Next Article in Journal
Hierarchical Voting-Based Feature Selection and Ensemble Learning Model Scheme for Glioma Grading with Clinical and Molecular Characteristics
Next Article in Special Issue
Novel p.G250A Mutation Associated with Chronic Pancreatitis Highlights Misfolding-Prone Region in Carboxypeptidase A1 (CPA1)
Previous Article in Journal
Pathogenesis and Preventive Tactics of Immune-Mediated Non-Pulmonary COVID-19 in Children and Beyond
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Glycogen Synthase Kinase 3β: A True Foe in Pancreatic Cancer

by
Omer H. M. Elmadbouh
1,
Stephen J. Pandol
1,2,* and
Mouad Edderkaoui
1,2
1
Department of Medicine, Division of Gastroenterology and Hepatology, Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
2
Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(22), 14133; https://doi.org/10.3390/ijms232214133
Submission received: 20 October 2022 / Accepted: 14 November 2022 / Published: 16 November 2022

Abstract

:
Glycogen synthase kinase 3 beta (GSK-3β) is a serine/threonine protein kinase involved in multiple normal and pathological cell functions, including cell signalling and metabolism. GSK-3β is highly expressed in the onset and progression of multiple cancers with strong involvement in the regulation of proliferation, apoptosis, and chemoresistance. Multiple studies showed pro- and anti-cancer roles of GSK-3β creating confusion about the benefit of targeting GSK-3β for treating cancer. In this mini-review, we focus on the role of GSK-3β in pancreatic cancer. We demonstrate that the proposed anti-cancer roles of GSK-3β are not relevant to pancreatic cancer, and we argue why GSK-3β is, indeed, a very promising therapeutic target in pancreatic cancer.

1. Introduction

Glycogen synthase kinase 3 beta (GSK-3β) is a serine/threonine protein kinase originally identified as a regulator of glycogen synthase activity. GSK3β is involved in glycogen synthesis and multiple signalling pathways linking the protein to disorders ranging from neurodegenerative diseases to cancers, and involving differentiation, proliferation, motility, and apoptosis.
Many studies showed pro- and anti-oncogenic roles of GSK-3β in many cancers including in pancreatic ductal adenocarcinoma (PDAC). The detailed mechanisms of the GSK-3β pathways have been previously discussed and can be found in many interesting reviews [1,2,3,4,5,6,7,8]. Here, we do not discuss the detailed pathways regulated by GSK-3β in cancer, but we focus on discussing the relevance to PDAC of the previously proposed anti-cancer and pro-cancer roles of GSK-3β.

2. Regulation of GSK-3β Activity

The activity of GSK-3β is mainly regulated by phosphorylation at serine 9 which results in its inhibition. This phosphorylation is regulated by protein kinase A (PKA), Akt kinase, ribosomal protein S6 kinase beta-1 (p70S6K), and p90 ribosomal S6 kinase (p90RSK), which inactivate GSK-3β and inhibit its ability to phosphorylate downstream substrates [9]. Other inactivating phosphorylation reactions of GSK-3β include Threonine 390 and Threonine 43 by p38 mitogen-activated protein kinases (p38 MAPK), and extracellular signal-regulated kinases (ERK) respectively [10,11]. Both p38 MAPK and ERK are important proteins in cancer development and progression.
Phosphorylation of Tyrosine 216, by autophosphorylation or by Src tyrosine kinase, activates GSK-3β [12,13]. Other regulators of the Tyr 216 phosphorylation/activation include the Ca2+-dependent tyrosine kinase Pyk2 and the proto-oncogene tyrosine-protein kinase Fyn [14,15]. Regulators of GSK-3β inhibitory and activating phosphorylations are summarized in Figure 1.
Protein phosphatases 1 and 2A (PP1 and PP2A) act as activating factors by dephosphorylating serine 9 to increase GSK-3β stability and activity. Other regulations of GSK-3β activity include subcellular localization and the formation of protein complexes [16].
GSK-3β catalyses the last step of glycogen synthesis and regulates β-catenin, a critical step in the Wnt pathway. These two processes were the first GSK-3β pathways studied in detail. They are also characteristics of aggressive cancers and contribute to the therapeutic hurdle of chemoresistance and high metastatic frequency that continues to maintain survival rates of pancreatic ductal adenocarcinoma (PDAC) patients at low levels.
The role of GSK-3β in the Wnt pathway, initially, caused GSK-3β to appear as if it has played an anti-cancer role for many years. Indeed, many reviews showed detailed roles of GSK-3β in cancer and suggested it as an anti-cancer protein. The detailed pathways regulated by GSK-3β in cancer in general were discussed in detail in many published reviews [1,2,3,4,5,6,7,8]. In this mini-review, we discuss the suggested anti-cancer roles of GSK-3β and question their relevance to PDAC.

3. GSK3-β Expression in Cancer

Overexpression of GSK-3β has been observed in various tumour types, including colon, liver, ovarian, and pancreatic cancers [17,18,19,20]. GSK-3β’s pathological role is marked by upregulation of expression and activity in cancer cells compared to non-neoplastic cells and defined by differential phosphorylation [17]. Increased GSK-3β activity is observed in cancer cells, as well as in metastatic lesions, and is responsible for the inherent chemoresistance observed in pancreatic cancer [17,21].
In addition, cell localization of GSK-3β seems to play an important role in cancer development and promotion. For example, nuclear localization of GSK-3β was demonstrated in 70% of human breast carcinomas and 73% of squamous cell head and neck carcinomas, whereas no detectable expression of nuclear GSK-3β was found in benign breast tissue and in benign salivary gland and other benign head and neck tissues [19]. In pancreatic cancer, a study by Ougolkov et al., showed that nuclear localization of GSK-3β is increased with the loss of differentiation of the cancer cells. In fact, they found that PanINs = 1 lesions, which are pre-cancer lesions, had only cytoplasmic localisation of GSK-3β, whereas well-differentiated cancer cells had increased cytoplasmic expression and moderate nuclear localization, and poorly differentiated cancer cells showed strong nuclear localization and little presence in the cytoplasm [22]. Nuclear localization of GSK-3β is associated with a worse outcome and drug resistance in cancer, and the inhibition of GSK-3β activation decreases its nuclear accumulation [22,23].

4. GSK-3β Proposed Onco-Suppressive Effect in Pancreatic Cancer

One of the first discovered roles of GSK-3β is its regulation of the Wnt/β-catenin signalling pathway. When the Wnt ligand is present, it binds to specific membrane-bound receptors. This binding activates an intracellular signalling cascade resulting in β-catenin stabilization and nuclear localization. In the nucleus, β-catenin associates with members of the TCF/LEF family of transcription factors to regulate the transcription of various Wnt targets, which are responsible for cell growth and motility, and both are important for cancer progression and metastasis.
GSK-3β phosphorylates β-catenin, triggering its degradation, and consequently reducing β-catenin nuclear accumulation and reversing the pro-cancer Wnt/βcatenin pathway [24]. The β-catenin destruction complex is assembled through the interactions between Adenomatous polyposis coli (APC), AXIN, GSK-3β, and Casein kinase 1 alpha (CK1α). Specifically, APC directly binds AXIN via Ser-Ala-Met-Pro (SAMP) repeat sequences and β-catenin via three 15-amino acid repeats and seven 20-amino acid repeats. AXIN interacts with GSK-3β, CK1α, and β-catenin, in addition to its binding to APC. The complex is stabilized via GSK-3β and CK1α-mediated AXIN phosphorylation. CK1α phosphorylates serine 45 of β-catenin, which primes the subsequent, sequential, phosphorylation of Threonine 41, serines 33 and 37 by GSK-3β. After these phosphorylation events, β-catenin is retained in the complex, thereby promoting its recognition by the Beta-transducin repeats-containing proteins (β-TrCP) for polyubiquitination on Lys-19 [25]. Therefore, in this case, inhibition of GSK-3β will lead to activation of β-catenin and its pro-cancer activated pathway leading to cancer promotion.
In contrast to this process, recent studies have shown that inhibition of GSK-3β leads to the stabilization of β-catenin and induces PDAC cell death with β-catenin having no effect on PDAC cell survival [26,27]. Importantly, recently published data show that β-catenin partly mediates the killing effects of GSK-3β pharmacological and molecular inhibitors in K-ras-dependent tumors [28]. Pancreatic cancer cells infected with lentivirus containing Cas9 and guide RNA (gRNA) to β-catenin or scrambled were treated with GSK-3β inhibitor SB-732881-H to test whether β-catenin inhibition will rescue PDAC cells from apoptosis induced by the GSK-3β inhibition. The results showed that in scrambled gRNA-infected cells, GSK-3β inhibition upregulated β-catenin, and induced caspase-3 activation and apoptosis. In contrast, a targeted knockout of β-catenin in cells infected with β-catenin gRNA abrogated GSK-3β inhibition induced apoptosis [28]. Taken together, apoptosis induced by GSK-3β inhibition in PDAC tumors is in part mediated by β-catenin. Interestingly, extended overexpression of β-catenin in various cancer cell lines from different origins induced apoptosis [29]. Based on this data, we can state that the GSK-3β/β-catenin pathway is a pro-cancer pathway in PDAC.
In addition to the GSK-3β/β-catenin pathway, multiple studies indicate that GSK-3β-mediated regulation of the Wnt pathway can modulate downstream signalling pathways independently of β-catenin. One major pathway is the inhibition of mTOR by GSK-3β mediated by phosphorylation of Tuberous sclerois complex 2 (TSC2). This pathway is activated by Wnt/GSK-3β independently of β-catenin [30] and is considered another anti-oncogenic effect of GSK-3β in PDAC as mTOR is a major pro-cancer effector in PDAC [31,32]. However, these findings were obtained in healthy cells including fibroblasts, bone marrow, and kidney cells; while data in cancer cells showed that overexpression of GSK-3β activated mTOR complex-1 (mTORC1) leading to activation of the mTOR pathway, indicating that the GSK-3β/mTOR interaction in cancer cells is different from normal cells [33].
In addition to the Wnt pathway, another proposed onco-suppressive effect of GSK-3β is its phosphorylation of the tumour suppressor protein p53, inducing its activation and transcriptional activity [34]. GSK-3β contributes to mitochondrial p53 apoptotic signalling as inhibition of GSK-3β by lithium or 2-Thio(3-iodobenzyl)-5-(1-pyridyl)-[1,3,4]-oxadiazole blocked cytochrome c release and caspase-3 activation [35]. However, the GSK-3β/p53 onco-suppressive effect loses its importance in many cancers including pancreatic cancer, where up to 76% of PDAC patients have a p53 mutation inducing its loss of function [36].
An important onco-suppressive effect of GSK-3β is that constitutively active GSK-3β was found to induce Snail degradation leading to increased E-cadherin and inhibition of the epithelial to mesenchymal transition (EMT), the driving force behind invasion and metastasis [37]. Inhibition of GSK-3β by lithium results in the upregulation of Snail and downregulation of E-cadherin in vivo [37]. In contradiction with this data, GSK-3β inhibitors such as BIO, TWS119, and LiCl, decreased the expression of mesenchymal markers in different cancer cell lines with a mesenchymal phenotype [38]. Furthermore, pharmacological and molecular inhibitions of GSK-3β reduced the migration ability of cancer cells. This effect is most likely induced by decreasing cancer stem cell-related cell surface marker CD44+/24 as cancer stemness is associated with EMT and migration/invasion [38].
In a different study, GSK-3β was found to down-regulate the c-Myc level by causing its proteasomal degradation, leading to onco-suppressive effects including increased apoptosis and decreased proliferation [39]. However, a study using PDAC cells infected with lentivirus containing Cas9 and guide RNA (gRNA) to c-Myc or scrambled were treated with GSK-3β inhibitor SB-732881-H, to test whether c-Myc inhibition will rescue PDAC cells from apoptosis induced by the GSK-3β inhibition, showed that in scrambled gRNA-infected cells, GSK-3β inhibition induced apoptosis. In contrast, targeted knockout of c-Myc in cells infected with c-Myc gRNA abrogated GSK-3β inhibition induced apoptosis [28]. Taken together, apoptosis induced by GSK-3β inhibition in PDAC tumors is in part mediated by c-Myc, and therefore, the proposed onco-suppressive effect of GSK-3β through inhibition of c-Myc is not relevant to PDAC.
Finally, Ben-Josef et al. showed in 2015 that the GSK-3β expression level is a strong prognosticator in PDAC, independent of other known factors such as tumor (T) stage, nodal status, surgical margins and CA19-9 [40]. More importantly, they found that patient survival was significantly increased in PDAC patients with a high level of GSK-3β compared to patients with a low level of GSK-3β. However, the study included a relatively small number of patients (163 patients) and did not associate survival with the activity of GSK-3β or localization (nuclear vs. cytoplasmic) [40]. The increased protein level of GSK-3β is not necessarily associated with the increased activity of GSK-3β, making the conclusions of the study not relevant to the real association between GSK-3β and survival of PDAC patients.
Figure 2 illustrates the proposed tumour suppressor pathways regulated by GSK-3β in pancreatic cancer.
The above-mentioned studies lead to conclude that most of the data showing the anti-oncogenic effect of GSK-3β is either weak, not relevant to pancreatic cancer, or overcome by other pro-cancer effects. Most of these studies were published between 2001 and 2010 when the pro-cancer role of GSK-3β was not well known. Data published in the last 15 years have demonstrated the oncogenic role of GSK-3β, making it a very promising target for treating cancer. This data is summarized in the next section.

5. GSK3-β Oncogenic Effect in Pancreatic Cancer

Differently from the proposed onco-suppressive roles of GSK-3β, strong data has been accumulating for many years supporting the oncogenic role of GSK-3β [2,3]. In pancreatic cancer, GSK-3β is involved in regulating cancer cell proliferation, resistance to apoptosis and autophagy, chemo-resistance, and EMT, leading to invasion and metastasis [1,2,3,41].
GSK-3β is a key regulator of the NF-κB transcription factor responsible for the expression of genes that regulate proliferation, apoptosis, inflammation, angiogenesis, invasion, and chemo-resistance in pancreatic cancer cells [42]. GSK-3β nuclear accumulation was found to significantly correlate with human pancreatic cancer de-differentiation and regulation of NF-κB. Inhibition of GSK-3β activity by AR-A014418 or by a dominant negative form of GSK-3β represses its nuclear accumulation via proteasomal degradation and arrests pancreatic tumour growth in vivo and decreases NF-κB-mediated pancreatic cancer cell survival and proliferation in established tumour xenografts [22]. During the last few years, we have gained more knowledge about the mechanism through which GSK-3β regulates NF-κB. Medunjanin et al. identified the NF-κB Essential Modifier (NEMO) as a GSK-3β substrate that is phosphorylated within the N-terminal domain at different serine residues [43]. NEMO activates IκB kinase (IKK) leading to phosphorylation of IκB and, subsequently, its proteasomal degradation, thus liberating NF-κB for nuclear translocation, enhancing transcriptional regulation [43]. GSK-3β phosphorylates NEMO leading to its stabilization and therefore, degradation of IκB and enhanced activity of NF-κB. Using NEMO mutants which could not be phosphorylated by GSK-3β, Medunjanin found that TNFα-induced NF-κB activation was hampered [44].
In addition to the GSK-3β up-regulation of the NF-κB pathway, which has a broad pro-cancer effect, GSK-3β is also involved in another major pro-cancer pathway in PDAC, namely the k-ras pathway, which is activated in 95% of PDAC patients. First, GSK-3β is a downstream target of the pancreatic cancer initiating and promoting K-ras pathway. Mutant/active K-ras enhances GSK-3β promoter activity and protein expression via the MAPK signalling, leading to increased and sustained expression of GSK-3β in PDAC cells [45].
Next, Ras-mutated tumours were also associated with increased activation of mitogenic PI3k signalling, and reduced PTEN phosphatase activity, which both drive tumour growth and survival [46]. Therefore, increasing GSK-3β expression by induction of Ras-driven MAPK results in increased cancer cell plasticity and increased EMT. Pharmacological and molecular inhibitions of GSK-3β in K-ras-mutant PDAC cancer models showed upregulation of c-Myc and β-catenin leading to apoptosis induction, and indicative treatment selection of only malignant pancreatic epithelial cells [21,39].
Importantly, Palanivel et al. showed that GSK-3β regulates the Leucine-zipper-like transcriptional regulator 1 (LZTR1)-dependent mechanism that controls the stability of Ras proteins and proliferation of pancreatic cancer cells. LZTR1 protein is a substrate adaptor for E3 ubiquitin ligases that promote the degradation of both wild type and mutant Ras proteins. The data shows that GSK-3β inhibits LZTR1, leading to a sustained level of K-ras and increased proliferation of the pancreatic cancer cells [47].
Furthermore, in a recent study, mice expressing the inducible nuclear-targeted GSK-3β transgene were crossed with K-rasG12D;Pdx1-Cre (KC) mice, leading to profound pancreatic cyst development and expansion of cytokeratin-19+ ductal cells as early as at the age of 4 weeks. This data showed that nuclear GSK-3β worked in cooperation with K-ras mutation to promote the reprogramming of pancreatic progenitors to the ductal lineage, resulting in loss of acinar cells and expansion of ductal cells with PDAC characteristics. This data suggests a role of GSK-3β in the early stage of the PDAC development and confirms the strong pro-cancer role of GSK-3β [48].
Depletion of GSK-3β sensitized PDAC cells to tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) in PDAC cells and in PDAC animal models, and the inhibition of GSK-3β function by AR-A014418 or LY2064827 in PDAC cells impaired the expression of the anti-apoptotic Bcl-xL and cIAP2 proteins [49,50].
In addition to the effect of GSK-3β on proliferation and inhibition of apoptosis, GSK-3β phosphorylates/inhibits Cyclase-Associated Protein 1 (CAP-1), leading to increased migration and invasion of pancreatic cancer cells [51]. CAP1 phosphorylated at S308/S310 regulatory site is associated with the increased activity of GSK-3β observed in pancreatic cancer. This suggests the inhibition of GSK-3β could effectively inhibit cancer cell invasion through regulation of CAP1.
The viability of therapeutic targeting of GSK-3β has been demonstrated via pharmacological inhibition and RNA interference to reduce cancer cell survival and proliferation and induce apoptosis in gastrointestinal and pancreatic cancer cells, glioblastoma, hematologic malignancies, osteo-sarcoma, gynaecologic and urogenital cancers, and lung cancers [52,53]. The inhibition of GSK-3β significantly reduced the proliferation and survival of cancer cells, sensitized them to gemcitabine and ionizing radiation, and attenuated their migration and invasion. These effects were associated with decreases in cyclin D1 expression and Rb phosphorylation. The inhibition of GSK-3β by RNA interference or by its specific inhibitor AR-A014418 also altered the subcellular localization of Rac1 and F-actin and the cellular microarchitecture, including lamellipodia. Coinciding with these changes were the reduced secretion of matrix metalloproteinase-2 (MMP-2) and decreased phosphorylation of focal adhesion kinase (FAK). The effects of GSK-3β inhibition on tumour invasion, susceptibility to gemcitabine, MMP-2 expression and FAK phosphorylation were observed in vitro as well as in tumour animal models [17,52].
Transcriptomic sub-types of pancreatic cancer were found to acquire sensitivity or tolerance to GSK-3β inhibitors. Loss of genes that drive endodermal lineage specification, HNF4A and GATA6, switch the metabolic profiles from classical to squamous, with GSK-3β as a key regulator of glycolysis. The inhibition of GSK-3β using TDZD-8 or Tideglusib resulted in selective sensitivity in the squamous subtype [54].
In addition to PDAC proliferation, apoptosis, and EMT/invasion, GSK-3β also regulates autophagy. Recently published data showed that pancreatic cancer cells treated with the anti-cancer drug salicylanilide derivative Niclosamide (Nic) have increased phosphorylation/inhibition of GSK-3β leading to inhibition of non-canonical hedgehog/Gli cascade via upregulation of Sufu and Gli3 as well as stimulation of autophagy and causing pancreatic cancer cell death [55]. Inhibited serine 9 p-GSK-3β mediates Sufu/Gli3 cascade, negatively regulating Hh/Gli1 cascade and positively regulating mTORC1 autophagy-mediated cell death [55]. This Hh-GLI impairment has also been demonstrated in preclinical models by a combination of the GSK-3β inhibitor lithium and gemcitabine dosing in pancreatic cancer [56]. Other PDAC GSK3β inhibitory treatments revolve around synergistic pharmacological interaction with Gemcitabine to treat tumour stages ranging from preclinical models to refractory, to advanced or metastatic [26,57,58]. This synergistic inhibitory treatment catalyses on the varied GSK-3β involvement in multiple signalling pathways to increase DNA damage and impair DNA repair, as well as alter the tumour microenvironment to reduce cell migration, cancer stemness, and metabolic pathways.
Inhibiting GSK-3β activity results in decreased cellular ATP-production and increased AMP/ATP ratio which contributes to AMP-activated protein kinase (AMPK) activation. These effects are coupled to increased LC-3B biosynthesis and p62 protein reduction indicating autophagy induction. Liver kinase B1 (LKB1) is involved in AMPK activation, suggesting the therapeutic use of GSK-3β inhibitor molecules, such as TDZD8, Tideglusib, TWS119, and peptide L803-mts, that promote LKB1 translocation to cytoplasm and enhanced LKB1 interaction to prevent resulting autophagy [59]. By inhibiting GSK-3β we can provide a therapeutic approach to controlling autophagy induction by manipulation of the LKB1-AMPK pathway to selectively treat tumours or metastatic lesions.
In addition, deregulation of GSK-3β expression and activity promotes tumour suppressor activity via JNK, Rb, Notch, TFEB, and C-Myc signalling pathways [60]. All this data strongly supports the pro-tumour function of GSK-3β. Figure 3 recapitulates the pro-oncogenic pathways regulated by GSK-3β in pancreatic cancer.
Based on the previous data, multiple laboratories have been working on developing GSK-3β inhibitors for treating cancer. There are multiple ongoing clinical trials testing GSK-3β inhibitors in cancer patients including four studies in PDAC patients [61,62]. Table 1 shows a list of GSK-3β inhibitors with their therapeutic activity, diseases tested on, and observed results. The table also includes a list of ongoing clinical trials testing GSK-3β inhibitors in cancers.

6. Conclusions

For many years, the role of GSK-3β in cancer cells as a pro-oncogenic or tumour-suppressor protein has been a much-disputed topic, which has stunted therapeutic targeting of this kinase. It is now clear that GSK3-β is a powerful pro-cancer protein and is a perfect candidate for treating PDAC and many other cancers.
The review of the tumour-suppressive role of GSK-3β in cancer reveals that most of these effects are either not active in pancreatic cancer, such as in the case of p53 regulation, or overcome by other pro-oncogenic effects of GSK-3β, such as in the case of β-catenin, c-Myc, and Snail pathways. On the other hand, the pro-oncogenic effects of GSK-3β are supported by strong data and interact with critical up-regulated proteins in the cancer progression such as K-ras and NF-κB. GSK3-β is expressed at higher levels in tumour cells and metastatic lesions compared to normal tissue. Therapeutic inhibition of GSK-3β has been shown to reduce cancer cell proliferation and survival in different malignant neoplasms, with combination treatments suggesting a viable method to avoid treatment resistance. The ongoing trials using GSK-3β inhibitors for treating PDAC and other cancers will reveal more data about this treatment strategy. GSK-3β is a very relevant target for treating PDAC.

Author Contributions

Conceptualization, O.H.M.E. and M.E.; investigation, O.H.M.E. and M.E.; data curation, O.H.M.E. and M.E.; writing—original draft preparation, O.H.M.E.; writing—review and editing, O.H.M.E., M.E. and S.J.P.; visualization, O.H.M.E. and M.E.; supervision, M.E.; project administration, M.E., funding acquisition, S.J.P. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the NCI P01 award CA233452.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors Edderkaoui and Pandol disclose their relationship with Avenzoar Pharmaceuticals, which is developing dual inhibitors of GSK-3β and histone deacetylases for treating cancer. Edderkaoui and Pandol are co-inventors in the patents related to Metavert.

References

  1. Cormier, K.W.; Woodgett, J.R. Recent advances in understanding the cellular roles of GSK-3. F1000Research 2017, 6, 167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Ding, L.; Billadeau, D.D. Glycogen synthase kinase-3beta: A novel therapeutic target for pancreatic cancer. Expert Opin. Ther. Targets 2020, 24, 417–426. [Google Scholar] [CrossRef] [PubMed]
  3. Uehara, M.; Domoto, T.; Takenaka, S.; Bolidong, D.; Takeuchi, O.; Miyashita, T.; Minamoto, T. Glycogen synthase kinase-3beta participates in acquired resistance to gemcitabine in pancreatic cancer. Cancer Sci. 2020, 111, 4405–4416. [Google Scholar] [CrossRef] [PubMed]
  4. Augello, G.; Emma, M.R.; Cusimano, A.; Azzolina, A.; Montalto, G.; McCubrey, J.A.; Cervello, M. The Role of GSK-3 in Cancer Immunotherapy: GSK-3 Inhibitors as a New Frontier in Cancer Treatment. Cells 2020, 9, 1427. [Google Scholar] [CrossRef]
  5. Domoto, T.; Uehara, M.; Bolidong, D.; Minamoto, T. Glycogen Synthase Kinase 3beta in Cancer Biology and Treatment. Cells 2020, 9, 1388. [Google Scholar] [CrossRef]
  6. He, R.; Du, S.; Lei, T.; Xie, X.; Wang, Y. Glycogen synthase kinase 3beta in tumorigenesis and oncotherapy (Review). Oncol. Rep. 2020, 44, 2373–2385. [Google Scholar] [CrossRef]
  7. Sahin, I.; Eturi, A.; De Souza, A.; Pamarthy, S.; Tavora, F.; Giles, F.J.; Carneiro, B.A. Glycogen synthase kinase-3 beta inhibitors as novel cancer treatments and modulators of antitumor immune responses. Cancer Biol. Ther. 2019, 20, 1047–1056. [Google Scholar] [CrossRef]
  8. Walz, A.; Ugolkov, A.; Chandra, S.; Kozikowski, A.; Carneiro, B.A.; O’Halloran, T.V.; Giles, F.J.; Billadeau, D.D.; Mazar, A.P. Molecular Pathways: Revisiting Glycogen Synthase Kinase-3beta as a Target for the Treatment of Cancer. Clin. Cancer Res. 2017, 23, 1891–1897. [Google Scholar] [CrossRef] [Green Version]
  9. Beurel, E.; Grieco, S.F.; Jope, R.S. Glycogen synthase kinase-3 (GSK3): Regulation, actions, and diseases. Pharmacol. Ther. 2015, 148, 114–131. [Google Scholar] [CrossRef] [Green Version]
  10. Thornton, T.M.; Pedraza-Alva, G.; Deng, B.; Wood, C.D.; Aronshtam, A.; Clements, J.L.; Sabio, G.; Davis, R.J.; Matthews, D.E.; Doble, B.; et al. Phosphorylation by p38 MAPK as an alternative pathway for GSK3beta inactivation. Science 2008, 320, 667–670. [Google Scholar] [CrossRef]
  11. Ding, Q.; Xia, W.; Liu, J.C.; Yang, J.Y.; Lee, D.F.; Xia, J.; Bartholomeusz, G.; Li, Y.; Pan, Y.; Li, Z.; et al. Erk associates with and primes GSK-3beta for its inactivation resulting in upregulation of beta-catenin. Mol. Cell 2005, 19, 159–170. [Google Scholar] [CrossRef] [PubMed]
  12. Cole, A.; Frame, S.; Cohen, P. Further evidence that the tyrosine phosphorylation of glycogen synthase kinase-3 (GSK3) in mammalian cells is an autophosphorylation event. Biochem. J. 2004, 377, 249–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Goc, A.; Al-Husein, B.; Katsanevas, K.; Steinbach, A.; Lou, U.; Sabbineni, H.; DeRemer, D.L.; Somanath, P.R. Targeting Src-mediated Tyr216 phosphorylation and activation of GSK-3 in prostate cancer cells inhibit prostate cancer progression in vitro and in vivo. Oncotarget 2014, 5, 775–787. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Sayas, C.L.; Ariaens, A.; Ponsioen, B.; Moolenaar, W.H. GSK-3 is activated by the tyrosine kinase Pyk2 during LPA1-mediated neurite retraction. Mol. Biol. Cell 2006, 17, 1834–1844. [Google Scholar] [CrossRef] [Green Version]
  15. Lesort, M.; Jope, R.S.; Johnson, G.V. Insulin transiently increases tau phosphorylation: Involvement of glycogen synthase kinase-3beta and Fyn tyrosine kinase. J. Neurochem. 1999, 72, 576–584. [Google Scholar] [CrossRef]
  16. Jope, R.S.; Yuskaitis, C.J.; Beurel, E. Glycogen synthase kinase-3 (GSK3): Inflammation, diseases, and therapeutics. Neurochem. Res. 2007, 32, 577–595. [Google Scholar] [CrossRef] [Green Version]
  17. Kitano, A.; Shimasaki, T.; Chikano, Y.; Nakada, M.; Hirose, M.; Higashi, T.; Ishigaki, Y.; Endo, Y.; Takino, T.; Sato, H.; et al. Aberrant glycogen synthase kinase 3beta is involved in pancreatic cancer cell invasion and resistance to therapy. PLoS ONE 2013, 8, e55289. [Google Scholar] [CrossRef] [Green Version]
  18. Vidri, R.J.; Fitzgerald, T.L. GSK-3: An important kinase in colon and pancreatic cancers. Biochimica et biophysica acta. Mol. Cell Res. 2020, 1867, 118626. [Google Scholar] [CrossRef]
  19. Ugolkov, A.V.; Matsangou, M.; Taxter, T.J.; O’Halloran, T.V.; Cryns, V.L.; Giles, F.J.; Mazar, A.P. Aberrant expression of glycogen synthase kinase-3beta in human breast and head and neck cancer. Oncol. Lett. 2018, 16, 6437–6444. [Google Scholar] [CrossRef] [Green Version]
  20. Xie, Y.; Zhu, S.; Zhong, M.; Yang, M.; Sun, X.; Liu, J.; Kroemer, G.; Lotze, M.; Zeh, H.J., 3rd; Kang, R.; et al. Inhibition of Aurora Kinase A Induces Necroptosis in Pancreatic Carcinoma. Gastroenterology 2017, 153, 1429–1443. [Google Scholar] [CrossRef]
  21. Pecoraro, C.; Faggion, B.; Balboni, B.; Carbone, D.; Peters, G.J.; Diana, P.; Assaraf, Y.G.; Giovannetti, E. GSK3beta as a novel promising target to overcome chemoresistance in pancreatic cancer. Drug Resist. Updates 2021, 58, 100779. [Google Scholar] [CrossRef] [PubMed]
  22. Ougolkov, A.V.; Fernandez-Zapico, M.E.; Bilim, V.N.; Smyrk, T.C.; Chari, S.T.; Billadeau, D.D. Aberrant nuclear accumulation of glycogen synthase kinase-3beta in human pancreatic cancer: Association with kinase activity and tumor dedifferentiation. Clin. Cancer Res. 2006, 12, 5074–5081. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Ignatz-Hoover, J.J.; Wang, V.; Mackowski, N.M.; Roe, A.J.; Ghansah, I.K.; Ueda, M.; Lazarus, H.M.; de Lima, M.; Paietta, E.; Fernandez, H.; et al. Aberrant GSK3beta nuclear localization promotes AML growth and drug resistance. Blood Adv. 2018, 2, 2890–2903. [Google Scholar] [CrossRef] [PubMed]
  24. Reya, T.; Clevers, H. Wnt signalling in stem cells and cancer. Nature 2005, 434, 843–850. [Google Scholar] [CrossRef] [PubMed]
  25. Wu, D.; Pan, W. GSK3: A multifaceted kinase in Wnt signaling. Trends Biochem. Sci. 2010, 35, 161–168. [Google Scholar] [CrossRef] [Green Version]
  26. Edderkaoui, M.; Chheda, C.; Soufi, B.; Zayou, F.; Hu, R.W.; Ramanujan, V.K.; Pan, X.; Boros, L.G.; Tajbakhsh, J.; Madhav, A.; et al. An Inhibitor of GSK3B and HDACs Kills Pancreatic Cancer Cells and Slows Pancreatic Tumor Growth and Metastasis in Mice. Gastroenterology 2018, 155, 1985–1998. [Google Scholar] [CrossRef]
  27. Nesteruk, K.; Smits, R.; Bruno, M.; Peppelenbosch, M.P.; Fuhler, G.M. Upregulated beta-catenin signaling does not affect survival of pancreatic cancer cells during dual inhibition of GSK3B and HDAC. Pancreatology 2020, 20, 558–561. [Google Scholar] [CrossRef]
  28. Kazi, A.; Xiang, S.; Yang, H.; Delitto, D.; Trevino, J.; Jiang, R.H.Y.; Ayaz, M.; Lawrence, H.R.; Kennedy, P.; Sebti, S.M. GSK3 suppression upregulates beta-catenin and c-Myc to abrogate KRas-dependent tumors. Nat. Commun. 2018, 9, 5154. [Google Scholar] [CrossRef] [Green Version]
  29. Kim, K.; Pang, K.M.; Evans, M.; Hay, E.D. Overexpression of beta-catenin induces apoptosis independent of its transactivation function with LEF-1 or the involvement of major G1 cell cycle regulators. Mol. Biol. Cell 2000, 11, 3509–3523. [Google Scholar] [CrossRef]
  30. Inoki, K.; Ouyang, H.; Zhu, T.; Lindvall, C.; Wang, Y.; Zhang, X.; Yang, Q.; Bennett, C.; Harada, Y.; Stankunas, K.; et al. TSC2 integrates Wnt and energy signals via a coordinated phosphorylation by AMPK and GSK3 to regulate cell growth. Cell 2006, 126, 955–968. [Google Scholar] [CrossRef]
  31. Hassan, Z.; Schneeweis, C.; Wirth, M.; Veltkamp, C.; Dantes, Z.; Feuerecker, B.; Ceyhan, G.O.; Knauer, S.K.; Weichert, W.; Schmid, R.M.; et al. MTOR inhibitor-based combination therapies for pancreatic cancer. Br. J. Cancer 2018, 118, 366–377. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Iriana, S.; Ahmed, S.; Gong, J.; Annamalai, A.A.; Tuli, R.; Hendifar, A.E. Targeting mTOR in Pancreatic Ductal Adenocarcinoma. Front. Oncol. 2016, 6, 99. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Azoulay-Alfaguter, I.; Elya, R.; Avrahami, L.; Katz, A.; Eldar-Finkelman, H. Combined regulation of mTORC1 and lysosomal acidification by GSK-3 suppresses autophagy and contributes to cancer cell growth. Oncogene 2015, 34, 4613–4623. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Turenne, G.A.; Price, B.D. Glycogen synthase kinase3 beta phosphorylates serine 33 of p53 and activates p53’s transcriptional activity. BMC Cell Biol. 2001, 2, 12. [Google Scholar] [CrossRef] [PubMed]
  35. Watcharasit, P.; Bijur, G.N.; Song, L.; Zhu, J.; Chen, X.; Jope, R.S. Glycogen synthase kinase-3beta (GSK3beta) binds to and promotes the actions of p53. J. Biol. Chem. 2003, 278, 48872–48879. [Google Scholar] [CrossRef] [Green Version]
  36. Cicenas, J.; Kvederaviciute, K.; Meskinyte, I.; Meskinyte-Kausiliene, E.; Skeberdyte, A.; Cicenas, J. KRAS, TP53, CDKN2A, SMAD4, BRCA1, and BRCA2 Mutations in Pancreatic Cancer. Cancers 2017, 9, 42. [Google Scholar] [CrossRef] [Green Version]
  37. Zhou, B.P.; Deng, J.; Xia, W.; Xu, J.; Li, Y.M.; Gunduz, M.; Hung, M.C. Dual regulation of Snail by GSK-3beta-mediated phosphorylation in control of epithelial-mesenchymal transition. Nat. Cell Biol. 2004, 6, 931–940. [Google Scholar] [CrossRef]
  38. Vijay, G.V.; Zhao, N.; Den Hollander, P.; Toneff, M.J.; Joseph, R.; Pietila, M.; Taube, J.H.; Sarkar, T.R.; Ramirez-Pena, E.; Werden, S.J.; et al. GSK3beta regulates epithelial-mesenchymal transition and cancer stem cell properties in triple-negative breast cancer. Breast Cancer Res. 2019, 21, 37. [Google Scholar] [CrossRef] [Green Version]
  39. Gregory, M.A.; Qi, Y.; Hann, S.R. Phosphorylation by glycogen synthase kinase-3 controls c-myc proteolysis and subnuclear localization. J. Biol. Chem. 2003, 278, 51606–51612. [Google Scholar] [CrossRef] [Green Version]
  40. Ben-Josef, E.; George, A.; Regine, W.F.; Abrams, R.; Morgan, M.; Thomas, D.; Schaefer, P.L.; DiPetrillo, T.A.; Fromm, M.; Small, W., Jr.; et al. Glycogen Synthase Kinase 3 Beta Predicts Survival in Resected Adenocarcinoma of the Pancreas. Clin. Cancer Res. 2015, 21, 5612–5618. [Google Scholar] [CrossRef]
  41. Garcea, G.; Manson, M.M.; Neal, C.P.; Pattenden, C.J.; Sutton, C.D.; Dennison, A.R.; Berry, D.P. Glycogen synthase kinase-3 beta; a new target in pancreatic cancer? Curr. Cancer Drug Targets 2007, 7, 209–215. [Google Scholar] [CrossRef] [PubMed]
  42. Li, Q.; Yang, G.; Feng, M.; Zheng, S.; Cao, Z.; Qiu, J.; You, L.; Zheng, L.; Hu, Y.; Zhang, T.; et al. NF-kappaB in pancreatic cancer: Its key role in chemoresistance. Cancer Lett. 2018, 421, 127–134. [Google Scholar] [CrossRef] [PubMed]
  43. Heydendael, W.; Sengupta, A.; Bhatnagar, S. Putative genes mediating the effects of orexins in the posterior paraventricular thalamus on neuroendocrine and behavioral adaptations to repeated stress. Brain Res. Bull. 2012, 89, 203–210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Medunjanin, S.; Schleithoff, L.; Fiegehenn, C.; Weinert, S.; Zuschratter, W.; Braun-Dullaeus, R.C. GSK-3beta controls NF-kappaB activity via IKKgamma/NEMO. Sci. Rep. 2016, 6, 38553. [Google Scholar] [CrossRef] [Green Version]
  45. Zhang, J.S.; Koenig, A.; Harrison, A.; Ugolkov, A.V.; Fernandez-Zapico, M.E.; Couch, F.J.; Billadeau, D.D. Mutant K-Ras increases GSK-3beta gene expression via an ETS-p300 transcriptional complex in pancreatic cancer. Oncogene 2011, 30, 3705–3715. [Google Scholar] [CrossRef] [Green Version]
  46. Fitzgerald, T.L.; Lertpiriyapong, K.; Cocco, L.; Martelli, A.M.; Libra, M.; Candido, S.; Montalto, G.; Cervello, M.; Steelman, L.; Abrams, S.L.; et al. Roles of EGFR and KRAS and their downstream signaling pathways in pancreatic cancer and pancreatic cancer stem cells. Adv. Biol. Regul. 2015, 59, 65–81. [Google Scholar] [CrossRef]
  47. Palanivel, C.; Chaudhary, N.; Seshacharyulu, P.; Cox, J.L.; Yan, Y.; Batra, S.K.; Ouellette, M.M. The GSK3 kinase and LZTR1 protein regulate the stability of Ras family proteins and the proliferation of pancreatic cancer cells. Neoplasia 2022, 25, 28–40. [Google Scholar] [CrossRef]
  48. Ding, L.; Roeck, K.; Zhang, C.; Zidek, B.; Rodman, E.; Hernandez-Barco, Y.; Zhang, J.S.; Bamlet, W.; Oberg, A.; Zhang, L.; et al. Nuclear GSK-3beta and Oncogenic KRas Lead to the Retention of Pancreatic Ductal Progenitor Cells Phenotypically Similar to Those Seen in IPMN. Front. Cell Dev. Biol. 2022, 10, 853003. [Google Scholar] [CrossRef]
  49. Mamaghani, S.; Simpson, C.D.; Cao, P.M.; Cheung, M.; Chow, S.; Bandarchi, B.; Schimmer, A.D.; Hedley, D.W. Glycogen synthase kinase-3 inhibition sensitizes pancreatic cancer cells to TRAIL-induced apoptosis. PLoS ONE 2012, 7, e41102. [Google Scholar] [CrossRef] [Green Version]
  50. Zhang, J.S.; Herreros-Villanueva, M.; Koenig, A.; Deng, Z.; de Narvajas, A.A.; Gomez, T.S.; Meng, X.; Bujanda, L.; Ellenrieder, V.; Li, X.K.; et al. Differential activity of GSK-3 isoforms regulates NF-kappaB and TRAIL- or TNFalpha induced apoptosis in pancreatic cancer cells. Cell Death Dis. 2014, 5, e1142. [Google Scholar] [CrossRef]
  51. Wu, H.; Hasan, R.; Zhang, H.; Gray, J.; Williams, D.; Miller, M.; Allen, F.; Lee, V.; Kelly, T.; Zhou, G.L. Phosphorylation Regulates CAP1 (Cyclase-Associated Protein 1) Functions in the Motility and Invasion of Pancreatic Cancer Cells. Sci. Rep. 2019, 9, 4925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Shimasaki, T.; Ishigaki, Y.; Nakamura, Y.; Takata, T.; Nakaya, N.; Nakajima, H.; Sato, I.; Zhao, X.; Kitano, A.; Kawakami, K.; et al. Glycogen synthase kinase 3beta inhibition sensitizes pancreatic cancer cells to gemcitabine. J. Gastroenterol. 2012, 47, 321–333. [Google Scholar] [CrossRef] [PubMed]
  53. Domoto, T.; Pyko, I.V.; Furuta, T.; Miyashita, K.; Uehara, M.; Shimasaki, T.; Nakada, M.; Minamoto, T. Glycogen synthase kinase-3beta is a pivotal mediator of cancer invasion and resistance to therapy. Cancer Sci. 2016, 107, 1363–1372. [Google Scholar] [CrossRef] [PubMed]
  54. Brunton, H.; Caligiuri, G.; Cunningham, R.; Upstill-Goddard, R.; Bailey, U.M.; Garner, I.M.; Nourse, C.; Dreyer, S.; Jones, M.; Moran-Jones, K.; et al. HNF4A and GATA6 Loss Reveals Therapeutically Actionable Subtypes in Pancreatic Cancer. Cell Rep. 2020, 31, 107625. [Google Scholar] [CrossRef]
  55. Kaushal, J.B.; Bhatia, R.; Kanchan, R.K.; Raut, P.; Mallapragada, S.; Ly, Q.P.; Batra, S.K.; Rachagani, S. Repurposing Niclosamide for Targeting Pancreatic Cancer by Inhibiting Hh/Gli Non-Canonical Axis of Gsk3beta. Cancers 2021, 13, 3105. [Google Scholar] [CrossRef]
  56. Peng, Z.; Ji, Z.; Mei, F.; Lu, M.; Ou, Y.; Cheng, X. Lithium inhibits tumorigenic potential of PDA cells through targeting hedgehog-GLI signaling pathway. PLoS ONE 2013, 8, e61457. [Google Scholar] [CrossRef] [Green Version]
  57. Kunnimalaiyaan, S.; Gamblin, T.C.; Kunnimalaiyaan, M. Glycogen synthase kinase-3 inhibitor AR-A014418 suppresses pancreatic cancer cell growth via inhibition of GSK-3-mediated Notch1 expression. HPB 2015, 17, 770–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Ding, L.; Madamsetty, V.S.; Kiers, S.; Alekhina, O.; Ugolkov, A.; Dube, J.; Zhang, Y.; Zhang, J.S.; Wang, E.; Dutta, S.K.; et al. Glycogen Synthase Kinase-3 Inhibition Sensitizes Pancreatic Cancer Cells to Chemotherapy by Abrogating the TopBP1/ATR-Mediated DNA Damage Response. Clin. Cancer Res. 2019, 25, 6452–6462. [Google Scholar] [CrossRef]
  59. Sun, A.; Li, C.; Chen, R.; Huang, Y.; Chen, Q.; Cui, X.; Liu, H.; Thrasher, J.B.; Li, B. GSK-3beta controls autophagy by modulating LKB1-AMPK pathway in prostate cancer cells. Prostate 2016, 76, 172–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Nagini, S.; Sophia, J.; Mishra, R. Glycogen synthase kinases: Moonlighting proteins with theranostic potential in cancer. Semin. Cancer Biol. 2019, 56, 25–36. [Google Scholar] [CrossRef] [PubMed]
  61. Garcia-Sampedro, A.; Gaggia, G.; Ney, A.; Mahamed, I.; Acedo, P. The State-of-the-Art of Phase II/III Clinical Trials for Targeted Pancreatic Cancer Therapies. J. Clin. Med. 2021, 10, 566. [Google Scholar] [CrossRef] [PubMed]
  62. Abrams, S.L.; Akula, S.M.; Meher, A.K.; Steelman, L.S.; Gizak, A.; Duda, P.; Rakus, D.; Martelli, A.M.; Ratti, S.; Cocco, L.; et al. GSK-3beta Can Regulate the Sensitivity of MIA-PaCa-2 Pancreatic and MCF-7 Breast Cancer Cells to Chemotherapeutic Drugs, Targeted Therapeutics and Nutraceuticals. Cells 2021, 10, 816. [Google Scholar] [CrossRef] [PubMed]
  63. Gaisina, I.N.; Gallier, F.; Ougolkov, A.V.; Kim, K.H.; Kurome, T.; Guo, S.; Holzle, D.; Luchini, D.N.; Blond, S.Y.; Billadeau, D.D.; et al. From a natural product lead to the identification of potent and selective benzofuran-3-yl-(indol-3-yl)maleimides as glycogen synthase kinase 3beta inhibitors that suppress proliferation and survival of pancreatic cancer cells. J. Med. Chem. 2009, 52, 1853–1863. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Ding, L.; Liou, G.Y.; Schmitt, D.M.; Storz, P.; Zhang, J.S.; Billadeau, D.D. Glycogen synthase kinase-3beta ablation limits pancreatitis-induced acinar-to-ductal metaplasia. J. Pathol. 2017, 243, 65–77. [Google Scholar] [CrossRef] [Green Version]
  65. Marchand, B.; Tremblay, I.; Cagnol, S.; Boucher, M.J. Inhibition of glycogen synthase kinase-3 activity triggers an apoptotic response in pancreatic cancer cells through JNK-dependent mechanisms. Carcinogenesis 2012, 33, 529–537. [Google Scholar] [CrossRef]
  66. Carbone, D.; Parrino, B.; Cascioferro, S.; Pecoraro, C.; Giovannetti, E.; Di Sarno, V.; Musella, S.; Auriemma, G.; Cirrincione, G.; Diana, P. 1,2,4-Oxadiazole Topsentin Analogs with Antiproliferative Activity against Pancreatic Cancer Cells, Targeting GSK3beta Kinase. ChemMedChem 2021, 16, 537–554. [Google Scholar] [CrossRef]
  67. Ougolkov, A.V.; Billadeau, D.D. Inhibition of glycogen synthase kinase-3. Methods Mol. Biol. 2008, 468, 67–75. [Google Scholar] [CrossRef]
  68. Zamek-Gliszczynski, M.J.; Abraham, T.L.; Alberts, J.J.; Kulanthaivel, P.; Jackson, K.A.; Chow, K.H.; McCann, D.J.; Hu, H.; Anderson, S.; Furr, N.A.; et al. Pharmacokinetics, metabolism, and excretion of the glycogen synthase kinase-3 inhibitor LY2090314 in rats, dogs, and humans: A case study in rapid clearance by extensive metabolism with low circulating metabolite exposure. Drug Metab. Dispos. 2013, 41, 714–726. [Google Scholar] [CrossRef] [Green Version]
  69. Bhat, R.; Xue, Y.; Berg, S.; Hellberg, S.; Ormo, M.; Nilsson, Y.; Radesater, A.C.; Jerning, E.; Markgren, P.O.; Borgegard, T.; et al. Structural insights and biological effects of glycogen synthase kinase 3-specific inhibitor AR-A014418. J. Biol. Chem. 2003, 278, 45937–45945. [Google Scholar] [CrossRef] [Green Version]
  70. Mamaghani, S.; Patel, S.; Hedley, D.W. Glycogen synthase kinase-3 inhibition disrupts nuclear factor-kappaB activity in pancreatic cancer, but fails to sensitize to gemcitabine chemotherapy. BMC Cancer 2009, 9, 132. [Google Scholar] [CrossRef]
  71. Ban, J.O.; Oh, J.H.; Son, S.M.; Won, D.; Song, H.S.; Han, S.B.; Moon, D.C.; Kang, K.W.; Song, M.J.; Hong, J.T. Troglitazone, a PPAR agonist, inhibits human prostate cancer cell growth through inactivation of NFkappaB via suppression of GSK-3beta expression. Cancer Biol. Ther. 2011, 12, 288–296. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Meijer, L.; Skaltsounis, A.L.; Magiatis, P.; Polychronopoulos, P.; Knockaert, M.; Leost, M.; Ryan, X.P.; Vonica, C.A.; Brivanlou, A.; Dajani, R.; et al. GSK-3-selective inhibitors derived from Tyrian purple indirubins. Chem. Biol. 2003, 10, 1255–1266. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Duda, P.; Akula, S.M.; Abrams, S.L.; Steelman, L.S.; Martelli, A.M.; Cocco, L.; Ratti, S.; Candido, S.; Libra, M.; Montalto, G.; et al. Targeting GSK3 and Associated Signaling Pathways Involved in Cancer. Cells 2020, 9, 1110. [Google Scholar] [CrossRef] [PubMed]
  74. Kubic, J.D.; Mascarenhas, J.B.; Iizuka, T.; Wolfgeher, D.; Lang, D. GSK-3 promotes cell survival, growth, and PAX3 levels in human melanoma cells. Mol. Cancer Res. 2012, 10, 1065–1076. [Google Scholar] [CrossRef] [Green Version]
  75. Piazza, F.; Manni, S.; Tubi, L.Q.; Montini, B.; Pavan, L.; Colpo, A.; Gnoato, M.; Cabrelle, A.; Adami, F.; Zambello, R.; et al. Glycogen Synthase Kinase-3 regulates multiple myeloma cell growth and bortezomib-induced cell death. BMC Cancer 2010, 10, 526. [Google Scholar] [CrossRef]
  76. Aguilar-Morante, D.; Morales-Garcia, J.A.; Sanz-SanCristobal, M.; Garcia-Cabezas, M.A.; Santos, A.; Perez-Castillo, A. Inhibition of glioblastoma growth by the thiadiazolidinone compound TDZD-8. PLoS ONE 2010, 5, e13879. [Google Scholar] [CrossRef] [Green Version]
  77. Dickey, A.; Schleicher, S.; Leahy, K.; Hu, R.; Hallahan, D.; Thotala, D.K. GSK-3beta inhibition promotes cell death, apoptosis, and in vivo tumor growth delay in neuroblastoma Neuro-2A cell line. J. Neurooncol. 2011, 104, 145–153. [Google Scholar] [CrossRef] [Green Version]
  78. Beurel, E.; Blivet-Van Eggelpoel, M.J.; Kornprobst, M.; Moritz, S.; Delelo, R.; Paye, F.; Housset, C.; Desbois-Mouthon, C. Glycogen synthase kinase-3 inhibitors augment TRAIL-induced apoptotic death in human hepatoma cells. Biochem. Pharmacol. 2009, 77, 54–65. [Google Scholar] [CrossRef]
  79. Georgievska, B.; Sandin, J.; Doherty, J.; Mortberg, A.; Neelissen, J.; Andersson, A.; Gruber, S.; Nilsson, Y.; Schott, P.; Arvidsson, P.I.; et al. AZD1080, a novel GSK3 inhibitor, rescues synaptic plasticity deficits in rodent brain and exhibits peripheral target engagement in humans. J. Neurochem. 2013, 125, 446–456. [Google Scholar] [CrossRef]
  80. Rosa, A.O.; Egea, J.; Martinez, A.; Garcia, A.G.; Lopez, M.G. Neuroprotective effect of the new thiadiazolidinone NP00111 against oxygen-glucose deprivation in rat hippocampal slices: Implication of ERK1/2 and PPARgamma receptors. Exp. Neurol. 2008, 212, 93–99. [Google Scholar] [CrossRef]
  81. Kushner, B.H.; Cheung, I.Y.; Modak, S.; Basu, E.M.; Roberts, S.S.; Cheung, N.K. Humanized 3F8 Anti-GD2 Monoclonal Antibody Dosing With Granulocyte-Macrophage Colony-Stimulating Factor in Patients With Resistant Neuroblastoma: A Phase 1 Clinical Trial. JAMA Oncol. 2018, 4, 1729–1735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Kramer, K.; Pandit-Taskar, N.; Humm, J.L.; Zanzonico, P.B.; Haque, S.; Dunkel, I.J.; Wolden, S.L.; Donzelli, M.; Goldman, D.A.; Lewis, J.S.; et al. A phase II study of radioimmunotherapy with intraventricular (131) I-3F8 for medulloblastoma. Pediatr. Blood Cancer 2018, 65, e26754. [Google Scholar] [CrossRef] [PubMed]
  83. Kushner, B.H.; Cheung, N.K. GM-CSF enhances 3F8 monoclonal antibody-dependent cellular cytotoxicity against human melanoma and neuroblastoma. Blood 1989, 73, 1936–1941. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Kushner, B.H.; Modak, S.; Basu, E.M.; Roberts, S.S.; Kramer, K.; Cheung, N.K. Posterior reversible encephalopathy syndrome in neuroblastoma patients receiving anti-GD2 3F8 monoclonal antibody. Cancer 2013, 119, 2789–2795. [Google Scholar] [CrossRef] [Green Version]
  85. Heiner, J.P.; Miraldi, F.; Kallick, S.; Makley, J.; Neely, J.; Smith-Mensah, W.H.; Cheung, N.K. Localization of GD2-specific monoclonal antibody 3F8 in human osteosarcoma. Cancer Res. 1987, 47, 5377–5381. [Google Scholar]
  86. Cheung, N.K.; Kushner, B.H.; Yeh, S.D.; Larson, S.M. 3F8 monoclonal antibody treatment of patients with stage 4 neuroblastoma: A phase II study. Int. J. Oncol. 1998, 12, 1299–1306. [Google Scholar] [CrossRef]
  87. Montecillo-Aguado, M.; Morales-Martinez, M.; Huerta-Yepez, S.; Vega, M.I. KLF4 inhibition by Kenpaullone induces cytotoxicity and chemo sensitization in B-NHL cell lines via YY1 independent. Leuk. Lymphoma 2021, 62, 1422–1431. [Google Scholar] [CrossRef]
  88. Kitabayashi, T.; Dong, Y.; Furuta, T.; Sabit, H.; Jiapaer, S.; Zhang, J.; Zhang, G.; Hayashi, Y.; Kobayashi, M.; Domoto, T.; et al. Identification of GSK3beta inhibitor kenpaullone as a temozolomide enhancer against glioblastoma. Sci. Rep. 2019, 9, 10049. [Google Scholar] [CrossRef] [Green Version]
  89. Yeo, M.; Chen, Y.; Jiang, C.; Chen, G.; Wang, K.; Chandra, S.; Bortsov, A.; Lioudyno, M.; Zeng, Q.; Wang, P.; et al. Repurposing cancer drugs identifies kenpaullone which ameliorates pathologic pain in preclinical models via normalization of inhibitory neurotransmission. Nat. Commun. 2021, 12, 6208. [Google Scholar] [CrossRef]
  90. Hu, S.; Ueda, M.; Stetson, L.; Ignatz-Hoover, J.; Moreton, S.; Chakrabarti, A.; Xia, Z.; Karan, G.; de Lima, M.; Agrawal, M.K.; et al. A Novel Glycogen Synthase Kinase-3 Inhibitor Optimized for Acute Myeloid Leukemia Differentiation Activity. Mol. Cancer Ther. 2016, 15, 1485–1494. [Google Scholar] [CrossRef]
Figure 1. Phosphorylation sites of GSK-3β. Green arrows show phosphorylation. Blue lanes show dephosphorylation.
Figure 1. Phosphorylation sites of GSK-3β. Green arrows show phosphorylation. Blue lanes show dephosphorylation.
Ijms 23 14133 g001
Figure 2. Proposed onco-suppressive pathways regulated by GSK-3β but found not relevant to pancreatic cancer.
Figure 2. Proposed onco-suppressive pathways regulated by GSK-3β but found not relevant to pancreatic cancer.
Ijms 23 14133 g002
Figure 3. Oncogenic pathways up-regulated by GSK-3β.
Figure 3. Oncogenic pathways up-regulated by GSK-3β.
Ijms 23 14133 g003
Table 1. Inhibitors of GSK-3β in Cancer Treatment.
Table 1. Inhibitors of GSK-3β in Cancer Treatment.
GSK-3β InhibitorsTherapeutic ActivityGSK-3β SpecificityResultant TargetingDiseases EffectedReference/Clinical Trial
benzofuran-3-yl- (indol-3-yl) maleimidesATP CompetitiveYesSupresses proliferation and survival of cancer cells Pancreatic Cancer[63]
9-ING-41ATP CompetitiveYesImpaired glucose metabolism
Autophagy
Differentiation of Renal Cancer
Breast Cancer,
Renal Cell Carcinoma
[64]
NCT03678883, 1801 phase 1/2 study
CHIR98014ATP CompetitiveYesROS generation and mitochondrial dysfunctionLung Cancer[65]
NCT01287520, 2018
Library of synthetic (1,2,4-oxadiazole) topsentin analogsATP Competitive
EMT markers SNAIL-1/2 and metalloproteinase-9
YesApoptosis InductionPancreatic Cancer[66]
MetavertDual Inhibitor- GSK-3β/ HDACYes (with inhibition of HDAC)Reduced EMT markers and cancer cell survivalPancreatic Cancer[26]
Lithium
(Lithium Carbonate)
Non-ATP Competitive- Mg2+ competitorNoMood stabilizerOsteosarcoma,
Esophageal Cancer
[56]
Tideglusib
NP031112
NP-12
Non- ATP CompetitiveYesBeta-Catenin
C-MYC
Pancreatic Cancer[28,67]
AZD-1080ATP CompetitiveYesBeta-Catenin
C-MYC
Pancreatic Cancer[7]
LY2090314ATP Competitive-Phosphorylation InhibitionYesTAK1Pancreatic Cancer,
Leukemia
[68]
NCT01632306, 2019
NCT01287520
NCT01214603
SB-732881-HATP CompetitiveBoth α and β isoforms of GSK-3Apoptosis InductionPancreatic Cancer[65]
AR-A014418ATP CompetitiveYesNOTCH1,
Cell differentiation/ proliferation inhibition
Pancreatic Cancer, Prostate Cancer[69,70,71]
BIOATP Competitive- Phosphorylation Inhibition
Inhibits CDKs
Both α and β isoforms of GSK-3Caspase 3
Caspase 9
Beta-Catenin
C-MYC
Cell proliferation ROS generation and mitochondrial dysfunction
Glioblastoma multiforme,
Pancreatic Cancer,
Osteosarcoma,
Lung Cancer
[28,72]
6BIOAndrogen receptor and signalling downregulationBoth α and β isoforms of GSK-3Androgen Receptor Signalling
Induced autophagy
Reduced metastasis
Prostate Cancer[73]
SB216763ALP expressionBoth α and β isoforms of GSK-3Androgen receptor activity, cell proliferation
FAK
Prostate Cancer,
Cervical Adenocarcinoma,
Colon Adenocarcinoma,
Osteosarcoma
[73,74,75]
TWS119Induces neuronal and CD8(+) T cell differentiationYesγδT cell survival and proliferation activatedBreast Cancer,
Colon Cancer,
Lung cancer
[76]
SB415286PD1Both α and β isoforms of GSK-3Improves osteoblastic differentiationLymphoma,
Melanoma
[73,77,78]
ABC1183CDK9
Beta Catenin
TNFalpha/IL6
NoGrowth inhibitoryMelanoma,
Gastrointestinal cancers
[19,73]
TDZD-8ATP non-competitive XIAPYesApoptosisNeuroblastoma,
Glioblastoma
[73,79]
CHIR99021Wnt activatorBoth α and β isoforms of GSK-3Microtubule stability,
Impaired chromosomal alignment
Non-small-cell Carcinoma[65,73]
NCT01287520, 2018
Indirubin
(Indirubin-3′-oxime)
Inhibits CDKs
Inhibits protein kinases
NoCell cycle arrest and apoptosisLeukemia[73]
NP00111ATP non-competitiveNoProtection against oxygen and glucose deprivationCancer[80]
3F8GD2YesEnhanced cellular cytotoxicityNeuroblastoma,
Melanoma,
Osteosarcoma,
Medulloblastoma
[81,82,83,84,85,86]
KenpaulloneInhibits CDKsNoInduced cytotoxicity and chemosensitivityGlioblastoma,
Breast Cancer
[87,88,89]
GS87MAPK signalling activationBoth α and β isoforms of GSK-3differentiation and growth arrestAcute Myeloid Leukemia[90]
2-Thio(3-iodobenzyl)-5-(1-pyridyl)-[1,3,4]-oxadiazoleCaspase-3 and cytochrome c inhibitionYesMitochondrial p53 apoptotic signallingNeuroblastoma and lung carcinoma [35]
LY2064827NF-κB and TRAIL- or TNFα induced apoptosisBoth α and β isoforms of GSK-3Induced chemosensitivityPancreatic cancer[50]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Elmadbouh, O.H.M.; Pandol, S.J.; Edderkaoui, M. Glycogen Synthase Kinase 3β: A True Foe in Pancreatic Cancer. Int. J. Mol. Sci. 2022, 23, 14133. https://doi.org/10.3390/ijms232214133

AMA Style

Elmadbouh OHM, Pandol SJ, Edderkaoui M. Glycogen Synthase Kinase 3β: A True Foe in Pancreatic Cancer. International Journal of Molecular Sciences. 2022; 23(22):14133. https://doi.org/10.3390/ijms232214133

Chicago/Turabian Style

Elmadbouh, Omer H. M., Stephen J. Pandol, and Mouad Edderkaoui. 2022. "Glycogen Synthase Kinase 3β: A True Foe in Pancreatic Cancer" International Journal of Molecular Sciences 23, no. 22: 14133. https://doi.org/10.3390/ijms232214133

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