We aimed to investigate the mechanistic, functional, and therapeutic role of glycogen synthase kinase 3β (GSK-3β) in the regulation and activation of the proinflammatory oncogenic transcription factor nuclear factor of activated T cells (NFATc2) in pancreatic cancer. IHC, qPCR, immunoblotting, immunofluorescence microscopy, and proliferation assays were used to analyze mouse and human tissues and cell lines. Protein–protein interactions and promoter regulation were analyzed by coimmunoprecipitation, DNA pulldown, reporter, and ChIP assays. Preclinical assays were performed using a variety of pancreatic cancer cells lines, xenografts, and a genetically engineered mouse model (GEMM). GSK-3β–dependent SP2 phosphorylation mediates NFATc2 protein stability in the nucleus of pancreatic cancer cells stimulating pancreatic cancer growth. In addition to protein stabilization, GSK-3β also maintains NFATc2 activation through a distinct mechanism involving stabilization of NFATc2–STAT3 complexes independent of SP2 phosphorylation. For NFATc2–STAT3 complex formation, GSK-3β–mediated phosphorylation of STAT3 at Y705 is required to stimulate euchromatin formation of NFAT target promoters, such as cyclin-dependent kinase-6, which promotes tumor growth. Finally, preclinical experiments suggest that targeting the NFATc2–STAT3–GSK-3β module inhibits proliferation and tumor growth and interferes with inflammation-induced pancreatic cancer progression in KrasG12D mice. In conclusion, we describe a novel mechanism by which GSK-3β fine-tunes NFATc2 and STAT3 transcriptional networks to integrate upstream signaling events that govern pancreatic cancer progression and growth. Furthermore, the therapeutic potential of GSK-3β is demonstrated for the first time in a relevant Kras and inflammation-induced GEMM for pancreatic cancer. Mol Cancer Ther; 15(3); 491–502. ©2016 AACR.

Pancreatic ductal adenocarcinoma (PDAC) is among the most lethal of solid malignancies with more than 40,000 new cases diagnosed in the United States each year, thus posing a significant health problem at the beginning of the twenty-first century (1). Mouse models and genetic studies have highlighted that pancreatic carcinogenesis occurs through chronologic accumulation of genetic alterations, of which activating mutations of the Kras oncogene represent a signature event that governs a multitude of mitogenic signaling events driving tumor initiation and progression (2). Importantly, preneoplastic cells and pancreatic cancer cells show a marked upregulation of inflammatory cytokines and transcription factors and are surrounded by a pronounced inflammatory microenvironment that enables Kras-mutant cells to progress toward frank malignancy (3–8). Moreover, chronic pancreatitis represents an important risk factor for the development of pancreatic cancer, underscoring the crucial role of inflammation in the process of malignant transformation in the pancreas (9–11). Blockade of these inflammatory factors, either directly or indirectly via interference with relevant signaling nodes, is likely to be important for effective Kras-directed therapy.

Interestingly, a recent study discovered Kras-dependent epigenetic induction of the glycogen synthase kinase 3β (GSK-3β) in a genetically engineered mouse model (GEMM) of PDAC. The authors showed that Kras signaling recruits the ETS/p300 transcriptional complex to the GSK-3β promoter, whose increased expression stimulated cancer cell growth (12). GSK-3β is a constitutive active and multifunctional Ser/Thr kinase, which, due to its broad substrate specificity, is involved in numerous cellular processes (13, 14). GSK-3β was initially recognized as an important tumor suppressor in various cancers predominantly acting through destabilization of the oncogene β-catenin (15). However, GSK-3β also plays important protumorigenic roles in several human malignancies, most notably in pancreatic cancer, regulating proliferation and survival (16–20). Increased expression of active GSK-3β and nuclear accumulation of active GSK-3β significantly correlated with a dedifferentiated state in pancreatic cancer cells (18). Furthermore, GSK-3β is commonly involved in inflammatory responses such as enhanced cytokine production driven by NF-κB and STAT3 pathways (21–24), and inhibition was shown to inhibit tumor cell growth via downregulation of NF-κB–mediated expression of antiapoptotic and proinflammatory genes (18, 25). Thus, GSK-3β has recently received increasing attention as a therapeutic target in preclinical and clinical therapeutics. However, the downstream effects of GSK-3β are complex and by far not restricted to the NF-κB pathway. Therefore, further studies are needed to fully comprehend the multiple molecular and cellular mechanisms by which GSK-3β contributes to the pathogenesis of cancer.

NFAT (nuclear factor of activated T cells) proteins are oncogenic transcription factors, which are induced upon inflammatory conditions and act as crucial promoters of Kras-induced pancreatic cancer progression and cellular plasticity (26). Moreover, nuclear NFAT is highly overexpressed in the majority of invasively growing pancreatic cancers (27–30). Interestingly, recent data suggest that NFATc2 is regulated by GSK-3β (31, 32); however, the exact mechanism and subsequent functional implications on tumor growth and inflammation-induced tumor progression are unknown.

In this study, we sought to investigate the mechanistic role of GSK-3β in the regulation and transcriptional activation of oncogenic NFATc2. Furthermore, we use pharmacologic as well as genetic approaches in vitro and in several mouse models of pancreatic cancer to address the therapeutic potential of this oncogenic-inflammatory signaling axis.

Cell culture

The human pancreatic adenocarcinoma cell line IMIM-PC1 was provided by FX Real (Institute Municipale de Investigacion Medica, Barcelona, Spain), Suit-028 and Suit-007 were kindly gifted from T. Iwamura (Miyazaki Medical College, Miyazaki, Japan). PaTu8988t were obtained from H.P. Elsaesser (Department of Cell Biology, Marburg, Germany). LinxA cells were purchased from Open Biosystems. All cell lines were obtained between 2004 to 2010. Mouse KrasG12D;p53−/− cells were established from mouse Pdx1-KrasG12D;p53−/− pancreatic tumors and generously gifted by J. Siveke (Technische Universität München, Munich, Germany) in 2012. Testing and authentication of human cell lines were not performed by the authors. All human pancreatic cancer cells were cultured in DMEM (Gibco, Invitrogen Corp.), supplemented with 10% FCS (Gibco) and 50 μg/mL gentamicin (Gibco). LinxA cells were maintained in DMEM (PAA Laboratories GmbH), supplemented with 10% FCS and 100 μg/mL hygromycin (Carl Roth GmbH). Mouse KrasG12D;p53−/− cells were grown in DMEM, 10% FCS, 1% nonessential amino acids (Gibco), and 1% penicillin/streptomycin (Gibco). GSK-3β inhibitor AR-A014418 was purchased from Calbiochem, caerulein, ionomycin, and IL6 were from Sigma-Aldrich. GSK-3β inhibitor 9-ING-41 has been previously described (33).

Plasmids

The full-length human NFATc2 expression vector as well as the bacterial expression vector pENTR11 NFATc2 were kindly provided by A. Rao (Department of Signaling and Gene Expression, La Jolla Institute for Allergy and Immunology, La Jolla, CA). GSK-3β S9A was from J. Woodgett (Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada). The wt STAT3 and STAT3 Y705F constructs were from J. Darnell (Laboratory of Molecular Cell Biology, The Rockefeller University, New York, NY). pBabe V5-GSK-3β S9A was from B. Manning (Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA). The NFAT-luc reporter construct harboring three consensus sites for NFAT fused to a luciferase gene was purchased from Stratagene. The HA-tagged NFATc2 construct was generated by cloning of HA-wt NFATc2 into the pcDNA3.1 (+) vector supplied by Invitrogen into HindIII and XbaI restriction sites. The pSP2 mutation was generated as previously described (31).

Generation of viral vectors and stable cell lines

Retroviral vectors and stable cell lines were generated as described previously (28). Briefly, HA-tagged NFATc2 constructs were cloned into a pQCXIP vector (BD Biosciences) and transfected into the packaging cell line LinxA. The supernatant containing retroviral particles was filtered through a 0.45-μm filter and supplemented with 2 μg/mL polybrene (Millipore) to infect the target cells PaTu8988t, Suit-028, and Suit-007. Spin infection was carried out at 2,500 × g and at 37°C. Transduced cells were selected with 1 μg/mL puromycin (Sigma-Aldrich) for at least two weeks. The successful overexpression of the NFATc2 constructs was tested by Western blotting.

qPCR analysis

RNA from cell lines was isolated using RNeasy Mini Kit (Qiagen), and first-strand cDNA was synthesized from 2 μg total RNA using random primers and Omniscript Reverse Transkriptase Kit (Qiagen). xs-13 was used as housekeeping gene. The following primer pairs were used: xs-13; 5′-gtcggaggagtcggacgag-3′, 5′-gcctttatttccttgttttgcaaa-3′, CDK-6; 5′-tcccgttccactgcgtcc-3′, 5′-caagccgcttaatccttcctggtt-3′. All primers were supplied by Biomers. Real-time PCR experiments were performed in triplicates and are displayed ± SD.

Transient transfection and siRNA

The transient transfection of expression constructs was carried out 24 hours after seeding using TransFast (Promega) and Lipofectamine 2000 (Invitrogen) as transfection reagents following manufacturer's instructions. siRNA was transfected using the siLentFect Lipid Reagent (Bio-Rad) according to the manufacturer's protocol. The RNAi oligonucleotide sequences were as follows: NFATc2, 5′-ccauuaaacaggagcagaatt-3′; STAT3, 5′-gcacaaucuacgaagaauctt-3′ (Applied Biosystems); GSK-3β, siGENOME ON-TARGETplus SMARTpool duplex (Thermo Scientific). The Silencer Negative Control from Ambion (Applied Biosystems) was used as a negative control.

Subcellular fractionation and coimmunoprecipitation

Subcellular fractionation was performed as described previously (2). Briefly, cells were washed with ice-cold PBS and centrifuged at 2,500 × g at 4°C. Pellets were subsequently resuspended in buffer A (10 mmol/L HEPES, pH 7.9, 10 mmol/L KCl, 0.1 mmol/L EDTA, 0.1 mmol/L EGTA, 1 mmol/L DTT, and protease inhibitors) for 30 minutes and consequently centrifuged for 20 minutes at 5,000 × g. Supernatants were transferred to new tubes and centrifuged for 20 minutes at 12,000 × g. Pellets were resuspended in buffer C (20 mmol/L HEPES, pH 7.9, 0.4 mmol/L NaCl, 1 mmol/L EDTA, 1 mmol/L EGTA, 1 mmol/L DTT, and protease inhibitors) and incubated on ice for additional 30 minutes. The final centrifugation step was carried out at 12,000 × g for 10 minutes.

For coimmunoprecipitation, cells were lysed with lysis buffer (50 mmol/L HEPES; pH 7.5–7.9, 150 mmol/L NaCl, 1 mmol/L EGTA, 10% glycerin, 1% Triton X-100, 100 mmol/L NaF, and 10 mmol/L Na4P2O7 × 10 H20) containing protease inhibitors. Five hundred micrograms of the total lysates or 100 μg of nuclear lysates were immunoprecipitated with the indicated antibodies and protein G- or A-agarose (Roche Diagnostics).

For isolation of proteins from murine tissues, the tissues were homogenized using a mortar and subsequently mixed with lysis buffer. After sonication, cells were centrifuged at 12,000 × g for 10 minutes and subjected to immunoblotting.

Chromatin immunoprecipitation

Chromatin immunoprecipitation assays using IgG (Upstate Biotechnology) or antibodies specific to NFATc2, H3K4me3 (all Abcam), RNA Polymerase II (Millipore), or STAT3 (Cell Signaling Technology) were performed as described previously (27). Bound regions were detected by qPCR using the following primer: CDK-6 promoter; 5′-tcccgttccactgcgtcc-3′, 5′-caagccgcttaatccttcctggtt-3′ (Biomers).

Reporter gene assays

Cells were seeded in 12-well plates and were transfected after 24 hours with the indicated constructs. Luciferase activity was measured using the Lumat LB 9501 Luminometer (Berthold Technologies) and the Dual-Luciferase Reporter Assay System (Promega) according to the manufacturer's instructions. Firefly luciferase values were normalized to Renilla luciferase activity and are shown as mean values ± SD.

DNA pulldown assays

For DNA pulldown assays, 100 μg to 150 μg of nuclear protein per sample was incubated for 3 hours with 1 μg of biotinylated double-stranded oligonucleotides containing the consensus NFAT-binding site (5′-tctaaggaggaaaaactgtttcatg-3′ and 5′-catgaaacagtttttcctccttaga-3′). DNA–protein complexes were further incubated with streptavidin–agarose beads (Sigma-Aldrich) for 1 hour, washed twice with lysis buffer, and subjected to immunoblotting.

Proliferation assays

Cell proliferation was measured by 3Hthymidine and bromodeoxyuridine (BrdUrd) incorporation. Cells were seeded in 24-well plates and cultured in medium containing 10% FCS. Nineteen hours after the indicated treatment or transfection, 3Hthymidine (0.5 μCi/well) was added during the last 5 hours of incubation. The cells were washed with 5% trichloroacetic acid, and the acid-insoluble fraction was dissolved by incubation with 1 μmol/L NaOH for 30 minutes at 37°C. Radioactivity was evaluated with a scintillation counter (Pharmacia). For BrdUrd assays, (transfected) cells were seeded in 96-well plates and treated as indicated. Incorporation of BrdUrd was assayed with Cell Detection ELISA, BrdU Kit (Roche) according to the manufacturer's instructions. The intensity of incorporation was measured with a DLReady Luminometer (Berthold Technologies). All proliferation assays were performed in triplicate in at least three independent experiments.

Mouse strains and in vivo experiments

Chronic pancreatitis was induced in 8-week-old Pdx1-Cre;KrasG12D mice by single daily intraperitoneal injections of caerulein (0.2 mg/kg BW; Sigma-Aldrich) and the GSK-3β inhibitor 9-ING-41 (20 mg/kg BW) 3 days per week for a period of 4 weeks (33). Mice were sacrificed at 12 weeks of age.

For the nude mice studies, 6- to 8-week-old pathogen-free athymic female nu/nu mice were obtained from Harlan Winkelmann. A total of 1 × 106 IMIM-PC1 was mixed with Matrigel (BD Biosciences) and injected subcutaneously in both flanks of 12 nude mice. After the establishment of visible tumors, mice were randomized into two groups and were treated 3 times weekly with either 10% DMSO or 10 mg/kg of AR-A014418 (Calbiochem). Mice and tumor sizes were monitored twice weekly. Furthermore, we injected 1 × 106 parental PaTu8988t control cells or PaTu8988t cells stably overexpressing wt NFATc2 and NFATc2 pSP2 subcutaneously into the flank of 6 nude mice each and weekly measured the tumor sizes. The tumor volumes (V) were estimated using the formula V = 4/3 × π × (L/2) × (i/2) × (H/2) where L = length, W = width, and H = height. After 4 weeks, mice were sacrificed and tumors were explanted, analyzed by size, and subjected to IHC and qPCR or Western Blot analyses. Animal experiments were carried out using protocols approved by the Institutional Animal Care and Use Committee at the University of Marburg (Marburg, Germany).

Ki67 proliferation analysis

High-power images (20× objective) were taken from Ki67-stained slides from at least 3 animals for each group. Nuclei positive for Ki67 were counted as actively proliferating cells in 20 metaplastic lesions and were related to total numbers of nuclei per lesion. Data were expressed as percentage of positive nuclei per total nuclei.

Fluorescence microscopy, IHC, and Western blotting

Immunofluorescence and IHC were performed on formalin-fixed, paraffin-embedded tumor sections as described previously (28). The following antibodies were used for immunofluorescence: HA (1:1,000) and STAT3 (1:200; all Cell Signaling Technology) and for IHC: NFATc2 (1:200; Abcam), p-NFATc2 (S213/217/221; 1:50, Santa Cruz Biotechnology), STAT3 (1:100, Cell Signaling Technology), p-STAT3 (Y705; 1:50, Cell Signaling Technology), p-GS (S641/645; 1:25, Cell Signaling Technology), Ki67 (1:600, NeoMarker), GSK-3β (1:200, Epitomics), and CD45 (BD Pharmingen, 1:100). Immunoblotting was performed using standard methods on uncultured, macrodissected tumor protein lysates or lysates from cultured cell lines. Membranes were probed with NFATc2 (Abcam), p-NFATc2 (S213/217/221; Abcam), Flag (Sigma-Aldrich), HA, STAT3, GSK-3β, CDK-6, p-STAT3 (Y705), p-GS (S641/645), GS (all Cell Signaling Technology), V5 (Invitrogen), and β-actin (Sigma-Aldrich) antibodies. Densitometric analysis was carried out with ImageQuant 5.1.

TMA staining and analysis

All staining carried out on human specimens was approved by the Mayo Clinic Institutional Review Board (Rochester, MN). Written confirmation was obtained from each patient. Tissue microarrays (TMA) containing samples from PDAC patients were stained as indicated and analyzed for NFATc2 and p-STAT3 expression in the Pathology Research Core. TMA slides were placed in the BOND III stainer (Leica Biosystems) for online processing. They were treated with Epitope Retrieval 2 Solution for 20 minutes and stained with NFATc2 (Abcam) or p-STAT3 (Cell Signaling Technology) for 15 minutes, and detection was achieved using the Polymer Refine Detection Kit per manufacturer's instructions (Leica Biosystems). Counterstaining was performed for 5 minutes with hematoxylin. Slides were dehydrated through increasing concentrations of alcohol, cleared in xylene, and cover-slipped in xylene-based mounting media.

Data analysis

The TMAs were evaluated for NFATc2 and p-STAT3 expression by a trained pancreatic pathologist and were scored as positive (score 1, 2, or 3) or negative (score 0). Stain positivity across the multiple evaluable cores per patient was reduced to 1 observation per unique subject by using the core, which stained with the maximum intensity for each stain.

Statistical analyses

Data were presented as averages ± SD or SE as noted and were analyzed by built-in t test using Microsoft Excel. P < 0.05 was considered significant.

GSK-3β confers pancreatic tumor growth stimulation by maintenance of NFAT expression levels through specific phosphorylation

To confirm and extend existing data on GSK-3β inhibition in pancreatic cancer, we applied a GSK-3β inhibitor (hereafter designated GSK-3i) to block kinase activity in different established pancreatic cancer cell lines. As expected, diminished GSK-3β activity was accompanied by a time- and dose-dependent decrease of cancer cell proliferation (Fig. 1A and B). We next treated nude mice bearing IMIM-PC1 xenograft tumors 3× weekly with GSK-3i and observed a significant tumor growth inhibition (Fig. 1C and Supplementary Fig. S1A), supporting the notion of a growth-promoting role of GSK-3β in pancreatic cancer.

Figure 1.

GSK-3β stabilizes NFATc2 in vitro and in vivo. A and B, PaTu8988t (A) and IMIM-PC1 (B) cells were treated with indicated concentrations of GSK-3β inhibitor AR-A014418 (hereafter referred to as GSK-3i) and analyzed for BrdUrd incorporation after 24 hours and 48 hours. C, nude mice bearing IMIM-PC1 xenograft tumors were treated 3× weekly with 10 mg/kg BW GSK-3i (n = 5) or 10% DMSO (n = 5) intraperitoneally. Tumors were extracted after 4 weeks. The tumor growth was normalized to the respective starting volumes. Means ± SE. *, P < 0.05; **, P < 0.01. D, PaTu8988t cells were starved and nuclear translocation of NFATc2 was induced by ionomycin treatment (0.5 μmol/L). Cells were then treated for 1 hour with 2.5, 25, and 50 μmol/L of GSK-3i and analyzed for NFATc2 and p-GS expression. Reduction of the well-described GSK-3β target p-GS serves as treatment control. E, PaTu8988t cells were transfected with GSK-3β siRNA, and lysates were tested for NFATc2 and GSK-3β expression after 48 hours. F and G, tumor lysates from three representative mice treated with 10% DMSO or GSK-3i were assessed for NFATc2 and p-GS (F) or NFATc2 and p-NFATc2 (G) expression as indicated. H, nuclear extracts from Suit-028 cells with stable expression of wt NFATc2 and NFATc2 pSP2 were treated for 3 hours with 25 μmol/L GSK-3i and then subjected to immunoblot analysis with the indicated antibodies. Lamin a/c serves as loading control. I, nuclear lysates from Suit-007 cells with stable expression of wt NFATc2 and NFATc2 pSP2 were subjected to DNA pulldown assays with oligonucleotides bearing the NFAT consensus sequence. DNA-bound NFATc2 was then analyzed by immunoblotting. J, PaTu8988t cells with stable wt NFATc2 and NFATc2 pSP2 expression or untransfected control cells were transfected with calcineurin and a reporter construct harboring three NFAT consensus binding sites (cisNFAT). Luciferase activity was measured after 24 hours. Displayed are means ± SD related to the control (first bar). K, nude mice were injected with PaTu8988t cells with stable wt NFATc2 or NFATc2 pSP2 expression or control cells, and tumor volumes were estimated after 4 weeks (n ≥ 5). Shown are means ± SE. *, P < 0.05; **, P < 0.01. L, representative IHC pictures of human PDA showing nuclear p-NFATc2 expression.

Figure 1.

GSK-3β stabilizes NFATc2 in vitro and in vivo. A and B, PaTu8988t (A) and IMIM-PC1 (B) cells were treated with indicated concentrations of GSK-3β inhibitor AR-A014418 (hereafter referred to as GSK-3i) and analyzed for BrdUrd incorporation after 24 hours and 48 hours. C, nude mice bearing IMIM-PC1 xenograft tumors were treated 3× weekly with 10 mg/kg BW GSK-3i (n = 5) or 10% DMSO (n = 5) intraperitoneally. Tumors were extracted after 4 weeks. The tumor growth was normalized to the respective starting volumes. Means ± SE. *, P < 0.05; **, P < 0.01. D, PaTu8988t cells were starved and nuclear translocation of NFATc2 was induced by ionomycin treatment (0.5 μmol/L). Cells were then treated for 1 hour with 2.5, 25, and 50 μmol/L of GSK-3i and analyzed for NFATc2 and p-GS expression. Reduction of the well-described GSK-3β target p-GS serves as treatment control. E, PaTu8988t cells were transfected with GSK-3β siRNA, and lysates were tested for NFATc2 and GSK-3β expression after 48 hours. F and G, tumor lysates from three representative mice treated with 10% DMSO or GSK-3i were assessed for NFATc2 and p-GS (F) or NFATc2 and p-NFATc2 (G) expression as indicated. H, nuclear extracts from Suit-028 cells with stable expression of wt NFATc2 and NFATc2 pSP2 were treated for 3 hours with 25 μmol/L GSK-3i and then subjected to immunoblot analysis with the indicated antibodies. Lamin a/c serves as loading control. I, nuclear lysates from Suit-007 cells with stable expression of wt NFATc2 and NFATc2 pSP2 were subjected to DNA pulldown assays with oligonucleotides bearing the NFAT consensus sequence. DNA-bound NFATc2 was then analyzed by immunoblotting. J, PaTu8988t cells with stable wt NFATc2 and NFATc2 pSP2 expression or untransfected control cells were transfected with calcineurin and a reporter construct harboring three NFAT consensus binding sites (cisNFAT). Luciferase activity was measured after 24 hours. Displayed are means ± SD related to the control (first bar). K, nude mice were injected with PaTu8988t cells with stable wt NFATc2 or NFATc2 pSP2 expression or control cells, and tumor volumes were estimated after 4 weeks (n ≥ 5). Shown are means ± SE. *, P < 0.05; **, P < 0.01. L, representative IHC pictures of human PDA showing nuclear p-NFATc2 expression.

Close modal

Interestingly, previous work by our group had pointed toward an oncogenic GSK-3β/NFATc2 stabilization pathway in pancreatic and breast cancer cells (31). To further investigate the GSK-3β/NFATc2 axis, we assessed NFATc2 expression levels in pancreatic cancer cell lines after pharmacologic inhibition or genetic depletion of GSK-3β. Treatment with the GSK-3i reduced nuclear NFATc2 protein expression in a dose-dependent fashion, as did transfection with a GSK-3β–specific siRNA (Fig. 1D and E and Supplementary Fig. S1B). Similarly, tissues derived from nude mice following GSK-3i treatment showed substantially less NFATc2 (Fig. 1F).

As GSK-3β was reported to specifically phosphorylate three conserved serines in the NFATc2 SP2 (S213, S217, and S221) region, we aimed to assess the functional relevance of this posttranslational modification. Importantly, a phospho-specific SP2 antibody detected wild-type NFATc2, but not a mutant NFATc2 lacking these phosphorylation sites (Supplementary Fig. S1C). Notably, whereas GSK-3β depletion resulted in diminished SP2 phosphorylation, expression of a constitutively active mutant (GSK-3β S9A) increased SP2 phosphorylation (Supplementary Fig. S1D and S1E). Immunoblot and immunohistologic analyses from GSK-3i–treated xenograft tumors revealed reduced levels of p-NFATc2, p-glycogen synthase (GS), and Ki67 (P < 0.01; Fig. 1G and Supplementary Fig. S1F and 1G). Moreover, nuclear NFATc2 protein levels were substantially reduced in pancreatic cancer cells treated with the GSK-3i, whereas overexpression of an NFATc2 mutant mimicking constitutive phosphorylation (NFATc2 pSP2) rescued the effect of GSK-3i treatment on NFATc2 expression (Fig. 1H and Supplementary Fig. S1H).

It is noteworthy that in immune cells, GSK-3β–mediated phosphorylation of the SP2 region terminated DNA binding of NFAT proteins and induced shuttling of NFAT proteins to the cytoplasm, followed by inactivation of NFAT activity (34–36). To address this issue in pancreatic cancer, we performed immunoblot analysis and DNA pulldown assays that detected high NFATc2 pSP2 levels in the nucleus as well as efficient binding to the NFAT consensus sequence GGAAA (Fig. 1I). Moreover, reporter gene assays demonstrated profound activation of an NFAT-responsive “cisNFAT” promoter by NFATc2 pSP2 compared with wild-type NFATc2 (Fig. 1J). Functionally, stable NFATc2 pSP2 expression significantly stimulated tumor growth of PaTu8988t xenograft tumors after 4 weeks (Fig. 1K). Immunohistochemical analysis of human PDA tissues showed nuclear NFATc2 expression (Fig. 1L). To extend and quantify this finding, we analyzed a human TMA consisting of 217 human pancreatic cancer tissues for NFATc2 expression and found a positive maximum nuclear tumor staining intensity in 96 of 217 (44.2%) of subjects, indicating the presence of active p-NFATc2, especially in neoplastic regions (Supplementary Table S1A). Collectively, our data suggest that contrary to the function in immune cells, SP2 phosphorylation by GSK-3β in pancreatic cancer cells maintains active NFATc2 protein in the nucleus, resulting in accelerated pancreatic cancer growth in vivo.

GSK-3β regulates NFATc2 activity through stabilization of complex formation with STAT3

Next, we assessed whether NFATc2 activity is also regulated by GSK-3β or whether the observed elevated NFATc2 pSP2 activation levels were due to increased protein levels following nuclear stabilization. Activation of the NFAT-responsive “cisNFAT” promoter correlated well with GSK-3β activity (Fig. 2A and B; Supplementary Fig. S2A and S2B). However, expression of the NFATc2 pSP2 construct, which is resistant to degradation upon GSK-3β inhibition, could not activate the cisNFAT promoter following GSK-3 depletion (Fig. 2A and Supplementary Fig. S2B). This was surprising, as we expected that recombinant NFATc2 pSP2 expression levels would overrule GSK-3β depletion. Moreover, expression of constitutively active GSK-3β (S9A) resulted in enhanced activity of the cisNFAT promoter when in the presence of wt or pSP2 NFATc1. Therefore, our data suggest that GSK-3β not only regulates NFATc2 protein stability through SP2 phosphorylation but also stimulates transcriptional activation by a distinct mechanism.

Figure 2.

SP2-independent regulation of NFATc2 activity by GSK-3β. Luciferase reporter gene assay demonstrating regulation of the cisNFAT promoter in PaTu8988t cells with stable wt NFATc2 and NFATc2 pSP2 expression or control cells following transfection with GSK-3β siRNA (A) or GSK-3β S9A (B). Means ± SD. C, PaTu8988t cells with stable expression of indicated constructs were transfected with wt STAT3 and cisNFAT and subjected to reporter gene assay. Shown are means ± SD normalized to controls. D, PaTu8988t cells with stable expression of indicated constructs were treated with 25 μmol/L GSK-3i, and after 3 hours nuclear lysates were subjected to coimmunoprecipitation and subsequent immunoblotting as indicated. E, reporter gene assay of Suit-007 cells with stable expression of indicated constructs after transfection with wt STAT3 and cisNFAT and subsequent administration of 25 μmol/L GSK-3i for 6 hours. Means ± SD. Successful knockdown of STAT3 is shown on right.

Figure 2.

SP2-independent regulation of NFATc2 activity by GSK-3β. Luciferase reporter gene assay demonstrating regulation of the cisNFAT promoter in PaTu8988t cells with stable wt NFATc2 and NFATc2 pSP2 expression or control cells following transfection with GSK-3β siRNA (A) or GSK-3β S9A (B). Means ± SD. C, PaTu8988t cells with stable expression of indicated constructs were transfected with wt STAT3 and cisNFAT and subjected to reporter gene assay. Shown are means ± SD normalized to controls. D, PaTu8988t cells with stable expression of indicated constructs were treated with 25 μmol/L GSK-3i, and after 3 hours nuclear lysates were subjected to coimmunoprecipitation and subsequent immunoblotting as indicated. E, reporter gene assay of Suit-007 cells with stable expression of indicated constructs after transfection with wt STAT3 and cisNFAT and subsequent administration of 25 μmol/L GSK-3i for 6 hours. Means ± SD. Successful knockdown of STAT3 is shown on right.

Close modal

We recently established an oncogenic NFATc1-STAT3 cooperativity governing pancreatic cancer progression (27). Thus, we were interested whether other NFAT family members such as NFATc2 also interact and cooperate with STAT3 to promote cancer growth and whether this cooperation is regulated by GSK-3β. We detected comparable and robust physical interaction of endogenous STAT3 with all NFATc2 SP2 mutants (Supplementary Fig. S2C). Correspondingly, STAT3 overexpression remarkably elevated NFATc2 activation of the cisNFAT promoter, irrespective of NFATc2 phosphorylation on SP2 (Fig. 2C). We next assessed whether this NFATc2/STAT3 complex formation is regulated by GSK-3β in pancreatic cancer. Significantly, GSK-3β inhibition disrupted the physical interaction of STAT3 with both wt and pSP2 NFATc2 (Fig. 2D). In addition, depletion of STAT3 abrogated the ability of GSK-3β S9A to enhance the transactivation of the cisNFAT promoter in the presence of wt NFATc2 (Fig. 2E). Moreover, STAT3 could not transactivate NFATc2 without GSK-3β activity (Supplementary Fig. S2D). Taken together, these data suggest that a functional NFATc2–STAT3 transcriptional complex requires the presence of GSK-3β activity not only to prevent NFATc2 nuclear degradation but also to engender NFATc2–STAT3 complex formation and transcriptional activity.

NFATc2 physically and functionally cooperates with canonical p-STAT3 (Y705) signaling to maintain cell growth

We next sought to characterize the NFATc2–STAT3 complex and its functional implication in pancreatic cancer. Immunohistochemical costaining of 217 human pancreatic cancer tissues revealed that 81 (37.3%) of cases were positive for both nuclear NFATc2 and nuclear p-STAT3 (Fig. 3A; Supplementary Table S1B). Remarkably, those tumors that showed NFATc2 expression stained positive for p-STAT3 in 84% (Fig. 3A). These observations were confirmed by immunofluorescence studies detecting simultaneous nuclear NFATc2 and STAT3 expression levels in pancreatic cancer cells (Fig. 3B). Interestingly, the inflammatory cytokine IL6 could induce a robust physical STAT3–NFATc2 interaction that was further enhanced by the addition of serum (Fig. 3C) and that required STAT3 Y705 phosphorylation (Fig. 3D). In addition, DNA pulldown assays demonstrated an interaction of the transcription factors on the NFAT consensus sequence that was also dependent on STAT3 phosphorylation (Fig. 3E). The expression of mutant STAT3 Y705F most notably antagonized the activating effect of wt STAT3 on the NFAT-responsive “cisNFAT” promoter, indicating that STAT3 phosphorylation is not only required for complex formation but also for efficient NFATc2 transactivation (Fig. 3F). It is noteworthy that GSK-3i treatment reduced STAT3 phosphorylation at Y705 in different pancreatic cancer cell lines (Supplementary Fig. S3A–S3C), suggesting that GSK-3β stabilizes NFATc2–STAT3 complexes by maintaining STAT3 phosphorylation. Finally, proliferation assays revealed that the loss of STAT3 expression reversed the NFATc2-dependent stimulation of cancer cell proliferation, highlighting its importance for NFATc2-induced growth stimulation (Fig. 3G). Accordingly, cell proliferation of PaTu8988t was reduced after STAT3 siRNA transfection, independent of SP2 phosphorylation (Supplementary Fig. S3D). In summary, p-STAT3 (Y705) efficiently binds to and transactivates NFATc2 on responsive promoters under mitogenic and inflammatory conditions, and in turn, acts to sustain NFATc2-dependent cell growth in a GSK-3β–dependent manner. Collectively, GSK-3β maintains NFATc2 activation through stabilization of NFATc2–STAT3 complexes independent of SP2 phosphorylation, presumably through phosphorylation of STAT3 at Y705.

Figure 3.

STAT3 Y705 phosphorylation is required for efficient physical and functional cooperation with NFATc2. A, TMA analysis of 217 cases of PDACs shows nuclear NFATc2 expression in 44% of cases, of which 84% also stained positive for p-STAT3. B, immunofluorescence staining displays nuclear colocalization of STAT3 (green) and HA-tagged NFATc2 (red) in PaTu8988t cells with stable expression of HA-NFATc2. Nuclei are visualized by DAPI staining (blue). C, Suit-007 cells with stable wt NFATc2 expression were transfected with Flag-tagged wt STAT3 and incubated with serum-free or serum medium for 24 hours with subsequent stimulation with IL6 (50 ng/mL). Lysates were subjected to coimmunoprecipitation and Western blotting as indicated. D, coimmunoprecipitation demonstrating reduced interaction of nuclear HA-tagged wt NFATc2 with Flag-tagged STAT3 Y705F compared with wt STAT3 in Suit-028 cells with stable HA-NFATc2 expression. E, PaTu8988t cells were transfected with STAT3 constructs as indicated and subjected to DNA pulldown assays with oligonucleotides bearing the NFAT consensus sequence. F, reporter gene assay demonstrating the inducibility of cisNFAT upon transfection of wt STAT3 and STAT3 Y705F in Suit-028 control cells and cells with stable expression of wt NFATc2 under serum-free conditions. Means ± SD. G, proliferation of Suit-007 cells with and without stable expression of wt NFATc2 following 72 hours of STAT3 siRNA transfection. Means ± SD. Efficient knockdown was tested by immunoblotting and is shown in Fig. 2E.

Figure 3.

STAT3 Y705 phosphorylation is required for efficient physical and functional cooperation with NFATc2. A, TMA analysis of 217 cases of PDACs shows nuclear NFATc2 expression in 44% of cases, of which 84% also stained positive for p-STAT3. B, immunofluorescence staining displays nuclear colocalization of STAT3 (green) and HA-tagged NFATc2 (red) in PaTu8988t cells with stable expression of HA-NFATc2. Nuclei are visualized by DAPI staining (blue). C, Suit-007 cells with stable wt NFATc2 expression were transfected with Flag-tagged wt STAT3 and incubated with serum-free or serum medium for 24 hours with subsequent stimulation with IL6 (50 ng/mL). Lysates were subjected to coimmunoprecipitation and Western blotting as indicated. D, coimmunoprecipitation demonstrating reduced interaction of nuclear HA-tagged wt NFATc2 with Flag-tagged STAT3 Y705F compared with wt STAT3 in Suit-028 cells with stable HA-NFATc2 expression. E, PaTu8988t cells were transfected with STAT3 constructs as indicated and subjected to DNA pulldown assays with oligonucleotides bearing the NFAT consensus sequence. F, reporter gene assay demonstrating the inducibility of cisNFAT upon transfection of wt STAT3 and STAT3 Y705F in Suit-028 control cells and cells with stable expression of wt NFATc2 under serum-free conditions. Means ± SD. G, proliferation of Suit-007 cells with and without stable expression of wt NFATc2 following 72 hours of STAT3 siRNA transfection. Means ± SD. Efficient knockdown was tested by immunoblotting and is shown in Fig. 2E.

Close modal

We next analyzed the GSK-3β–dependent NFATc2/STAT3 interaction on the previously described NFATc2 target promoter, CDK-6 (Fig. 4A and B). Importantly, GSK-3i treatment resulted not only in the loss of NFATc2 binding to the CDK-6 promoter but also reduced the binding of STAT3 (Fig. 4C). Furthermore, binding of RNA-polymerase II and the activating histone modification H3K4-me3 was significantly decreased upon GSK-3i administration (Fig. 4C), followed by loss of CDK-6 gene and protein expression (Fig. 4D and E). Consistent with the role of GSK-3β in promoting the transactivation of NFATc2–STAT3 complexes, CDK-6 expression was decreased after upon GSK-3i treatment (Supplementary Fig. S3E). Taken together, our data strongly point to a multilevel regulatory impact of GSK-3β on NFATc2, which is, on one hand, mediated by SP2 phosphorylation-dependent stabilization of protein levels in the nucleus and on the other hand by recruitment of NFATc2–STAT3 complexes to target promoters and subsequent transcription of cell-cycle promoters independent of SP2 phosphorylation.

Figure 4.

Stimulation of euchromatin formation on CDK-6 target promoter through NFATc2–STAT3 complexes. A, transcription start site (TSS) binding motifs for NFAT and STAT3. B, human CDK-6 promoter with putative NFATc2 and STAT3 binding sites. C, chromatin immunoprecipitation analysis determines NFATc2, STAT3, RNA-polymerase II (RNA-Pol II), and H3K4me3 binding to the CDK-6 promoter with and without AR-A014418 (GSK-3i) treatment (25 μmol/L for 12 hours). Means ± SD. D, qPCR illustrating reduced CDK-6 mRNA expression in PaTu8988t cells following GSK-3β inhibition with GSK-3i (25 μmol/L for 3 hours). E, lysates from PaTu8988t cells with stable expression of GSK-3β S9A or following transfection with GSK-3β siRNA were subjected to immunoblotting with indicated antibodies.

Figure 4.

Stimulation of euchromatin formation on CDK-6 target promoter through NFATc2–STAT3 complexes. A, transcription start site (TSS) binding motifs for NFAT and STAT3. B, human CDK-6 promoter with putative NFATc2 and STAT3 binding sites. C, chromatin immunoprecipitation analysis determines NFATc2, STAT3, RNA-polymerase II (RNA-Pol II), and H3K4me3 binding to the CDK-6 promoter with and without AR-A014418 (GSK-3i) treatment (25 μmol/L for 12 hours). Means ± SD. D, qPCR illustrating reduced CDK-6 mRNA expression in PaTu8988t cells following GSK-3β inhibition with GSK-3i (25 μmol/L for 3 hours). E, lysates from PaTu8988t cells with stable expression of GSK-3β S9A or following transfection with GSK-3β siRNA were subjected to immunoblotting with indicated antibodies.

Close modal

Targeting the NFATc2–STAT3–GSK-3β cooperativity interferes with inflammation-induced pancreatic cancer progression in KrasG12D mice

To assess the therapeutic potential of targeting the identified NFATc2–STAT3–GSK-3β cooperativity in vivo, we utilized a well-established model of pancreatitis-associated carcinogenesis to ensure profound induction of NFATc2 and STAT3, as these transcription factors were shown to be overexpressed or activated in the majority of Kras-mutated tumor specimen, especially upon inflammatory conditions (37, 38). In line with previously published data, we observed that cancer progression was massively accelerated when transgenic mice with pancreas-specific expression of oncogenic KrasG12D were challenged 3× weekly with the CCK analogue caerulein to induce a persistent organ inflammation (9, 10). Mice developed the full spectrum of precursor lesions ranging from noninvasive PanIN1 to PanIN2/3 and carcinoma in situ lesions (Fig. 5A and B). This was accompanied by induced p-NFATc2 (SP2) and p-STAT3 (Y705) levels (Fig. 5C and D). When mice were additionally treated with a GSK-3i, however, cancer progression was significantly blocked in terms of proliferation, neoplastic transformation, and tumor incidence, as evidenced by restored normal ducts, decreased precursor lesions (Fig. 5B), and significantly reduced Ki67 expression levels (Fig. 5E). Moreover, we observed a reduced tendency to develop tumors upon GSK-3i treatment (30% after caerulein; n = 7 vs. 0% after caerulein and GSK-3i; n = 8). Finally, induction of p-NFATc2 and p-STAT3 was antagonized upon treatment with GSK-3i (Fig. 5C and D), clearly highlighting the regulatory impact of GSK-3 on NFATc2/STAT3–mediated cancer progression and growth. Interestingly, increased immune cell infiltrations upon caerulein treatment were markedly reduced upon GSK-3 inhibition (Supplementary Fig. S4).

Figure 5.

Targeting GSK-3 prolongs pancreatic cancer progression in KrasG12D mice. A, representative H&E staining in Pdx1;KrasG12D mice after treatment with PBS (n = 10) caerulein (n = 7) or caerulein along with the GSK-3 inhibitor 9-ING-41 (designated GSK-3i; n = 8), showing retarded tumor progression upon GSK-3 inhibition. Scale bars, 100 μmol/L (top) and 200 μmol/L (bottom). B, quantification of normal and preneoplastic ducts in Pdx1;KrasG12D mice upon indicated treatment. Means ± SD (n ≥ 6). *, P values are related to untreated Pdx1;KrasG12D cohorts and Δ values indicate caerulein versus caerulein and GSK-3i treatment. * and Δ, P < 0.05; ** and ΔΔ, P < 0.01; *** and ΔΔΔ, P < 0.001. C, immunohistologic p-STAT3, p-NFATc2, and Ki67 staining in Pdx1;KrasG12D mice after treatment as indicated. Scale bars, 100 μmol/L. D, tissues from Pdx1;KrasG12D mice after treatment as indicated were assayed for p-STAT3, STAT3, p-NFATc2, and NFATc2 expression. E, proliferation index was measured in Ki67-stained pancreatic sections (n ≥ 3) of Pdx1;KrasG12D mice after indicated treatment. Means ± SE. *, P < 0.05; ***, P < 0.001.

Figure 5.

Targeting GSK-3 prolongs pancreatic cancer progression in KrasG12D mice. A, representative H&E staining in Pdx1;KrasG12D mice after treatment with PBS (n = 10) caerulein (n = 7) or caerulein along with the GSK-3 inhibitor 9-ING-41 (designated GSK-3i; n = 8), showing retarded tumor progression upon GSK-3 inhibition. Scale bars, 100 μmol/L (top) and 200 μmol/L (bottom). B, quantification of normal and preneoplastic ducts in Pdx1;KrasG12D mice upon indicated treatment. Means ± SD (n ≥ 6). *, P values are related to untreated Pdx1;KrasG12D cohorts and Δ values indicate caerulein versus caerulein and GSK-3i treatment. * and Δ, P < 0.05; ** and ΔΔ, P < 0.01; *** and ΔΔΔ, P < 0.001. C, immunohistologic p-STAT3, p-NFATc2, and Ki67 staining in Pdx1;KrasG12D mice after treatment as indicated. Scale bars, 100 μmol/L. D, tissues from Pdx1;KrasG12D mice after treatment as indicated were assayed for p-STAT3, STAT3, p-NFATc2, and NFATc2 expression. E, proliferation index was measured in Ki67-stained pancreatic sections (n ≥ 3) of Pdx1;KrasG12D mice after indicated treatment. Means ± SE. *, P < 0.05; ***, P < 0.001.

Close modal

Pancreatic cancer exhibits a high therapeutic resistance against cytotoxic and apoptosis-inducing agents (39), and although genetic approaches targeting Kras in mice have shown encouraging results (40, 41), the oncogene is still deemed “undruggable” in mice and man. Persistent inflammation contributes to tumor progression and therapeutic resistance in pancreatic cancer, and the activation of proinflammatory cytokines and transcription factors is a molecular feature of this disease. Although GSK-3β is not induced by inflammation itself, it governs a variety of inflammatory responses such as enhanced cytokine production driven by NF-κB and STAT3 pathways (21–24). Interestingly, the anti-inflammatory potential of GSK-3 inhibition was evidenced in our study by a clear reduction of immune cell infiltrations in GEMM of pancreas cancer.

NFAT proteins represent another class of major proinflammatory transcription factors that were originally described as inducible factors binding to the IL2 promoter in activated T cells (42). Strongly induced upon inflammatory stimuli, NFAT proteins have been shown to be overexpressed in the majority of invasively growing cancers, presumably in those with persistent inflammation, and to dramatically accelerate Kras-driven carcinogenesis in the pancreas (27, 28, 30). However, as therapeutic targeting of transcription factors is difficult, it is critical to identify upstream regulatory pathways that may be targetable for cancer therapeutics.

Here, we investigated the role of GSK-3β on the regulation and activation of NFATc2 in pancreatic cancer progression and growth. We propose the following model: in contrast to results in immune cells, where GSK-3β phosphorylates three serine residues in the N-terminal SP2 domain and thus terminates DNA binding of NFAT (34–36), we show that specific phosphorylation of the SP2 region of NFATc2 by GSK-3β induces NFATc2 protein stability in the nucleus of pancreatic cancer cells (Fig. 6). Thus, we extend our existing knowledge of the GSK-3β–NFATc2 stabilization pathway by uncovering nuclear activation of p-NFATc2 as a potentially specific diagnostic tool and therapeutic target, as targeting p-NFATc2 would supposedly not harm NFAT signaling in immune cells. However, protein stability itself is not the exclusive mechanism governing robust protumorigenic effects driven by the GSK-3β–NFATc2 axis. In fact, our data highlight that GSK-3β orchestrates an SP2 phosphorylation–independent mechanism involving NFATc2–STAT3 complex formation that is required to stimulate euchromatin formation of NFAT target promoters, such as CDK-6. This is likely dominant over SP2-dependent NFATc2 protein stabilization, as it can be disrupted through GSK-3β inhibition, even in the presence of p-NFATc2.

Figure 6.

Proposed model of GSK-3β–mediated regulation of the NFATc2 metabolism in pancreatic cancer cells. Activation of the phosphatase calcineurin following an intracellular calcium influx dephosphorylates NFAT proteins and stimulates their nuclear import. There, GSK-3β–mediated SP2 phosphorylation protects NFATc2 from degradation and further stimulates DNA binding and transcriptional activation of target genes (left). In addition, GSK-3β stabilizes NFATc2/STAT3 complex formation independent of and dominant to SP2 phosphorylation resulting in target gene expression (right).

Figure 6.

Proposed model of GSK-3β–mediated regulation of the NFATc2 metabolism in pancreatic cancer cells. Activation of the phosphatase calcineurin following an intracellular calcium influx dephosphorylates NFAT proteins and stimulates their nuclear import. There, GSK-3β–mediated SP2 phosphorylation protects NFATc2 from degradation and further stimulates DNA binding and transcriptional activation of target genes (left). In addition, GSK-3β stabilizes NFATc2/STAT3 complex formation independent of and dominant to SP2 phosphorylation resulting in target gene expression (right).

Close modal

In addition, our observations suggest that the activity of NFAT proteins is not only executed by a variety of phosphorylation events (some GSK-3β–mediated) but to a great extent by dynamic and complex protein–protein interactions influencing and fine-tuning DNA binding and specificity to target promoters. This cooperation with partner proteins serves to stabilize and modulate DNA binding of NFAT proteins, thus generating a sophisticated axis of interdependencies and feedback loops. STAT3 proteins are aberrantly activated in most cancers, and recently emerged as potent mediators of pancreatic malignant transformation and cancer progression, especially under inflammatory conditions (43, 44). In line with these observations, we have previously shown that NFATc1 induces STAT3 expression with subsequent formation of NFATc1/STAT3 nucleoprotein complexes in pancreatic cancer cells. Likewise, STAT3 affected NFATc1 transcriptional activity by maintaining its DNA binding on distant enhancers and hence stimulated enhancer–promoter communication leading to the expression of oncogenes such as EGFR, cyclin D3, or Wnt proteins (27, 28, 30).

From a therapeutic point of view, targeting of central upstream regulators, and subsequently the complete proinflammatory oncogenic network, appears feasible and may help overcome drug resistance and apoptosis inhibition. Notably, caution has been raised in the therapeutic field owing to the putative tumor suppressive functions of GSK-3β as inhibition may result in the activation of the Wnt pathway and subsequent tumorigenesis. Reassuringly, however, Wnt signaling and β-catenin nuclear accumulation are not perturbed in GSK-3β–deficient mice (14). Also, lithium, a known inhibitor of GSK-3, causes a reduced cancer morbidity in psychiatric patients compared with the general population and inhibits proliferation in hepatocellular carcinoma cell lines (45, 46).

Collectively, our data suggest that GSK-3β fine-tunes NFATc2 and STAT3 transcriptional networks to integrate upstream signaling events to eventually govern pancreatic cancer progression and growth. Furthermore, the therapeutic potential of GSK-3β to delay inflammation-induced malignant transformation is demonstrated for the first time in a relevant GEMM for pancreatic cancer. In a broader sense, our data add to the molecular understanding of GSK-3β–mediated tumorigenesis and may thus open new avenues for individually tailored therapies. For instance, one can imagine that patients with high expression of NFATc2 may benefit the most from GSK-3β inhibitor-based therapy. Our results are particularly germane given the fact that a large number of clinical GSK-3β inhibitors are available and currently being tested in several disease entities, including pancreatic cancer patients.

A.P. Kozikowski is the owner and founder of, has ownership interest (including patents) in, and is a consultant/advisory board member for Actuate Therapeutics. No potential conflicts of interest were disclosed by the other authors.

Conception and design: S. Baumgart, N.-M. Chen, J.-S. Zhang, S.K Singh, V. Ellenrieder, A. Neesse

Development of methodology: N.-M. Chen, J.-S. Zhang, A.P. Kozikowski

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): S. Baumgart, N.-M. Chen, J.-S. Zhang, I.N. Gaisina, S.K. Singh, D. Fink, C. Klindt, E. Hessmann, T.M. Gress, A. Neesse

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): S. Baumgart, N.-M. Chen, S.K. Singh, D. Fink, P. Ströbel, L. Zhang, W.R. Bamlet, A. Koenig, E. Hessmann, A. Neesse

Writing, review, and/or revision of the manuscript: S. Baumgart, N.-M. Chen, J.-S. Zhang, D.D. Billadeau, A.P. Kozikowski, P. Ströbel, A. Koenig, E. Hessmann, A. Neesse

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): N.-M. Chen, T.M. Gress

Study supervision: N.-M. Chen, S.K. Singh, V. Ellenrieder, A. Neesse

This work was supported in part by the NCI Pancreatic Cancer SPORE grant P50CA102701 (to D.D. Billadeau), by a Max Eder Fellowship (110972) Deutsche Krebshilfe (to A. Neesse), and by the National Natural Science Foundation of China (grant number 81472601, to J.S. Zhang).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Siegel
R
,
Naishadham
D
,
Jemal
A
. 
Cancer statistics, 2013
.
CA Cancer J Clin
2013
;
63
:
11
30
.
2.
Almoguera
C
,
Shibata
D
,
Forrester
K
,
Martin
J
,
Arnheim
N
,
Perucho
M
. 
Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes
.
Cell
1988
;
53
:
549
54
.
3.
Ardito
CM
,
Gruner
BM
,
Takeuchi
KK
,
Lubeseder-Martellato
C
,
Teichmann
N
,
Mazur
PK
, et al
EGF receptor is required for KRAS-induced pancreatic tumorigenesis
.
Cancer Cell
2012
;
22
:
304
17
.
4.
Bayne
LJ
,
Beatty
GL
,
Jhala
N
,
Clark
CE
,
Rhim
AD
,
Stanger
BZ
, et al
Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer
.
Cancer Cell
2012
;
21
:
822
35
.
5.
Eser
S
,
Reiff
N
,
Messer
M
,
Seidler
B
,
Gottschalk
K
,
Dobler
M
, et al
Selective requirement of PI3K/PDK1 signaling for Kras oncogene-driven pancreatic cell plasticity and cancer
.
Cancer Cell
2013
;
23
:
406
20
.
6.
Kapoor
A
,
Yao
W
,
Ying
H
,
Hua
S
,
Liewen
A
,
Wang
Q
, et al
Yap1 activation enables bypass of oncogenic kras addiction in pancreatic cancer
.
Cell
2014
;
158
:
185
97
.
7.
Pylayeva-Gupta
Y
,
Lee
KE
,
Hajdu
CH
,
Miller
G
,
Bar-Sagi
D
. 
Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia
.
Cancer Cell
2012
;
21
:
836
47
.
8.
Zhu
Z
,
Aref
AR
,
Cohoon
TJ
,
Barbie
TU
,
Imamura
Y
,
Yang
S
, et al
Inhibition of KRAS-driven tumorigenicity by interruption of an autocrine cytokine circuit
.
Cancer Discov
2014
;
4
:
452
65
.
9.
Guerra
C
,
Collado
M
,
Navas
C
,
Schuhmacher
AJ
,
Hernandez-Porras
I
,
Canamero
M
, et al
Pancreatitis-induced inflammation contributes to pancreatic cancer by inhibiting oncogene-induced senescence
.
Cancer Cell
2011
;
19
:
728
39
.
10.
Guerra
C
,
Schuhmacher
AJ
,
Canamero
M
,
Grippo
PJ
,
Verdaguer
L
,
Perez-Gallego
L
, et al
Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice
.
Cancer Cell
2007
;
11
:
291
302
.
11.
Lowenfels
AB
,
Maisonneuve
P
. 
Pancreatic disease: does “itis” lead to “oma”?
Gastroenterology
1998
;
114
:
859
61
.
12.
Zhang
JS
,
Koenig
A
,
Harrison
A
,
Ugolkov
AV
,
Fernandez-Zapico
ME
,
Couch
FJ
, et al
Mutant K-Ras increases GSK-3beta gene expression via an ETS-p300 transcriptional complex in pancreatic cancer
.
Oncogene
2011
;
30
:
3705
15
.
13.
Doble
BW
,
Woodgett
JR
. 
GSK-3: tricks of the trade for a multi-tasking kinase
.
J Cell Sci
2003
;
116
:
1175
86
.
14.
Hoeflich
KP
,
Luo
J
,
Rubie
EA
,
Tsao
MS
,
Jin
O
,
Woodgett
JR
. 
Requirement for glycogen synthase kinase-3beta in cell survival and NF-kappaB activation
.
Nature
2000
;
406
:
86
90
.
15.
Polakis
P
. 
Wnt signaling and cancer
.
Genes Dev
2000
;
14
:
1837
51
.
16.
Bang
D
,
Wilson
W
,
Ryan
M
,
Yeh
JJ
,
Baldwin
AS
. 
GSK-3alpha promotes oncogenic KRAS function in pancreatic cancer via TAK1-TAB stabilization and regulation of noncanonical NF-kappaB
.
Cancer Discov
2013
;
3
:
690
703
.
17.
Kitano
A
,
Shimasaki
T
,
Chikano
Y
,
Nakada
M
,
Hirose
M
,
Higashi
T
, et al
Aberrant glycogen synthase kinase 3beta is involved in pancreatic cancer cell invasion and resistance to therapy
.
PLoS One
2013
;
8
:
e55289
.
18.
Ougolkov
AV
,
Fernandez-Zapico
ME
,
Bilim
VN
,
Smyrk
TC
,
Chari
ST
,
Billadeau
DD
. 
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
81
.
19.
Ougolkov
AV
,
Fernandez-Zapico
ME
,
Savoy
DN
,
Urrutia
RA
,
Billadeau
DD
. 
Glycogen synthase kinase-3beta participates in nuclear factor kappaB-mediated gene transcription and cell survival in pancreatic cancer cells
.
Cancer Res
2005
;
65
:
2076
81
.
20.
Shimasaki
T
,
Kitano
A
,
Motoo
Y
,
Minamoto
T
. 
Aberrant glycogen synthase kinase 3beta in the development of pancreatic cancer
.
J Carcinog
2012
;
11
:
15
.
21.
Beurel
E
,
Jope
RS
. 
Glycogen synthase kinase-3 regulates inflammatory tolerance in astrocytes
.
Neuroscience
2010
;
169
:
1063
70
.
22.
Beurel
E
,
Yeh
WI
,
Michalek
SM
,
Harrington
LE
,
Jope
RS
. 
Glycogen synthase kinase-3 is an early determinant in the differentiation of pathogenic Th17 cells
.
J Immunol
2011
;
186
:
1391
8
.
23.
Espinosa
L
,
Ingles-Esteve
J
,
Aguilera
C
,
Bigas
A
. 
Phosphorylation by glycogen synthase kinase-3 beta down-regulates Notch activity, a link for Notch and Wnt pathways
.
J Biol Chem
2003
;
278
:
32227
35
.
24.
Kaidanovich-Beilin
O
,
Woodgett
JR
. 
GSK-3: functional insights from cell biology and animal models
.
Front Mol Neurosci
2011
;
4
:
40
.
25.
Zhang
JS
,
Herreros-Vilanueva
M
,
Koenig
A
,
Deng
Z
,
de Narvajas
AA
,
Gomez
TS
, 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
.
26.
Singh
SK
,
Chen
NM
,
Hessmann
E
,
Siveke
J
,
Lahmann
M
,
Singh
G
, et al
Antithetical NFATc1-Sox2 and p53-miR200 signaling networks govern pancreatic cancer cell plasticity
.
EMBO J
2015
;
34
:
517
30
.
27.
Baumgart
S
,
Chen
NM
,
Siveke
JT
,
Konig
A
,
Zhang
JS
,
Singh
SK
, et al
Inflammation-induced NFATc1-STAT3 transcription complex promotes pancreatic cancer initiation by KrasG12D
.
Cancer Discov
2014
;
4
:
688
701
.
28.
Baumgart
S
,
Glesel
E
,
Singh
G
,
Chen
NM
,
Reutlinger
K
,
Zhang
J
, et al
Restricted heterochromatin formation links NFATc2 repressor activity with growth promotion in pancreatic cancer
.
Gastroenterology
2012
;
142
:
388
98
.
29.
Buchholz
M
,
Schatz
A
,
Wagner
M
,
Michl
P
,
Linhart
T
,
Adler
G
, et al
Overexpression of c-myc in pancreatic cancer caused by ectopic activation of NFATc1 and the Ca2+/calcineurin signaling pathway
.
EMBO J
2006
;
25
:
3714
24
.
30.
Koenig
A
,
Linhart
T
,
Schlengemann
K
,
Reutlinger
K
,
Wegele
J
,
Adler
G
, et al
NFAT-induced histone acetylation relay switch promotes c-Myc-dependent growth in pancreatic cancer cells
.
Gastroenterology
2010
;
138
:
1189
99
.
31.
Singh
SK
,
Baumgart
S
,
Singh
G
,
Konig
AO
,
Reutlinger
K
,
Hofbauer
LC
, et al
Disruption of a nuclear NFATc2 protein stabilization loop confers breast and pancreatic cancer growth suppression by zoledronic acid
.
J Biol Chem
2011
;
286
:
28761
71
.
32.
Yoeli-Lerner
M
,
Chin
YR
,
Hansen
CK
,
Toker
A
. 
Akt/protein kinase b and glycogen synthase kinase-3beta signaling pathway regulates cell migration through the NFAT1 transcription factor
.
Mol Cancer Res
2009
;
7
:
425
32
.
33.
Pal
K
,
Cao
Y
,
Gaisina
IN
,
Bhattacharya
S
,
Dutta
SK
,
Wang
E
, et al
Inhibition of GSK-3 induces differentiation and impaired glucose metabolism in renal cancer
.
Mol Cancer Ther
2014
;
13
:
285
96
.
34.
Neal
JW
,
Clipstone
NA
. 
Glycogen synthase kinase-3 inhibits the DNA binding activity of NFATc
.
J Biol Chem
2001
;
276
:
3666
73
.
35.
Beals
CR
,
Sheridan
CM
,
Turck
CW
,
Gardner
P
,
Crabtree
GR
. 
Nuclear export of NF-ATc enhanced by glycogen synthase kinase-3
.
Science
1997
;
275
:
1930
4
.
36.
Beals
CR
,
Clipstone
NA
,
Ho
SN
,
Crabtree
GR
. 
Nuclear localization of NF-ATc by a calcineurin-dependent, cyclosporin-sensitive intramolecular interaction
.
Genes Dev
1997
;
11
:
824
34
.
37.
Corcoran
RB
,
Contino
G
,
Deshpande
V
,
Tzatsos
A
,
Conrad
C
,
Benes
CH
, et al
STAT3 plays a critical role in KRAS-induced pancreatic tumorigenesis
.
Cancer Res
2011
;
71
:
5020
9
.
38.
Navas
C
,
Hernandez-Porras
I
,
Schuhmacher
AJ
,
Sibilia
M
,
Guerra
C
,
Barbacid
M
. 
EGF receptor signaling is essential for k-ras oncogene-driven pancreatic ductal adenocarcinoma
.
Cancer Cell
2012
;
22
:
318
30
.
39.
Neesse
A
,
Frese
KK
,
Bapiro
TE
,
Nakagawa
T
,
Sternlicht
MD
,
Seeley
TW
, et al
CTGF antagonism with mAb FG-3019 enhances chemotherapy response without increasing drug delivery in murine ductal pancreas cancer
.
Proc Natl Acad Sci U S A
2013
;
110
:
12325
30
.
40.
Collins
MA
,
Bednar
F
,
Zhang
Y
,
Brisset
JC
,
Galban
S
,
Galban
CJ
, et al
Oncogenic Kras is required for both the initiation and maintenance of pancreatic cancer in mice
.
J Clin Invest
2012
;
122
:
639
53
.
41.
Ying
H
,
Kimmelman
AC
,
Lyssiotis
CA
,
Hua
S
,
Chu
GC
,
Fletcher-Sananikone
E
, et al
Oncogenic Kras maintains pancreatic tumors through regulation of anabolic glucose metabolism
.
Cell
2012
;
149
:
656
70
.
42.
Shaw
J
,
Meerovitch
K
,
Bleackley
RC
,
Paetkau
V
. 
Mechanisms regulating the level of IL-2 mRNA in T lymphocytes
.
J Immunol
1988
;
140
:
2243
8
.
43.
Lesina
M
,
Kurkowski
MU
,
Ludes
K
,
Rose-John
S
,
Treiber
M
,
Kloppel
G
, et al
Stat3/Socs3 activation by IL-6 transsignaling promotes progression of pancreatic intraepithelial neoplasia and development of pancreatic cancer
.
Cancer Cell
2011
;
19
:
456
69
.
44.
Scholz
A
,
Heinze
S
,
Detjen
KM
,
Peters
M
,
Welzel
M
,
Hauff
P
, et al
Activated signal transducer and activator of transcription 3 (STAT3) supports the malignant phenotype of human pancreatic cancer
.
Gastroenterology
2003
;
125
:
891
905
.
45.
Cohen
Y
,
Chetrit
A
,
Cohen
Y
,
Sirota
P
,
Modan
B
. 
Cancer morbidity in psychiatric patients: influence of lithium carbonate treatment
.
Med Oncol
1998
;
15
:
32
6
.
46.
Erdal
E
,
Ozturk
N
,
Cagatay
T
,
Eksioglu-Demiralp
E
,
Ozturk
M
. 
Lithium-mediated downregulation of PKB/Akt and cyclin E with growth inhibition in hepatocellular carcinoma cells
.
Int J Cancer
2005
;
115
:
903
10
.