binding by reinforcing autoinhibition. Genome Res. 2000, 14, 366–376. [Google Scholar]
  74. Dittmer, J. The role of the transcription factor Ets1 in carcinoma. Semin. Cancer Biol. 2015, 35, 20–38. [Google Scholar] [CrossRef]
  75. Davis, F.J.; Gupta, M.; Camoretti-Mercado, B.; Schwartz, R.J.; Gupta, M.P. Calcium/Calmodulin-dependent Protein Kinase Activates Serum Response Factor Transcription Activity by Its Dissociation from Histone Deacetylase, HDAC4. J. Biol. Chem. 2003, 278, 20047–20058. [Google Scholar] [CrossRef] [Green Version]
  76. MacDonnell, S.M.; Weisser-Thomas, J.; Kubo, H.; Hanscome, M.; Liu, Q.; Jaleel, N.; Berretta, R.; Chen, X.; Brown, J.H.; Sabri, A.-K.; et al. CaMKII Negatively Regulates Calcineurin–NFAT Signaling in Cardiac Myocytes. Circ. Res. 2009, 105, 316–325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Kreusser, M.M.; Lehmann, L.H.; Keranov, S.; Hoting, M.-O.; Oehl, U.; Kohlhaas, M.; Reil, J.-C.; Neumann, K.; Schneider, M.D.; Hill, J.A.; et al. Cardiac CaM Kinase II Genes δ and γ Contribute to Adverse Remodeling but Redundantly Inhibit Calcineurin-Induced Myocardial Hypertrophy. Circulation 2014, 130, 1262–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Van Oort, R.J.; Van Rooij, E.; Bourajjaj, M.; Schimmel, J.; Jansen, M.A.; Van Der Nagel, R.; Doevendans, P.A.; Schneider, M.D.; Van Echteld, C.J.A.; De Windt, L.J. MEF2 Activates a Genetic Program Promoting Chamber Dilation and Contractile Dysfunction in Calcineurin-Induced Heart Failure. Circulation 2006, 114, 298–308. [Google Scholar] [CrossRef] [Green Version]
  79. Macian, F. NFAT proteins: Key regulators of T-cell development and function. Nat. Rev. Immunol. 2005, 5, 472–484. [Google Scholar] [CrossRef]
  80. Müller, M.R.; Rao, A. NFAT, immunity and cancer: A transcription factor comes of age. Nat. Rev. Immunol. 2010, 10, 645–656. [Google Scholar] [CrossRef]
  81. Jain, J.; McCafffrey, P.G.; Miner, Z.; Kerppola, T.K.; Lambert, J.N.; Verdine, G.L.; Curran, T.; Rao, A. The T-cell transcription factor NFATp is a substrate for calcineurin and interacts with Fos and Jun. Nature 1993, 365, 352–355. [Google Scholar] [CrossRef]
  82. Rao, A.; Luo, C.; Hogan, P.G. Transcription factors of the NFAT family: Regulation and Function. Annu. Rev. Immunol. 1997, 15, 707–747. [Google Scholar] [CrossRef] [PubMed]
  83. Chen, N.M.; Singh, G.; Koenig, A.; Liou, G.Y.; Storz, P.; Zhang, J.S.; Regul, L.; Nagarajan, S.; Kühnemuth, B.; Johnsen, S.A.; et al. NFATc1 links EGFR signaling to induction of sox9 transcription and acinar-ductal transdifferentiation in the pan-creas. Gastroenterology 2015, 148, 1024–1034. [Google Scholar] [CrossRef] [Green Version]
  84. Chen, N.-M.; Neesse, A.; Dyck, M.L.; Steuber, B.; Koenig, A.O.; Lubeseder-Martellato, C.; Winter, T.; Forster, T.; Bohnenberger, H.; Kitz, J.; et al. Context-Dependent Epigenetic Regulation of Nuclear Factor of Activated T Cells 1 in Pancreatic Plasticity. Gastroenterology 2017, 152, 1507–1520.e15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Hessmann, E.; Zhang, J.-S.; Chen, N.-M.; Hasselluhn, M.; Liou, G.-Y.; Storz, P.; Ellenrieder, V.; Billadeau, D.D.; Koenig, A. NFATc4 RegulatesSox9Gene Expression in Acinar Cell Plasticity and Pancreatic Cancer Initiation. Stem Cells Int. 2016, 2016, 1–11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Baumgart, S.; Chen, N.-M.; Siveke, J.T.; König, A.; Zhang, J.-S.; Singh, S.K.; Wolf, E.; Bartkuhn, M.; Esposito, I.; Heßmann, E.; et al. Inflammation-Induced NFATc1–STAT3 Transcription Complex Promotes Pancreatic Cancer Initiation by KrasG12D. Cancer Discov. 2014, 4, 688–701. [Google Scholar] [CrossRef] [Green Version]
  87. Baumgart, S.; Glesel, E.; Singh, G.; Chen, N.; Reutlinger, K.; Zhang, J.; Billadeau, D.D.; Fernandez-Zapico, M.E.; Gress, T.M.; Singh, S.K.; et al. Restricted Heterochromatin Formation Links NFATc2 Repressor Activity With Growth Promotion in Pancreatic Cancer. Gastroenterology 2012, 142, 388–398.e7. [Google Scholar] [CrossRef] [Green Version]
  88. Singh, G.; Singh, S.K.; König, A.; Reutlinger, K.; Nye, M.D.; Adhikary, T.; Eilers, M.; Gress, T.M.; Fernandez-Zapico, M.E.; Ellenrieder, V. Sequential Activation of NFAT and c-Myc Transcription Factors Mediates the TGF-β Switch from a Suppressor to a Promoter of Cancer Cell Proliferation. J. Biol. Chem. 2010, 285, 27241–27250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Hasselluhn, M.C.; Schmidt, G.E.; Ellenrieder, V.; Johnsen, S.A.; Hessmann, E. Aberrant NFATc1 signaling counteracts TGFβ-mediated growth arrest and apoptosis induction in pancreatic cancer progression. Cell Death Dis. 2019, 10, 446. [Google Scholar] [CrossRef] [Green Version]
  90. Köenig, A.; Linhart, T.; Schlengemann, K.; Reutlinger, K.; Wegele, J.; Adler, G.; Singh, G.; Hofmann, L.; Kunsch, S.; Büch, T.; et al. NFAT-Induced Histone Acetylation Relay Switch Promotes c-Myc-Dependent Growth in Pancreatic Cancer Cells. Gastroenterology 2010, 138, 1189–1199.e2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Singh, S.K.; Jochen, G.; Hessmann, E.; Siveke, J.; Lahmann, M.; Singh, G.; Voelker, N.; Vogt, S.; Esposito, I.; Schmidt, A.; et al. Antithetical NFAT c1–Sox2 and p53–miR200 signaling networks govern pancreatic cancer cell plasticity. EMBO J. 2015, 34, 517–530. [Google Scholar] [CrossRef]
  92. Ma, Y.; Hendershot, L.M. The role of the unfolded protein response in tumour development: Friend or foe? Nat. Rev. Cancer 2004, 4, 966–977. [Google Scholar] [CrossRef] [PubMed]
  93. Zhang, Z.; Zhang, L.; Zhou, L.; Lei, Y.; Zhang, Y.; Huang, C. Redox signaling and unfolded protein response coordinate cell fate decisions under ER stress. Redox Biol. 2019, 25, 101047. [Google Scholar] [CrossRef] [PubMed]
  94. Da Conceicao, V.N.; Sun, Y.; Zboril, E.K.; De La Chapa, J.J.; Singh, B.B. Loss of Ca2+ entry via Orai-TRPC1 induces ER stress, initiating immune activation in macrophages. J. Cell Sci. 2019, 133, jcs237610. [Google Scholar] [CrossRef] [Green Version]
  95. Kono, T.; Tong, X.; Taleb, S.; Bone, R.N.; Iida, H.; Lee, C.-C.; Sohn, P.; Gilon, P.; Roe, M.W.; Evans-Molina, C. Impaired Store-Operated Calcium Entry and STIM1 Loss Lead to Reduced Insulin Secretion and Increased Endoplasmic Reticulum Stress in the Diabetic β-Cell. Diabetes 2018, 67, 2293–2304. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Flourakis, M.; Lehen’Kyi, V.; Beck, B.; Raphael, M.; Vandenberghe, M.; Van Denabeele, F.; 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]
  97. Eletto, D.; Chevet, E.; Argon, Y.; Appenzeller-Herzog, C. Redox controls UPR to control redox. J. Cell Sci. 2014, 127, 3649–3658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Zhang, X.; Gibhardt, C.S.; Will, T.; Stanisz, H.; Körbel, C.; Mitkovski, M.; Stejerean, I.; Cappello, S.; Pacheu-Grau, D.; Dudek, J.; et al. Redox signals at the ER–mitochondria interface control melanoma progression. EMBO J. 2019, 38, e100871. [Google Scholar] [CrossRef]
  99. Lièvremont, J.-P.; Rizzuto, R.; Hendershot, L.; Meldolesi, J. BiP, a Major Chaperone Protein of the Endoplasmic Reticulum Lumen, Plays a Direct and Important Role in the Storage of the Rapidly Exchanging Pool of Ca2+. J. Biol. Chem. 1997, 272, 30873–30879. [Google Scholar] [CrossRef] [Green Version]
  100. Giraldo, A.M.V.; Medus, M.L.; Lebrero, M.G.; Pagano, R.S.; Labriola, C.A.; Landolfo, L.; Delfino, J.M.; Parodi, A.J.; Caramelo, J.J. The Structure of Calreticulin C-terminal Domain Is Modulated by Physiological Variations of Calcium Concentration. J. Biol. Chem. 2010, 285, 4544–4553. [Google Scholar] [CrossRef] [Green Version]
  101. Smith, G.L.; Eisner, D.A. Calcium Buffering in the Heart in Health and Disease. Circulation 2019, 139, 2358–2371. [Google Scholar] [CrossRef] [PubMed]
  102. Ivessa, N.E.; De Lemos-Chiarandini, C.; Gravotta, D.; Sabatini, D.D.; Kreibich, G. The Brefeldin A-induced Retrograde Transport from the Golgi Apparatus to the Endoplasmic Reticulum Depends on Calcium Sequestered to Intracellular Stores. J. Biol. Chem. 1995, 270, 25960–25967. [Google Scholar] [CrossRef] [Green Version]
  103. Vassilakos, A.; Michalak, M.; Lehrman, M.A.; Williams, D.B. Oligosaccharide Binding Characteristics of the Molecular Chaperones Calnexin and Calreticulin. Biochemistry 1998, 37, 3480–3490. [Google Scholar] [CrossRef] [PubMed]
  104. Prins, D.; Michalak, M. Organellar Calcium Buffers. Cold Spring Harb. Perspect. Biol. 2011, 3, a004069. [Google Scholar] [CrossRef] [Green Version]
  105. Bertolotti, A.; Zhang, Y.; Hendershot, L.M.; Harding, H.P.; Ron, D. Dynamic interaction of BiP and ER stress transducers in the unfolded-protein response. Nat. Cell Biol. 2000, 2, 326–332. [Google Scholar] [CrossRef] [PubMed]
  106. Kopp, M.C.; Larburu, N.; Durairaj, V.; Adams, C.J.; Ali, M.M.U. UPR proteins IRE1 and PERK switch BiP from chaperone to ER stress sensor. Nat. Struct. Mol. Biol. 2019, 26, 1053–1062. [Google Scholar] [CrossRef]
  107. Hillary, R.F.; Fitzgerald, U. A lifetime of stress: ATF6 in development and homeostasis. J. Biomed. Sci. 2018, 25, 1–10. [Google Scholar] [CrossRef] [Green Version]
  108. Vekich, J.A.; Belmont, P.J.; Thuerauf, D.J.; Glembotski, C.C. Protein disulfide isomerase-associated 6 is an ATF6-inducible ER stress response protein that protects cardiac myocytes from ischemia/reperfusion-mediated cell death. J. Mol. Cell. Cardiol. 2012, 53, 259–267. [Google Scholar] [CrossRef] [Green Version]
  109. Hetz, C. The unfolded protein response: Controlling cell fate decisions under ER stress and beyond. Nat. Rev. Mol. Cell Biol. 2012, 13, 89–102. [Google Scholar] [CrossRef]
  110. Maas, N.L.; Diehl, J.A. Molecular Pathways: The PERKs and Pitfalls of Targeting the Unfolded Protein Response in Cancer. Clin. Cancer Res. 2015, 21, 675–679. [Google Scholar] [CrossRef] [Green Version]
  111. Pakos-Zebrucka, K.; Koryga, I.; Mnich, K.; Ljujic, M.; Samali, A.; Gorman, A.M. The integrated stress response. EMBO Rep. 2016, 17, 1374–1395. [Google Scholar] [CrossRef] [Green Version]
  112. Haze, K.; Yoshida, H.; Yanagi, H.; Yura, T.; Mori, K. Mammalian Transcription Factor ATF6 Is Synthesized as a Transmembrane Protein and Activated by Proteolysis in Response to Endoplasmic Reticulum Stress. Mol. Biol. Cell 1999, 10, 3787–3799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Ye, J.; Rawson, R.B.; Komuro, R.; Chen, X.; Davé, U.P.; Prywes, R.; Brown, M.S.; Goldstein, J.L. ER Stress Induces Cleavage of Membrane-Bound ATF6 by the Same Proteases that Process SREBPs. Mol. Cell 2000, 6, 1355–1364. [Google Scholar] [CrossRef]
  114. Schindler, A.J.; Schekman, R. In vitro reconstitution of ER-stress induced ATF6 transport in COPII vesicles. Proc. Natl. Acad. Sci. USA 2009, 106, 17775–17780. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Urra, H.; Dufey, E.; Avril, T.; Chevet, E.; Hetz, C. Endoplasmic Reticulum Stress and the Hallmarks of Cancer. Trends Cancer 2016, 2, 252–262. [Google Scholar] [CrossRef] [PubMed]
  116. Chen, X.; Cubillos-Ruiz, J.R. Endoplasmic reticulum stress signals in the tumour and its microenvironment. Nat. Rev. Cancer 2021, 21, 71–88. [Google Scholar] [CrossRef]
  117. Xu, C.; Bailly-Maitre, B.; Reed, J.C. Review series Endoplasmic reticulum stress: Cell life and death decisions. J. Clin. Investig. 2005, 115, 2656–2664. [Google Scholar] [CrossRef] [Green Version]
  118. Urano, F.; Wang, X.; Bertolotti, A.; Zhang, Y.; Chung, P.; Harding, H.P.; Ron, D. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 2000, 287, 664–666. [Google Scholar] [CrossRef] [Green Version]
  119. Yoshida, H.; Matsui, T.; Yamamoto, A.; Okada, T.; Mori, K. XBP1 mRNA Is Induced by ATF6 and Spliced by IRE1 in Response to ER Stress to Produce a Highly Active Transcription Factor. Cell 2001, 107, 881–891. [Google Scholar] [CrossRef] [Green Version]
  120. Shamu, C.E.; Walter, P. Oligomerization and phosphorylation of the Ire1p kinase during intracellular signaling from the endoplasmic reticulum to the nucleus. EMBO J. 1996, 15, 3028–3039. [Google Scholar] [CrossRef]
  121. Ali, M.M.U.; Bagratuni, T.; Davenport, E.L.; Nowak, P.R.; Silva-Santisteban, M.C.; Hardcastle, A.; McAndrews, C.; Rowlands, M.G.; Morgan, G.J.; Aherne, W.; et al. Structure of the Ire1 autophosphorylation complex and implications for the unfolded protein response. EMBO J. 2011, 30, 894–905. [Google Scholar] [CrossRef] [Green Version]
  122. Lee, A.-H.; Iwakoshi, N.N.; Glimcher, L.H. XBP-1 Regulates a Subset of Endoplasmic Reticulum Resident Chaperone Genes in the Unfolded Protein Response. Mol. Cell. Biol. 2003, 23, 7448–7459. [Google Scholar] [CrossRef] [Green Version]
  123. Cullinan, S.B.; Zhang, D.; Hannink, M.; Arvisais, E.; Kaufman, R.J.; Diehl, J.A. Nrf2 Is a Direct PERK Substrate and Effector of PERK-Dependent Cell Survival. Mol. Cell. Biol. 2003, 23, 7198–7209. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Mukaigasa, K.; Tsujita, T.; Nguyen, V.T.; Li, L.; Yagi, H.; Fuse, Y.; Nakajima-Takagi, Y.; Kato, K.; Yamamoto, M.; Kobayashi, M. Nrf2 activation attenuates genetic endoplasmic reticulum stress induced by a mutation in the phosphomannomutase 2 gene in zebrafish. Proc. Natl. Acad. Sci. USA 2018, 115, 2758–2763. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Carrara, M.; Prischi, F.; Nowak, P.R.; Ali, M.M. Crystal structures reveal transient PERK luminal domain tetramerization in endoplasmic reticulum stress signaling. EMBO J. 2015, 34, 1589–1600. [Google Scholar] [CrossRef] [PubMed]
  126. Harding, H.P.; Zhang, Y.; Scheuner, D.; Chen, J.-J.; Kaufman, R.J.; Ron, D. Ppp1r15 gene knockout reveals an essential role for translation initiation factor 2 alpha (eIF2α) dephosphorylation in mammalian development. Proc. Natl. Acad. Sci. USA 2009, 106, 1832–1837. [Google Scholar] [CrossRef] [Green Version]
  127. Wang, P.; Li, J.; Tao, J.; Sha, B. The luminal domain of the ER stress sensor protein PERK binds misfolded proteins and thereby triggers PERK oligomerization. J. Biol. Chem. 2018, 293, 4110–4121. [Google Scholar] [CrossRef] [Green Version]
  128. Hayes, J.D.; Dinkova-Kostova, A.T. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem. Sci. 2014, 39, 199–218. [Google Scholar] [CrossRef]
  129. Rouschop, K.M.A.; Beucken, T.V.D.; Dubois, L.; Niessen, H.; Bussink, J.; Savelkouls, K.; Keulers, T.; Mujcic, H.; Landuyt, W.; Voncken, J.W.; et al. The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5. J. Clin. Investig. 2010, 120, 127–141. [Google Scholar] [CrossRef]
  130. B’Chir, W.; Maurin, A.-C.; Carraro, V.; Averous, J.; Jousse, C.; Muranishi, Y.; Parry, L.; Stepien, G.; Fafournoux, P.; Bruhat, A. The eIF2α/ATF4 pathway is essential for stress-induced autophagy gene expression. Nucleic Acids Res. 2013, 41, 7683–7699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Kroemer, G.; Mariño, G.; Levine, B. Autophagy and the Integrated Stress Response. Mol. Cell 2010, 40, 280–293. [Google Scholar] [CrossRef] [Green Version]
  132. Ye, J.; Kumanova, M.; Hart, L.S.; Sloane, K.; Zhang, H.; De Panis, D.N.; Bobrovnikova-Marjon, E.; Diehl, J.A.; Ron, D.; Koumenis, C. The GCN2-ATF4 pathway is critical for tumour cell survival and proliferation in response to nutrient deprivation. EMBO J. 2010, 29, 2082–2096. [Google Scholar] [CrossRef] [Green Version]
  133. Galehdar, Z.; Swan, P.; Fuerth, B.; Callaghan, S.M.; Park, D.S.; Cregan, S.P. Neuronal Apoptosis Induced by Endoplasmic Reticulum Stress Is Regulated by ATF4-CHOP-Mediated Induction of the Bcl-2 Homology 3-Only Member PUMA. J. Neurosci. 2010, 30, 16938–16948. [Google Scholar] [CrossRef]
  134. Puthalakath, H.; O’Reilly, L.A.; Gunn, P.; Lee, L.; Kelly, P.N.; Huntington, N.D.; Hughes, P.D.; Michalak, E.M.; McKimm-Breschkin, J.; Motoyama, N.; et al. ER Stress Triggers Apoptosis by Activating BH3-Only Protein Bim. Cell 2007, 129, 1337–1349. [Google Scholar] [CrossRef] [Green Version]
  135. Ohoka, N.; Yoshii, S.; Hattori, T.; Onozaki, K.; Hayashi, H. TRB3, a novel ER stress-inducible gene, is induced via ATF4–CHOP pathway and is involved in cell death. EMBO J. 2005, 24, 1243–1255. [Google Scholar] [CrossRef] [PubMed]
  136. Salazar, M.; Lorente, M.; García-Taboada, E.; Pérez-Gómez, E.; Dávila, D.; Zúñiga-García, P.; Flores, J.M.; Rodríguez, A.; Hegedus, Z.; Mosén-Ansorena, D.; et al. Loss of Tribbles pseudokinase-3 promotes Akt-driven tumorigenesis via FOXO inactivation. Cell Death Differ. 2014, 22, 131–144. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Marciniak, S.J.; Yun, C.Y.; Oyadomari, S.; Novoa, I.; Zhang, Y.; Jungreis, R.; Nagata, K.; Harding, H.P.; Ron, D. CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev. 2004, 18, 3066–3077. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Bi, M.; Naczki, C.; Koritzinsky, M.; Fels, D.; Blais, J.; Hu, N.; Harding, H.; Novoa, I.; Varia, M.; Raleigh, J.; et al. ER stress-regulated translation increases tolerance to extreme hypoxia and promotes tumor growth. EMBO J. 2005, 24, 3470–3481. [Google Scholar] [CrossRef] [Green Version]
  139. Moore, P.C.; Qi, J.Y.; Thamsen, M.; Ghosh, R.; Peng, J.; Gliedt, M.J.; Meza-Acevedo, R.; Warren, R.E.; Hiniker, A.; Kim, G.E.; et al. Parallel Signaling through IRE1α and PERK Regulates Pancreatic Neuroendocrine Tumor Growth and Survival. Cancer Res. 2019, 79, 6190–6203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Terai, H.; Kitajima, S.; Potter, D.S.; Matsui, Y.; Quiceno, L.G.; Chen, T.; Kim, T.-J.; Rusan, M.; Thai, T.C.; Piccioni, F.; et al. ER Stress Signaling Promotes the Survival of Cancer “Persister Cells” Tolerant to EGFR Tyrosine Kinase Inhibitors. Cancer Res. 2017, 78, 1044–1057. [Google Scholar] [CrossRef] [Green Version]
  141. Dufey, E.; Urra, H.; Hetz, C. ER proteostasis addiction in cancer biology: Novel concepts. Semin. Cancer Biol. 2015, 33, 40–47. [Google Scholar] [CrossRef]
  142. Wang, Y.; Alam, G.N.; Ning, Y.; Visioli, F.; Dong, Z.; Nör, J.E.; Polverini, P.J. The Unfolded Protein Response Induces the Angiogenic Switch in Human Tumor Cells through the PERK/ATF4 Pathway. Cancer Res. 2012, 72, 5396–5406. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Pereira, E.R.; Frudd, K.; Awad, W.; Hendershot, L.M. Endoplasmic Reticulum (ER) Stress and Hypoxia Response Pathways Interact to Potentiate Hypoxia-inducible Factor 1 (HIF-1) Transcriptional Activity on Targets Like Vascular Endothelial Growth Factor (VEGF). J. Biol. Chem. 2014, 289, 3352–3364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. 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] [PubMed] [Green Version]
  145. Yuan, G.; Nanduri, J.; Bhasker, C.R.; Semenza, G.L.; Prabhakar, N.R. Ca2+/Calmodulin Kinase-dependent Activation of Hypoxia Inducible Factor 1 Transcriptional Activity in Cells Subjected to Intermittent Hypoxia. J. Biol. Chem. 2005, 280, 4321–4328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  146. Jian, W.; Junling, S.; Kaili, Z.; Jinmeng, H.; Jiuxing, D.; Jianwei, S.; Wang, J.; Shen, J.; Zhao, K.; Hu, J.; et al. STIM1 overexpression in hypoxia microenvironment contributes to pancreatic carcinoma progression. Cancer Biol. Med. 2019, 16, 100–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Azzi, J.R.; Sayegh, M.H.; Mallat, S.G. Calcineurin Inhibitors: 40 Years Later, Can’t Live Without…. J. Immunol. 2013, 191, 5785–5791. [Google Scholar] [CrossRef] [Green Version]
  148. Kaufman, D.B.; Shapiro, R.; Lucey, M.R.; Cherikh, W.S.; Bustami, R.T.; Dyke, D.B. Immunosuppression: Practice and trends. Arab. Archaeol. Epigr. 2004, 4, 38–53. [Google Scholar] [CrossRef]
  149. Kawahara, T.; Kashiwagi, E.; Ide, H.; Li, Y.; Zheng, Y.; Miyamoto, Y.; Netto, G.J.; Ishiguro, H.; Miyamoto, H. Cyclosporine A and tacrolimus inhibit bladder cancer growth through down-regulation of NFATc1. Oncotarget 2015, 6, 1582–1593. [Google Scholar] [CrossRef] [Green Version]
  150. Siamakpour-Reihani, S.; Caster, J.; Nepal, D.B.; Courtwright, A.; Hilliard, E.; Usary, J.; Ketelsen, D.; Darr, D.; Shen, X.J.; Patterson, C.; et al. The Role of Calcineurin/NFAT in SFRP2 Induced Angiogenesis—A Rationale for Breast Cancer Treatment with the Calcineurin Inhibitor Tacrolimus. PLoS ONE 2011, 6, e20412. [Google Scholar] [CrossRef] [Green Version]
  151. Mancini, M.; Toker, A. NFAT proteins: Emerging roles in cancer progression. Nat. Rev. Cancer 2009, 9, 810–820. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Dantal, J.; Soulillou, J.-P. Immunosuppressive Drugs and the Risk of Cancer after Organ Transplantation. N. Engl. J. Med. 2005, 352, 1371–1373. [Google Scholar] [CrossRef]
  153. Rezzani, R. Cyclosporine A and adverse effects on organs: Histochemical studies. Prog. Histochem. Cytochem. 2004, 39, 85–128. [Google Scholar] [CrossRef] [PubMed]
  154. Khan, H.Y.; Mpilla, G.B.; Sexton, R.; Viswanadha, S.; Penmetsa, K.V.; Aboukameel, A.; Diab, M.; Kamgar, M.; Al-Hallak, M.N.; Szlaczky, M.; et al. Calcium Release-Activated Calcium (CRAC) Channel Inhibition Suppresses Pancreatic Ductal Adenocarcinoma Cell Proliferation and Patient-Derived Tumor Growth. Cancers 2020, 12, 750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Tameire, F.; Verginadis, I.I.; Leli, N.M.; Polte, C.; Conn, C.S.; Ojha, R.; Salinas, C.S.; Chinga, F.; Monroy, A.M.; Fu, W.; et al. ATF4 couples MYC-dependent translational activity to bioenergetic demands during tumour progression. Nat. Cell Biol. 2019, 21, 889–899. [Google Scholar] [CrossRef]
  156. NCT03709342 A PK/PD Study of CM4620-IE in Patients with Acute Pancreatitis. Available online: https://clinicaltrials.gov/ct2/show/NCT03709342 (accessed on 4 March 2021).
  157. NCT03401190 CM4620 Injectable Emulsion Versus Supportive Care in Patients with Acute Pancreatitis and SIRS. Available online: https://clinicaltrials.gov/ct2/show/NCT03401190 (accessed on 4 March 2021).
  158. NCT04195347 Study of CM4620 to Reduce the Severity of Pancreatitis Due to Asparaginase. Available online: https://clinicaltrials.gov/ct2/show/NCT04195347 (accessed on 4 March 2021).
  159. NCT04345614 A Study of Auxora in Patients with Severe COVID-19 Pneumonia. Available online: https://clinicaltrials.gov/ct2/show/NCT04345614 (accessed on 4 March 2021).
  160. NCT04661540 A Study of Auxora in Patients with Critical COVID-19 Pneumonia. Available online: https://clinicaltrials.gov/ct2/show/NCT04661540 (accessed on 4 March 2021).
  161. NCT04681066 A Study of Auxora in Patients with Acute Pancreatitis and Accompanying SIRS (CARPO). Available online: https://clinicaltrials.gov/ct2/show/NCT04681066 (accessed on 4 March 2021).
  162. Wen, L.; Voronina, S.; Javed, M.A.; Awais, M.; Szatmary, P.; Latawiec, D.; Chvanov, M.; Collier, D.; Huang, W.; Barrett, J.; et al. Inhibitors of ORAI1 Prevent Cytosolic Calcium-Associated Injury of Human Pancreatic Acinar Cells and Acute Pancreatitis in 3 Mouse Models. Gastroenterology 2015, 149, 481–492.e7. [Google Scholar] [CrossRef] [Green Version]
  163. Gerasimenko, J.V.; Gryshchenko, O.; Ferdek, P.E.; Stapleton, E.; Hébert, T.O.G.; Bychkova, S.; Peng, S.; Begg, M.; Gerasimenko, O.V.; Petersen, O.H. Ca2+ release-activated Ca2+ channel blockade as a potential tool in antipancreatitis therapy. Proc. Natl. Acad. Sci. USA 2013, 110, 13186–13191. [Google Scholar] [CrossRef] [Green Version]
  164. Soboloff, J.; Rothberg, B.S.; Madesh, M.; Gill, D.L. STIM proteins: Dynamic calcium signal transducers. Nat. Rev. Mol. Cell Biol. 2012, 13, 549–565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Calcium-dependent signaling and transcription factors. Upon a stimulus, GPCRs or RTKs are activated, phosphorylating PLC and promoting the breakdown of PIP2 to DAG and IP3. The latter promotes the opening of the channel IP3R, raising cytosolic calcium levels. STIM and ORAI further promote the influx of calcium into the cytoplasm through SOCE. DAG and calcium activate PKC and consequently the MAPK pathway, fostering AP1 activity. SERCA pumps calcium from the cytoplasm into the ER negatively regulating PKC. CaM binds calcium, activating CaN and CAMKII. CaN dephosphorylates NFATs, promoting its translocation into the nucleus, where it can dimerize with AP1, among other factors, to activate target genes. In the cytosol, CAMKII activates NFκB by phosphorylating IKK2. CAMKII further activates HSF1, while repressing ETS1. CREB1 can be phosphorylated by CAMKII at two sites, one repressive (Ser142) and one activating (Ser133). CAMKII inhibits HDAC4, promoting the activation of MEF2 and SRF. SOCE inhibitors can be employed to target STIM1 or ORAI1 and consequently calcium-dependent programs, while calcineurin inhibitors block NFAT activation. In yellow are calcium pumps, in blue calcium channels and in green the components of SOCE, STIM and ORAI. Concerning signaling components, receptors are in purple and their downstream effectors in pink/red. Factors activated by CaN in green and factors activated and/or repressed by CAMKII in blue. GPCR: G protein-coupled receptor; RTK: receptor tyrosine kinase; PLC: phospholipase C; PIP2: phosphatidylinositol 4,5-bisphosphate; DAG: diacylglycerol; IP3: inositol 1,4,5-triphosphate; IP3R: IP3 receptor; STIM: stromal interaction molecule; ORAI: calcium release-activated calcium channel; SOCE: store-operated calcium entry; PKC: protein kinase C; MAPK: mitogen-activated protein kinase; AP1: activator protein 1; SERCA: sarco/endoplasmic reticulum calcium-ATPase; ER: endoplasmic reticulum; CaM: calmodulin; CaN: calcineurin; CAMKII: calcium/calmodulin-dependent protein kinase II; NFAT: nuclear factor of activated T cells; NFκB: nuclear factor-kappa B; IKK2: inhibitor of nuclear-factor kappa B kinase subunit beta; HSF1: heat shock factor 1; ETS1: c-ets-1; CREB1: cAMP response element-binding protein 1; HDAC4: histone deacetylase 4; MEF2: myocyte enhancer factor 2; SRF: serum response factor.
Figure 1. Calcium-dependent signaling and transcription factors. Upon a stimulus, GPCRs or RTKs are activated, phosphorylating PLC and promoting the breakdown of PIP2 to DAG and IP3. The latter promotes the opening of the channel IP3R, raising cytosolic calcium levels. STIM and ORAI further promote the influx of calcium into the cytoplasm through SOCE. DAG and calcium activate PKC and consequently the MAPK pathway, fostering AP1 activity. SERCA pumps calcium from the cytoplasm into the ER negatively regulating PKC. CaM binds calcium, activating CaN and CAMKII. CaN dephosphorylates NFATs, promoting its translocation into the nucleus, where it can dimerize with AP1, among other factors, to activate target genes. In the cytosol, CAMKII activates NFκB by phosphorylating IKK2. CAMKII further activates HSF1, while repressing ETS1. CREB1 can be phosphorylated by CAMKII at two sites, one repressive (Ser142) and one activating (Ser133). CAMKII inhibits HDAC4, promoting the activation of MEF2 and SRF. SOCE inhibitors can be employed to target STIM1 or ORAI1 and consequently calcium-dependent programs, while calcineurin inhibitors block NFAT activation. In yellow are calcium pumps, in blue calcium channels and in green the components of SOCE, STIM and ORAI. Concerning signaling components, receptors are in purple and their downstream effectors in pink/red. Factors activated by CaN in green and factors activated and/or repressed by CAMKII in blue. GPCR: G protein-coupled receptor; RTK: receptor tyrosine kinase; PLC: phospholipase C; PIP2: phosphatidylinositol 4,5-bisphosphate; DAG: diacylglycerol; IP3: inositol 1,4,5-triphosphate; IP3R: IP3 receptor; STIM: stromal interaction molecule; ORAI: calcium release-activated calcium channel; SOCE: store-operated calcium entry; PKC: protein kinase C; MAPK: mitogen-activated protein kinase; AP1: activator protein 1; SERCA: sarco/endoplasmic reticulum calcium-ATPase; ER: endoplasmic reticulum; CaM: calmodulin; CaN: calcineurin; CAMKII: calcium/calmodulin-dependent protein kinase II; NFAT: nuclear factor of activated T cells; NFκB: nuclear factor-kappa B; IKK2: inhibitor of nuclear-factor kappa B kinase subunit beta; HSF1: heat shock factor 1; ETS1: c-ets-1; CREB1: cAMP response element-binding protein 1; HDAC4: histone deacetylase 4; MEF2: myocyte enhancer factor 2; SRF: serum response factor.
Cells 10 00966 g001
Figure 2. ER stress and its calcium dependency. During resting conditions, GRP78 inhibits the activation of ER stress mediators, ATF6, IRE1 and PERK, which in turn hinder the ATPase activity of GRP78. Furthermore, GRP78 buffers 25% of ER calcium and calcium levels highly modulate the protein folding activity of GRP78 and other protein chaperones. Upon stress, GRP78 dissociates from ER stress mediators, leading to their activation and triggering the ER stress response. GRP78: Heat Shock Protein Family A (Hsp70) Member 5; ER: endoplasmic reticulum; ATF6: Activating Transcription Factor 6; IRE1: inositol requiring enzyme 1; PERK: PKR-like ER kinase.
Figure 2. ER stress and its calcium dependency. During resting conditions, GRP78 inhibits the activation of ER stress mediators, ATF6, IRE1 and PERK, which in turn hinder the ATPase activity of GRP78. Furthermore, GRP78 buffers 25% of ER calcium and calcium levels highly modulate the protein folding activity of GRP78 and other protein chaperones. Upon stress, GRP78 dissociates from ER stress mediators, leading to their activation and triggering the ER stress response. GRP78: Heat Shock Protein Family A (Hsp70) Member 5; ER: endoplasmic reticulum; ATF6: Activating Transcription Factor 6; IRE1: inositol requiring enzyme 1; PERK: PKR-like ER kinase.
Cells 10 00966 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kutschat, A.P.; Johnsen, S.A.; Hamdan, F.H. Store-Operated Calcium Entry: Shaping the Transcriptional and Epigenetic Landscape in Pancreatic Cancer. Cells 2021, 10, 966. https://doi.org/10.3390/cells10050966

AMA Style

Kutschat AP, Johnsen SA, Hamdan FH. Store-Operated Calcium Entry: Shaping the Transcriptional and Epigenetic Landscape in Pancreatic Cancer. Cells. 2021; 10(5):966. https://doi.org/10.3390/cells10050966

Chicago/Turabian Style

Kutschat, Ana P., Steven A. Johnsen, and Feda H. Hamdan. 2021. "Store-Operated Calcium Entry: Shaping the Transcriptional and Epigenetic Landscape in Pancreatic Cancer" Cells 10, no. 5: 966. https://doi.org/10.3390/cells10050966

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