Abstract
The receptor tyrosine kinase AXL promotes tumor progression, metastasis, and therapy resistance through the induction of epithelial-mesenchymal transition (EMT). Here, we found that activation of AXL resulted in the phosphorylation of TANK-binding kinase 1 (TBK1) and the downstream activation of AKT3 and Snail, a transcription factor critical for EMT. Mechanistically, we showed that TBK1 directly bound to and phosphorylated AKT3 in a manner dependent on the multiprotein complex mTORC1. Upon activation, AKT3 interacted with and promoted the nuclear accumulation of Snail, which led to increased EMT as assessed by marker abundance. In human pancreatic ductal adenocarcinoma tissue, nuclear AKT3 colocalized with Snail and correlated with worse clinical outcomes. Primary mouse pancreatic cancer cells deficient in AKT3 showed reduced metastatic spread in vivo, suggesting selective AKT3 inhibition as a potential therapeutic avenue for targeting EMT in aggressive cancers.
INTRODUCTION
Cancer metastasis, the leading cause of cancer mortality, correlates with epithelial plasticity/epithelial-to-mesenchymal transition (EMT) (1, 2). Metastasis of epithelial tumors, such as pancreatic cancer (PDA), requires cancer cells to escape epithelial nests, invade surrounding stroma, intravasate into blood or lymphatic vessels, survive circulation, and extravasate at a secondary site, where successful cells form micrometastases and eventually macrometastases (3). The escape of tumor cells from tumor cell nests encapsulated by a basement membrane can be facilitated by tumor cell epithelial plasticity, which results in epithelial tumor cells losing contact with the basement membrane and nearby cells while adopting mesenchymal-like features that enhance cell migration and invasion. Whereas epithelial plasticity alters morphology and cell-cell contact, it also enables tumor cell survival under stressful environmental conditions, such as chemotherapy and radiation (4–7).
In carcinomas, the activation of an EMT program is associated with tumor grade. High-grade cancer is characterized by a loss of normal tissue structure and architecture. High-grade tumors are often described as poorly differentiated, displaying tumor cells that have undergone full or partial EMT. In contrast, low-grade tumors that retain an epithelial phenotype are characterized as well differentiated. Across human cancer, tumors that are high grade and poorly differentiated carry a worse prognosis with a high likelihood of metastasizing to distant organs (8). Understanding the molecular mechanisms underlying cellular plasticity and metastasis may reveal previously unidentified ways to target these programs for effective therapies.
Many signaling pathways can mediate tumor cell epithelial plasticity, including the receptor tyrosine kinase (RTK) AXL (9–11), increased expression of which correlates with metastasis and resistance to therapy (9, 12). AXL is a member of the TAM (Tyro3, AXL, MerTK) family of RTKs (13) and is activated by its ligand, growth arrest–specific gene 6 (Gas6), to promote a variety of cellular processes, including epithelial plasticity, cell survival, proliferation, and migration (9). The serine threonine protein kinase TANK-binding kinase 1 (TBK1) promotes EMT downstream of AXL in PDA, providing insight into a function for TBK1 (14). Whereas the precise mechanism of how TBK1 drives EMT has yet to be determined, previous work found that TBK1 can directly activate AKT (15).
AKT is a key regulator of many cellular phenotypes associated with cancer, including cell survival, proliferation, and metastasis (16). Activation of AKT can drive EMT via the induction of EMT transcription factors (EMT-TFs) including Slug and Snail, which transcriptionally repress E-cadherin and induce vimentin, twist1, matrix metalloproteinase–2 (MMP-2), and MMP-9 that promote tumor cell invasion (7, 17, 18). There are three mammalian AKT isoforms: AKT1, AKT2, and AKT3. Whereas each isoform is encoded by a distinct gene, there is ~80% amino acid sequence identity, and each isoform appears to be activated by similar mechanisms (19, 20). Although the general function of AKT in cancer cell survival and growth has been well characterized, the contribution of different AKT isoforms has not been investigated as intensely and is often underappreciated. On the basis of a phosphoproteomics screen, AKT isoforms have specific expression patterns and serve different functions in cell signaling and cancer (21).
In this study, we investigated the contribution of each AKT isoform in AXL-mediated signaling and report that the AKT3 isoform is phosphorylated in a TBK1-dependent manner upon Gas6-mediated AXL stimulation, which resulted in increased accumulation of phosphorylated AKT3 (pAKT3) and Snail in the nucleus. We showed that AKT3 colocalized and interacted with TBK1 and Snail and that TBK1:AKT3:Snail complexes coimmunoprecipitated from nuclear fraction of cell lysates. Furthermore, in human tissues, nuclear AKT3 colocalized with Snail and correlated with aggressive PDA and breast cancer. In addition, CRISPR-Cas9 knockout (KO) of AKT3 in tumor cells reduced metastasis in mice, suggesting that selective AKT3 targeting represents a therapeutic avenue for treating aggressive cancer.
RESULTS
AXL activation induces TBK1-dependent AKT3 activation
For these studies, we used PDA cells where AXL activation has been shown to promote tumor cell migration and invasion (22). We and others have shown that AKT is activated downstream of AXL (14). To determine which AKT isoform is most associated with AXL-driven epithelial plasticity, we first evaluated the expression of AXL, AKT isoforms, and EMT markers in publicly available GEO RNA sequencing data of PDA cell lines (Fig. 1A). This analysis revealed a stronger correlation between AXL and AKT3 compared with the other AKT isoforms. This was corroborated by the results of gene expression profiling interactive analysis (GEPIA) for AKT isoforms and AXL in human PDA using the TCGA database (Fig. 1B) (23), where AKT3 and AXL expression showed the best correlation. Considering that increased AXL expression has been shown in other types of cancer, we examined expression profiles of AXL, EMT markers, and AKT isoforms in breast cancer. Up-regulated AXL expression correlated with expression of EMT markers N-cadherin and Twist2 by immunohistochemistry (IHC) in primary breast tumors (fig. S1A). Similar to PDA cells, AXL expression strongly correlated with AKT3, but not AKT1 or AKT2, expression in breast cancer cell lines and breast tumors (fig. S1, B and C). Moreover, in MCF10a breast epithelial cells, forced expression of Slug increased AKT3 expression and pAKT in an AXL-dependent manner with no effect on AKT1 expression (fig. S1D). These data suggest that AXL expression is essential for accumulation and activation of AKT3.
Fig. 1. AKT isoforms in AXL-TBK1 signaling.
(A) Gene coexpression analysis for AKT isoforms and EMT and stem cell–associated genes in pancreas cancer cell lines. Red and blue show positive and negative correlation for mRNA levels, respectively. (B) Gene coexpression analysis for AKT isoforms and AXL using GEPIA in PAAD tumors from TCGA and GTEx databases. X and Y axes show relative expression levels of AXL and an individual AKT isoforms, respectively. AXL:AKT1, P = 2.3 × 10−10, R = 0.45; AXL:AKT2, P = 3.4 × 10−9, R = 0.42; AXL:AKT3, P = 3.1 × 10−11, P = 0.47. (C) Phosphorylation status of TBK1 and AKT isoforms upon Gas6 stimulation of AXL in Panc1 cells by immunoblotting (n = 3 over three independent experiments). The cells were pretreated with AXLi and stimulated with Gas6 in the presence or absence of the inhibitor or left unstimulated (Control/UN). AKT1, AKT2, and AKT3 were immunoprecipitated with α-AKT1, α-AKT2, and α-AKT3 Ab, respectively, and blotted with α-pAKT (S473) Ab. Total protein levels were tested with α-AKT1, α-AKT2, and α-AKT3 Ab, respectively. Phosphorylation of AXL was tested by sandwich ELISA using α-AXL and α-pAXL (Tyr866) Ab. Means ± SEM pAKT/AKT3 is shown. (D) Modification of AKT3 by SUMO in Panc1 cells determined by immunoblotting (n = 2 over two independent experiments). Untreated Panc1 cells were immunoprecipitated with indicated α-AKT3 Ab and blotted with α-SUMO1 or α-Sumo2/3 Ab or α-AKT3 Ab. (E) Expression of indicated genes in the empty vector control KPfC CAS9-EV or AKT3 KO cell lines (AKT3 KO A, AKT3 KO B) by immunoblotting (n = 3 over three independent experiments). (F) Accumulation of indicated proteins and phosphorylation of AKT3 was tested in TBK1+/+, TBK1Δ/Δ, and TBK1Δ/Δ KIC PDA cells transduced with myrAKT3 (TBK1Δ/Δ-myrAKT3), n = 3 over three independent experiments. AKT3 was immunoprecipitated and blotted with α-pAKT (S473) and AKT3 Ab. (G) In vitro cell migration and invasion assay of TBK1+/+, TBK1Δ/Δ, and TBK1Δ/Δ-myrAKT3 cell lines (n = 3 over three independent experiments). The cells were stained with phalloidin (red) and Hoechst (blue). Scale bar, 50 μm.
Next, we tested the accumulation and phosphorylation of AKT isoforms downstream of AXL in human PDA cells (Panc1). Because Panc1 cells naturally express Gas6 and Gas6 activity is vitamin K dependent, the cells were pretreated with vitamin K inhibitor warfarin before stimulation (9). We evaluated phosphorylation of AXL [by enzyme-linked immunosorbent assay (ELISA) or immunoblotting] and TBK1 (by immunoblotting) (Fig. 1C and fig. S2A). Gas6 treatment increased accumulation of pAXL and pTBK1. Control samples were incubated with an AXL inhibitor (AXLi, BGB324/bemcentinib) or a TBK1-specific inhibitor (TBK1i, GSK8612) in combination with Gas6, which reduced accumulation of pAXL or pTBK1, respectively, to unstimulated (Control/UN) levels (Fig. 1C and fig. S2A). To test the phosphorylation of each AKT isoform, the samples were immunoprecipitated with AKT isoform–specific antibodies (Abs) and subsequently immunoblotted for pAKT. The results showed a 2.7-fold increase in phosphorylation of AKT3 with a molecular weight of ~90 kDa (Fig. 1C, “IP,” right) and a modest increase in the phosphorylation of the predicted size AKT3 band (~60 kDa; fig. S2A) in Gas6-stimulated cells. Unstimulated, AXLi-, and TBK1i-treated cells had similar basal levels of pAKT3. To determine the potential posttranslational modification of the ~90-kDa AKT3 band and confirm its identity, we tested AKT3 sumoylation status in Panc1 cells. Previous studies have shown the importance of AKT sumoylation for cell proliferation and tumorigenesis (24) and identified a SUMO-consensus site (K272) on AKT3 (25). AKT3 was immunoprecipitated with three AKT3-specific Abs and blotted with Abs specific for SUMO1 or SUMO2/3. Figure 1D shows ~90-kDa sumoylated AKT3 bands in each reaction, suggesting that the 90-kDa pAKT3 band detected in Fig. 1C is likely SUMO-modified AKT3. For pAKT1 and pAKT2, only bands of predicted molecular weight (~60 kDa) were detected (fig. S2A). AKT1 showed a less strong AXL-induced response but was not inhibited by TBK1i, suggesting that AKT1 is being activated by unidentified components of AXL signaling (fig. S2A). Basal levels of pAKT2 were elevated by Gas6 stimulation but were unaffected by AXLi (fig. S2A). We also tested the phosphorylation status of GSK3β, a known AKT substrate, but did not see any changes in its phosphorylation or total levels in response to Gas6 stimulation (fig. S2A).
To confirm the contribution of TBK1 to AXL-induced phosphorylation of AKT3, we used cells from a mouse genetic model (KIC Tbk1Δ/Δ) that harbors a deletion of TBK1 kinase domain, which affects TBK1 activity and reduces its accumulation to undetectable levels (26). KrasLSL-G12D; Cdkn2alox/lox; Ptf1aCre/+ (KIC) mice develop PDA that presents as low-grade ductal lesions at 3 weeks of age and progresses to pancreatic adenocarcinoma by 7 to 8 weeks of age. Stimulation with Gas6 induced pAXL in Tbk1+/+ and Tbk1Δ/Δ cell lines (fig. S2B). TBK1 phosphorylation was detectable only in KIC Tbk1+/+ cells and increased with Gas6 stimulation. AXLi efficiently inhibited phosphorylation of AXL in each cell line and TBK1 phosphorylation in Tbk1+/+ cells. Similar to Panc1 cells, AKT3 phosphorylation was up-regulated in an AXL-dependent manner by Gas6 stimulation (2.2-fold) in Tbk1+/+ but not in Tbk1Δ/Δ cells (fig. S2B). AKT1 phosphorylation was induced by Gas6 in Tbk1+/+ and Tbk1Δ/Δ cells. However, AKT2 phosphorylation levels dropped below unstimulated levels in Gas6-treated Tbk1+/+ cells and were restored in the presence of AXLi (fig. S2B). In Tbk1Δ/Δ cells, pAKT2 levels were nearly undetectable and did not change with Gas6 or Gas6/AXLi treatment (fig. S2B). In addition, no changes in total or phosphorylated GSK3β levels were detected in Gas6-treated cells in the presence or absence of AXLi and TBK1i in Tbk1+/+ cells. In Tbk1Δ/Δ cells, GSK3β phosphorylation was minimal and unchanged by stimulation with Gas6. Together, these results suggest that AKT isoforms respond differently to AXL stimulation and therefore may have distinct contributions to AXL-induced EMT. Only AKT3 showed increased phosphorylation and activation by AXL in a TBK1-dependent manner. AKT1 activity was modestly induced by AXL independent of TBK1, whereas Gas6-induced AKT2 phosphorylation was cell line dependent given that it was elevated in an AXL-independent manner in Panc1 cells but reduced by AXL activation in KIC cells.
AXL-induced AKT3 activation promotes EMT
Considering that AXL-induced stimulation of TBK1 is necessary for AXL-driven EMT in PDA (14) and AXL stimulation leads to TBK1-dependent AKT3 activation, we examined whether AKT3 is required for initiation of EMT. For this, AKT3 CRISPR-Cas9 KO was generated in a primary PDA cell line derived from KrasLSL − G12D/+Trp53fl/flPdx1Cre/+ (KPfC) mice. Next, we tested the expression of EMT markers in empty vector control (Cas9-EV) and AKT3 KO A and AKT3 KO B cells (Fig. 1E). Minor variations in protein expression were detectable between KO A and B, likely because of heterogeneity of the wild-type cells (WT) (Westermann et al., 2022). However, compared with Cas9-EV both KO lines showed reduced expression of mesenchymal markers vimentin and Slug and increased expression of epithelial marker E-cadherin, confirming that AKT3 is necessary for the expression of EMT markers in KPfC cells in vitro (Fig. 1E).
To test whether AKT3 phosphorylation induces EMT markers in a TBK1-dependent manner, we analyzed expression of EMT markers in KIC Tbk1+/+ and Tbk1Δ/Δ cells. Basal levels of pAKT3 were sufficient to induce the expression of vimentin and Slug in Tbk1+/+ cells. In Tbk1Δ/Δ cells, vimentin and Slug expression were reduced, whereas E-cadherin levels increased dramatically compared with KIC Tbk1+/+ cells. To determine the importance of TBK1, we transformed KIC Tbk1Δ/Δ cells with a construct expressing constitutively active myristoylated AKT3 (27), which was sufficient to reverse the expression of EMT markers to the levels detected in Tbk1+/+ cells (Fig. 1F). In parallel, we tested these cell lines in a three-dimensional (3D) invasion assay. Unlike Tbk1+/+ cells, Tbk1Δ/Δ cells formed tight spheroids and did not show any invasiveness (Fig. 1G). In correlation with the EMT expression data, myrAKT3 expression in Tbk1Δ/Δ cells reversed the phenotype to a more mesenchymal-like one displayed by KIC Tbk1+/+ cells (Fig. 1G). These results demonstrate that TBK1-dependent AKT3 activation contributes to AXL-mediated EMT.
TBK1 interacts with and phosphorylates AKT3
TBK1 can directly activate AKT (15, 16). We hypothesized that TBK1 directly binds to and activates AKT3 to drive EMT downstream of AXL. First, we examined localization of TBK1 and AKT3 in untreated Panc1 cells and determined that there is a considerable overlap in TBK1- and AKT3-specific signals suggesting that the proteins colocalize mostly in the cytoplasm but also in the nucleus (Fig. 2A). Similarly, in KIC Tbk1+/+ cells, TBK1 and AKT3 colocalized in the cytoplasm and the nuclei of unstimulated and Gas6-stimulated cells (fig. S2, C and D). Next, we tested TBK1 and AKT3 interactions in a series of pull-down assays. Using human embryonic kidney (HEK) 293T and Panc1 cells transfected with epitope-tagged TBK1 or AKT3, we demonstrated that exogenous epitope-tagged TBK1 and AKT3 coimmunoprecipitate with endogenous AKT3 and TBK1, respectively (Fig. 2, B and C). Second, we immunoprecipitated endogenous TBK1:AKT3 complexes from KIC, KPfC, and Panc1 cells using AKT3- and TBK1-specific Abs (Fig. 2D). These results demonstrate that TBK1 interacts with AKT3.
Fig. 2. TBK1 and AKT3 interaction and AKT3 activation.
(A) Subcellular localization of TBK1 and AKT3 in Panc1 cells by confocal microscopy. Representative images shown (n = 3 over three independent experiments). Scale bar, 20 μm. (B and C) Endogenous AKT3 or TBK1 pull-down with epitope-tagged TBK1 or AKT3, respectively (n = 2 over two independent experiments). HEK293 cells (B) were transfected with constructs expressing either MFlag-TBK1, MFlag-AKT3, or MHA-AKT3 or left untransfected (UN). Panc1 cells were transfected with a construct expressing HA-AKT3 or left untransfected (UN). Immunoprecipitation was performed with α-Flag or α-HA Ab. The lysates and immunoprecipitants were blotted with α-AKT3 or α-TBK1 Ab. (D) Pull-down of endogenous AKT3:TBK1 complexes with α-AKT3 or α-TBK1 Ab from unstimulated (UN) or Gas6-stimulated cell lysates (n = 2 over two independent experiments). The lysates and immunoprecipitants were probed with α-TBK1 and α-AKT3 Ab. Ag, agarose-only control. (E) In vitro kinase activity assay was done using recombinant TBK1 and AKT3 proteins and cold ATP. AKT3 phosphorylation sites were identified by MS (n = 1 over two experiments).
To investigate whether TBK1 can directly bind to and activate AKT3, we performed an in vitro kinase activity assay with human recombinant TBK1 and AKT3 using cold adenosine triphosphate (ATP). Kinase reactions were separated via SDS–polyacrylamide gel electrophoresis (SDS-PAGE) and analyzed by mass spectrometry (MS), which showed phosphorylation of AKT3 at T144, T193, T298, T302/309, and S472 (Fig. 2E and fig. S2D). The latter two sites correspond to most commonly detected phosphorylation sites on AKT isoforms, AKT1 T308/AKT2 T309 and AKT1 S473/AKT2 S474 (28–31), as shown by the amino acid alignment (fig. S2D). Phosphorylation at both sites is necessary for maximal activation of AKT [reviewed in (32)]. These results confirm that AKT3 is a direct substrate of TBK1. Given that these proteins colocalize and coimmunoprecipitate and AKT3 phosphorylation upon AXL-stimulation is TBK1 dependent, these data strongly suggest that AKT3 is phosphorylated by TBK1.
mTORC1 is important for AXL-induced AKT3 phosphorylation
AKT was previously shown to be activated by mammalian target of rapamycin complex 2 (mTORC2) in a positive-feedback loop (33, 34). To determine mTOR’s contribution to AXL/TBK1-induced AKT3 phosphorylation, KIC TBK1+/+ cells were pretreated with warfarin and inhibitors of mTORC1 (mTORC1i, rapamycin) or mTORC2 (mTORC2i, JR-AB2–011) and stimulated with Gas6. mTor inhibition did not affect AXL, TBK1, or AKT3 accumulation or AXL phosphorylation induced by Gas6 (fig. S2E). However, mTORC1i, but not mTORC2i, reduced total pTBK1 and pAKT3 (71 and 69%, respectively) compared with “No Drug” control in Gas6-treated cells (fig. S2E) suggesting that mTORC1 is involved in AXL/TBK1-induced AKT3 activation. Similar to mTORC2, mTORC1 is an AKT substrate, and it might activate AKT3 isoform in a positive-feedback loop. In addition, mTORC1 was shown to be a TBK1 target (35). Further, TBK1 coimmunoprecipitated with mTOR in No Drug and mTORC2i-treated cells. However, mTORC1i inhibited mTOR:TBK1 interactions (fig. S2E). Therefore, it is possible that mTORC1 is stimulated by AXL/TBK1 signaling and then phosphorylates AKT3.
AXL-TBK1 is required for accumulation of pAKT3 and Snail in the nucleus
To quantify accumulation levels of total and pAKT3 in the nucleus and the cytoplasm, Panc1 cells were pretreated with warfarin and stimulated with Gas6 in the presence or absence of AXLi or TBK1i. Lysates were fractionated and analyzed by immunoblotting. Tubulin and lamin A/C were used as controls for cytoplasmic and nuclear fractionation, respectively (Fig. 3A). TBK1 and AKT3 were found in the nuclear and cytoplasmic fractions. AKT3 was immunoprecipitated from the fractionated lysates and blotted for pAKT. Gas6-stimulation increased phosphorylation of AKT3 in the cytoplasm and the nucleus (Fig. 3A). However, pAKT:total AKT3 ratio was twofold higher in the nucleus compared with the cytoplasm. AXLi and TBK1i reduced accumulation of total AKT3 in cytoplasmic fraction and pAKT3 in both fractions. This demonstrates that (i) AXL-induced activation of AKT3 in the nucleus is TBK1 dependent and (ii) total nuclear AKT3 levels do not change upon AXL stimulation or treatment with AXLi and TBK1i. This suggests that, in Panc1 cells, AXL/TBK1 signaling does not induce additional AKT3 translocation to the nucleus. However, AKT3 has a nuclear export signal (NES) (36) and may shuttle out of the nucleus. Therefore, future experiments characterizing subcellular localization of AKT3 ΔNES mutant in Gas6-stimulated cells are necessary.
Fig. 3. Subcellular localization of activated AKT3 after Gas6 stimulation and AKT3 in cell invasion.
(A) Protein subcellular localization and accumulation in Panc1 cells before and after AXL stimulation (n = 3 over three independent experiments). Panc1 cells pretreated with indicated inhibitors and stimulated with Gas6 in the presence or absence of the inhibitors or left untreated (UN), lysed, and fractionated. Whole cell lysate (WCL), nuclear (Nuc), or cytoplasmic (Cyt) fractions were probed with indicated Abs. To determine Akt3 phosphorylation status, AKT3 was immunoprecipitated with α-AKT3 Abs and blotted with α-pAKT (S473) Ab. Total protein level of AKT3 are shown. AXL was immunoprecipitated with α-AXL Ab and blotted with α-pAXL (Tyr702) or α-AXL Ab. Means ± SEM nuclear Snail and mean ± SEM pAKT/AKT3 is shown. (B) In vitro cell migration and invasion assay for KPfC AKT3 KO A and AKT3 KO B and their derivative cell lines, WT AKT3 rescue AKT3 KO: AKT3 and mutant AKT3-NLS1 rescue AKT3 KO: AKT3-NLS1 cells. Representative images shown (n = 3 over three independent experiments). All cells including empty vector control CAS9-EV were stained for phalloidin (red) and Hoechst (blue). Scale bar, 50 μm. **P < 0.01; ***P < 0.005. Adjusted P values were calculated using Dunn’s multiple comparison test.
Because we have previously shown that AXL-driven TBK1 activation increases expression of EMT inducing TFs (EMT-TFs) Slug and Snail (14), we evaluated their expression in fractionated lysates. We also found that these EMT-TFs displayed different localization patterns. Slug was detected only in the cytoplasm, and its expression did not seem to respond to any treatment. Snail mainly accumulated in the nucleus, and its nuclear levels were increased 2.4-fold by Gas6 stimulation (Fig. 3A). Consistent with our prior data, AXLi and TBK1i reduced Snail levels but only in the nucleus.
To clarify the contribution of TBK1 to AKT3 nuclear localization, we compared AKT3 protein levels in fractionated KIC Tbk1+/+ and Tbk1Δ/Δ cells: Gas6 increased nuclear AKT3 in each cell line, but, unfortunately, nuclear AKT3 levels were too low to detect phosphorylation by immunoblotting. These results suggested that levels of nuclear AKT3 may be a cell line–specific feature, and in KIC cells, this process is TBK1 independent (fig. S3, A and B). In contrast, in MDA-MB-231 breast cancer cells and MCF10a normal breast epithelial cells, nuclear localization of AKT3 is readily detectable in all cells by immunofluorescence and is inhibited by AXL knockdown (KD) with AXL short hairpin RNA (shRNA) (fig. S4, A and B), indicating that in these cells, AKT3 is transported to the nucleus during AXL signaling.
Next, we tested Snail accumulation in fractionated KIC Tbk1+/+ and Tbk1Δ/Δ cell lysates. Similarly to Panc1 cells, AXL stimulation increased nuclear Snail ~1.3-fold in Tbk1+/+ cells (fig. S3A). In Gas6-treated Tbk1Δ/Δ cells, however, nuclear Snail accumulation did not change, whereas cytoplasmic Snail dropped ~12-fold compared with the unstimulated control (fig. S3B). These results support that TBK1 participates in Snail accumulation and nuclear localization during AXL signaling.
Nuclear localization of AKT3 is essential for cell invasion
AKT3 is found in the nucleus (37), but the mechanisms of targeting AKT3 to the nucleus are unclear. Proteins more than 40 kDa must be actively transported through the nuclear membrane by importins that recognize and bind a nuclear localization signal (NLS) (38). We used a web-based NLS mapper (39) and identified a weak bipartite NLS in the AKT3 amino acid sequence (accession number: Q9Y243) located in a flexible linker region between the PH domain and kinase domain (fig. S4C). On the basis of these in silico findings, we created two AKT3 mutant expression constructs: AKT3-NLS1 and AKT3-NLS2. AKT3-NLS1 carries two point mutations (K141R and R142A) that alter the leucine-rich NLS region to the sequence that resembles the linker area in AKT2 (fig. S4C). For AKT3-NLS2, a 10–amino acid sequence flanking the NLS was replaced to mimic a longer part of the linker region as coded in AKT2. WT AKT3 and the mutants were stably expressed in human breast HMLER cells and analyzed by fluorescence microscopy. Parental HMLER cells do not express endogenous AKT3 (40); therefore, no AKT3 signal was detectable in control HMLER cells stably expressing green fluorescent protein (HMLER:GFP; fig. S4D). WT AKT3 was predominantly nuclear (87%); however, AKT3-NLS1 and AKT3-NLS2 mutants were largely restricted to the cytoplasm with 18 and 29% nuclear localization, respectively (fig. S4E).
Next, to test whether AKT3 nuclear localization is important for EMT, we transduced pancreatic KPfC AKT3 KO cell clones A and B (KO A and KO B) with WT AKT3 or AKT3-NLS1 expression constructs and evaluated 3D invasion. Analysis of tumor cell morphology and invasion revealed that whereas AKT3 KO resulted in a reduced invasive phenotype compared with control, rescuing AKT3 expression with WT AKT3 restored the invasive phenotype (Fig. 3, B and C). However, AKT3-NLS expression in AKT3 KO cells did not restore invasiveness, suggesting that nuclear AKT3 localization is necessary to drive EMT.
AKT3 interacts with Snail in the nucleus
To validate cell fractionation data, we analyzed Snail expression and localization by confocal microscopy. In unstimulated Panc1 cells (UN) treated with warfarin to “silence” AXL stimulation, Snail expression was barely detectable. However, Gas6-stimulation upregulated nuclear levels of Snail, whereas its cytoplasmic levels remained the same (Fig. 4A). Similar to cell fractionation data, AXLi treatment reduced nuclear Snail levels to background levels.
Fig. 4. Subcellular localization of Snail and half-life after Gas6 stimulation.
(A) Subcellular localization of Snail in Gas6-stimulated Panc1 cells. Representative images shown (n = 3 over three independent experiments). Panc1 cells were pretreated with AXLi and stimulated with Gas6 in the presence or absence of the inhibitor or left unstimulated (UN). Scale bar, 25 μm. (B) Localization of Snail and AKT3 in Panc1 cells. Representative images shown (n = 3 over three independent experiments). Scale bar, 20 μm. (C) Pull-down of endogenous TBK1:AKT3:Snail complexes with α-AKT3 Ab from untreated Panc1 cell lysates (n = 2 over two independent experiments). WCL or fractionated cell lysates and the immunoprecipitants were probed with α-Snail, α-TBK1, and α-AKT3 Ab. Ag, agarose only. Asterisks (*) show protein bands of the predicted molecular weights. (D) AKT3 accumulation in parental and shAKT3 transduced Panc1 cells (n = 3 over three independent experiments). (E) Parental or AKT3 shRNA (shAKT3) transduced Panc1 cells were treated with CHX in the presence or absence of Gas6 and harvested at indicated time points (n = 3 over three independent experiments). The lysates were probed with α-Snail and α–β-actin or α–glyceraldehyde-3-phosphate dehydrogenase Ab. Means ± SEM. Snail expression was normalized against β-actin and shown as % CHX 0-hour chase.
Knowing that pAKT3 and Snail accumulate in the nucleus upon AXL activation, we assessed nuclear localization of AKT3 and Snail in Panc1 cells by confocal microscopy. Imaging (Fig. 4B) indicates that AKT3 and Snail overlap in the nucleus, suggesting that the proteins interact. Interaction of AKT3 and Snail was confirmed by co-immunoprecipitation. Snail was detected in the AKT3 IP from whole-cell lysate and in the nuclear fraction of untreated Panc1 cells. Snail was detected at the predicted molecular weight and at a higher molecular weight likely indicating that Snail is posttranslationally modified (Fig. 4C). Snail was detectable in the cytoplasmic fraction of the lysate, but it was not pulled down with AKT3. This suggests that AKT3:Snail interactions occur in the nucleus but not in the cytoplasm. Because AKT3 colocalizes and interacts with TBK1, we probed AKT3 pull downs for TBK1. TBK1 coprecipitated with AKT3 from both fractions; however, most of the TBK1 signal was detected in the nuclear fraction (Fig. 4C). Similar to Snail, we detected bands corresponding to the predicted TBK1 molecular weight and slower migrating TBK1-specific bands indicative of its potential posttranslational modifications. This suggests that AKT3 interacts with TBK1 and Snail and that all three proteins might function as part of the multiprotein signaling complex located in the nucleus.
AXL activation increases Snail half-life in an AKT3-dependent manner
To investigate the mechanism regulating Snail accumulation in AXL-stimulated cells, we knocked down AKT3 expression with shRNA (Fig. 4D) and tested Snail protein stability by cycloheximide (CHX) chase assay (Fig. 4E). Panc1 cells were treated with CHX in the presence or absence of Gas6 over a time course of 8 hours. Consistent with previous findings (41), Snail had a half-life of ~2 hours and was difficult to detect by 8 hours of chase when treated with CHX (Fig. 4E). However, AXL stimulation with Gas6 extended Snail half-life. Further, we found that Snail expression was nearly undetectable in shAKT3 Panc1 cells, and Gas6 treatment no longer had a stabilizing effect on Snail (Fig. 4E). These results demonstrate that AXL-induced accumulation of Snail in the nucleus is likely due to AKT3-dependent stabilization of the protein.
Selective targeting of AKT3 with an allosteric small-molecule inhibitor
Several drugs targeting pan-AKT activity (e.g., GDC0068, AXD5363, and MK-2206) are currently in various stages of clinical testing. However, many of these trials report toxicity, such as hyperglycemia and hyperinsulinemia, because of the essential functions of AKT1 and AKT2 in tissue homeostasis (42–45). An AKT3-selective inhibitor has the potential to overcome these issues. The similarity between AKT1, AKT2, and AKT3 in the kinase domain precludes selective kinase inhibition. However, an allosteric site located in a cleft between the PH domain and the kinase domain has been used to identify AKT1-, AKT2-, and AKT1/2-selective inhibitors (46–48). Sequence alignment around this allosteric site suggested that there are exploitable differences in this region (Fig. 5A). A structural model produced by comparison of the crystal structures of AKT1 (48) and the AKT2 kinase domain suggested that a single amino acid deletion in AKT2 and AKT3 compared with AKT1 leads to a change in the path that the protein backbone follows, opening a pocket at the front of the allosteric binding site (Fig. 5B). This pocket is small in the case of AKT2 because of the protrusion of the large side chain of Arg269, but the pocket is larger in AKT3 because of the presence of a glycine at this site. A series of allosteric small-molecule inhibitors of AKT3 were developed (BerGenBio ASA, patent number WO/2016/102672) with backbones that bind to the allosteric site on AKT3 specifically. One of these compounds, a potent and selective allosteric AKT3 inhibitor, BGB214 (N- (5- (4- (1-aminocyclobutyl)phenyl)- 4-pheny lpyridin-2-yl)-2-((1r,4r)-4-(N-methylacetamido) cyclohexyl) acetamide), referred to as AKT3i, was chosen for our studies (Fig. 5C). In biochemical assays using purified tag-free enzymes, AKT3i had a median inhibitory concentration (IC50) of 13 nM for AKT3 with approximately 1000-fold selectivity against AKT1 and >35-fold selectivity against AKT2 (Fig. 5D).
Fig. 5. Effect of AKT3 inhibitor BGB214 (AKT3i) on Snail accumulation.
(A) Differences in sequence between AKT1, AKT2, and AKT3 around the allosteric site include a deletion in AKT2 and AKT3 compared with AKT1. (B) Surface view of the front of the allosteric binding site of AKT3, including bound allosteric inhibitor AKT VIII (green). Homology model of AKT3 based on crystal structures of AKT1 bound to AKT VIII (PDB 3o96) and AKT2 kinase domain (PDB 1o6k). Side chains from the AKT1 crystal structure (Lys268, yellow) and the AKT2 crystal structure (Arg269, magenta) are superimposed. A molecule with similar structure to BGB214 (pink) docked at the allosteric site clashes with Lys268 of AKT1 (yellow). (C) Structure of BGB214. (D) Inhibition of AKT1, AKT2, and AKT3 enzymatic activity on GSK3α-derived Ultra U lightTM-labeled crosstide substrate (n = 3 over three independent experiments). (E) The effect of BGB214 (AKT3i) on Panc1 viability in vitro (n = 4 over four independent experiments). Eight different drug concentrations were tested with eight replicates per concentration. Relative cell number was determined by MTS assay. (F) Protein accumulation in Panc1 cells stimulated with Gas6 in the presence or absence of AXLi, TBK1i, and AKT3i were tested by immunoblotting (n = 3 over three independent experiments). Panc1 cells were pretreated with indicated inhibitors and stimulated with Gas6 in the presence or absence of the inhibitors or left unstimulated (UN). AKT3 was immunoprecipitated with α-AKT3 Ab and blotted with α-pAKT (S473) or α-AKT3 Ab. AXL was immunoprecipitated with α-AXL Ab and blotted with pAXL (Tyr702) or AXL Ab.
First, AKT3i efficacy was evaluated in cell viability assays. The calculated AKT3i IC50 for Panc1 cells (Fig. 5E) and MDA-MB-231 (fig. S5A) cells were 13.5 and 9.3 μM, respectively. AKT3i specificity was validated using HMLER cells transduced with vectors expressing GFP (control) or AKT3. Treatment of HMLER cells (GFP and AKT3) with increasing concentrations of AKT3i did not affect the accumulation or phosphorylation of AKT1 or AKT2 (fig. S5B). Because HMLER-GFP cells do not express endogenous AKT3, AKT3 signal was undetectable. In HMLER-AKT3 cells, AKT3i inhibited AKT3 phosphorylation at 0.3 μM concentration but did not affect total levels of AKT3 (fig. S5B), highlighting its specificity. AKT3i efficacy was validated in a 3D cell migration assay. MDA-MB-231 cells were plated in collagen/Matrigel and treated with Gas6 in the presence or absence of 3 μM AXLi or 2 μM AKT3i for 48 hours. Invasion more than 50 μm was determined and quantified, revealing that inhibition of AXL or AKT3 substantially reduced cell migration/invasion (fig. S5, C and D). However, cell viability was not noticeably affected by the drugs used at indicated concentrations (fig. S5E).
Last, we used AKT3i to confirm AKT3’s contribution to Snail upregulation in Gas6-stimulated Panc1 cells. AKT3 expression and phosphorylation and Snail expression levels were up-regulated in Gas6-treated cells compared with untreated (UN) cell levels (Fig. 5F). AXLi and TBK1i, used for control, reduced levels of pAKT and Snail. Strikingly, AKT3i treatment reduced AXL activity and potently reduced AKT3, pAKT3 and Snail levels. AKT3i also decreased phosphorylation of GSK3β. On this basis, we conclude that selective inhibition of AKT3 with the allosteric inhibitor (AKT3i) blocks AKT3-mediated up-regulation of Snail and inhibits migration/invasion properties of the cells.
AKT3 expression is associated with poorly differentiated tumors and increased metastasis
To assess the biological consequence of AKT3 activity, control KPfC cells (CAS9-EV), AKT3 KO KPfC cells (AKT3 KO), or AKT3 KO KPfC cells transduced with AKT3 (rescue) were injected orthotopically into the pancreases of C57BL/6J mice (Fig. 6, A to H). Primary tumor and metastatic burden was evaluated 19 days after injection. Although primary tumor weight did not differ between the three groups (Fig. 6B), gross metastatic burden was reduced in tumors lacking AKT3, an effect rescued by AKT3 reexpression (Fig. 6A). Hematoxylin and eosin (H&E) analysis as well as IHC for CK19, a PDA tumor cell marker (49), confirmed reduced metastasis to livers of AKT3 KO tumor–bearing mice (Fig. 6, C and D). Consistent with this observation, tumors lacking AKT3 were more differentiated and expressed higher levels of E-cadherin and lower levels of vimentin (Fig. 6, E to H). The expression of E-cadherin and vimentin in vivo was consistent with the expression of these proteins in vitro (Fig. 6F). In addition, we observed that the expression level of AKT3 correlated with the number of gross metastases (Fig. 6, A and F).
Fig. 6. The effect of AKT3 expression on tumor differentiation and metastasis in mice.
C57BL/6J mice were injected orthotopically with 250,000 KPfC PDA cells (CAS9-EV, AKT3 KO, or rescue). (A) Gross metastases (CAS9-EV, 3 of 9; AKT3 KO, 0 of 10; rescue, 6 of 9) and (B) primary tumor weights (n = 9 or 10 per group). (C and D) Representative images of H&E staining and CK19 IHC in livers. CK19 reactivity was quantified as a percent of total liver area [(C) and (D) n = 6 per group]. (E) Representative images of tumors stained using H&E and IHC for E-cadherin and vimentin (n = 6 per group). (F) Accumulations of AKT3, ECad, and vimentin were tested in KPfC CAS9-EV, AKT3 KO, and rescue cells by immunoblotting (n = 3 over three independent experiments). Cells were lysed and probed with indicated Ab. Percent area of Ecad (G) and vimentin (H) was quantified (n = 4 to 6 per group as indicated in bar graph). Adjusted P values were calculated using Dunn’s multiple comparison tests (*P < 0.05; **P < 0.01). ns, not significant.
Nuclear AKT3 is associated with aggressive cancer and worse survival in patients
To investigate the importance of nuclear AKT3 in cancer development, we assessed AKT3 localization in pancreatic and breast tumors from patients by IHC. In AXL-positive tumors, nuclear AKT3 was detected in single tumor cells outside epithelial ducts (Fig. 7A and quantification in Fig. 7B and fig. S6A). However, in AXL-negative tumors, AKT3 was cytoplasmic, supporting our findings that (i) AXL activity induces nuclear accumulation of AKT3 and (ii) nuclear AKT3 results in a less differentiated (more mesenchymal-like) tumor cell phenotype.
Fig. 7. Expression and localization of AKT3 and Snail in human PDA.
(A) Representative images of IHC for AKT3 and AXL in human PDA (n = 71). (B) Shows number of cases with nuclear and cytoplasmic AKT3 and AXL-positive and -negative human PDA. P value was calculated using Fisher’s exact test. (C) Representative images of IHC staining for AKT3 (red) and Snail (green) in human AXL-positive/nuclear AKT3 PDA (n = 8). Scale bar, 100 μm.
Next, to assess the influence of AKT3 signaling and its downstream targets on patient survival, genes that were found to be differentially expressed after AKT3 overexpression in MCF10A cells were used to generate an AKT3 score (table S1), which was then mapped against probes in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) database. The results showed that high AKT3 expression is associated with an EMT signature and worse survival outcome (fig. S6B). Further, we found a prominently different distribution between breast cancer subtypes (based on PAM50 intrinsic subtypes) and AKT3 scores (fig. S6C). In estrogen receptor (ER)–negative tumors and basal-like tumors, high levels of AKT3-associated gene expression correlates with more aggressive forms of breast cancer, worse overall outcome, and a higher hazard ratio. This is consistent with previous reports showing high AKT3 copy number alterations in patients with triple negative breast cancer (TNBC) (50, 51) and a correlation between AKT3 expression levels and higher grade breast tumors (52). To assess the effect of nuclear AKT3 localization on patient survival, we correlated IHC data for AKT3 in clinical breast cancer samples (fig. S6A) with the clinical outcomes. The results revealed that nuclear AKT3 predicted a worse overall outcome in this cohort of patients with breast cancer (fig. S6D).
Last, to validate our in vitro data showing nuclear AKT3:Snail interactions, we examined localization of these proteins in human PDA tissues using IHC. Nuclear AKT3 and Snail expression strongly correlated and completely overlapped in all tested samples (Fig. 7C).
Together, these results confirm that AXL-mediated signaling induces nuclear accumulation of pAKT3 and Snail, which promotes EMT and metastasis (Fig. 8). This suggests that (i) AKT3 may serve as a therapeutic target that avoids toxicity associated with pan-AKT inhibition and (ii) nuclear AKT3 can be used as a prognostic biomarker for predicting worse overall survival in aggressive cancers, including metastatic pancreas and breast tumors. Further investigation is needed to determine whether nuclear AKT3 is an important feature of additional types of aggressive cancer.
Fig. 8. Working model: AXL signaling induces TBK1-dependent phosphorylation of AKT3, accumulation of Snail in the nucleus, and promotes EMT.
AXL activation by its ligand Gas6 leads to phosphorylation and activation of TBK1 and subsequent phosphorylation of AKT3 either directly by TBK1 or via TBK1-mTORC1. Nuclear TBK1 phosphorylates AKT3 located in the nucleus. In addition, AKT3, phosphorylated by cytoplasmic TBK1, may travel to the nucleus. Accumulation of pAKT3 in the nucleus results in accumulation/stabilization of Snail, an EMT-TF in the nucleus, which leads to EMT.
DISCUSSION
Phosphatidylinositol 3-kinase/AKT is a downstream signaling pathway stimulated by Gas6-AXL [reviewed in (53, 54)]. The inhibition of AXL with AXLi (BGB324) reduces AKT phosphorylation in PDA cells in vivo and in vitro (12). The AKT family includes three isoforms: AKT1, AKT2, and AKT3. Despite high homology between the isoforms, each contributes distinctly to cell biology and in cancer (32, 55, 56). However, communication of AXL with downstream signaling cascades can be cell type and oncogenotype specific. Therefore, in the current study, we examined the response of individual AKT isoforms to Gas6-AXL stimulation. Here, we demonstrated that AXL-stimulation exerts a differential effect on phosphorylation of AKT isoforms. Axl activation induces phosphorylation and, therefore, activation of AKT3.
TBK1 directly phosphorylates AKT to support cell survival and oncogenic transformation (15). In addition, AXL-induced stimulation of TBK1 is necessary for AXL-driven EMT in PDA (14). In the present study, we investigated the mechanisms of TBK1-dependent AKT3 activation and the importance of AKT3 in EMT. Many studies have focused on the function of AKT1, AKT2, or panAKT in cancer. However, the contribution of AKT3 has not been studied as frequently. This may be explained by the fact that AKT3 is the least expressed of the three isoforms (57), and prior results on the function of AKT3 in tumorigenesis are inconsistent (58–63). However, a few reports suggest that AKT3 contributes to cancer progression (64–67). We believe that the inconsistency of AKT3 studies may be due to different genetic contexts and the fact that AKT3 can be expressed as two alternatively spliced variants, one of which lacks S472 (68). Overexpression of the AKT3 variant lacking S472 is associated with improved overall survival and reduced lung metastasis in preclinical models of breast cancer, whereas expression of WT AKT3 results in increased tumorigenesis (68). In our studies, we showed that AKT3 stimulates expression of EMT markers vimentin and Snail in a TBK1-dependent manner, whereas AKT3 deletion by CRISPR-Cas9 reverses the phenotype and up-regulates expression of E-cadherin. Using confocal microscopy, pull-down assays, and an in vitro kinase assay, we demonstrated that TBK1 colocalizes with AKT3 in the cytoplasm and the nucleus and directly interacts with and phosphorylates AKT3 at several sites, including S472 (Fig. 8). We identified a putative NLS sequence in AKT3 and showed that nuclear localization of AKT3 is critical for its role in cell invasion. TBK1, however, seemed dispensable for nuclear localization of AKT3, but it was necessary for its phosphorylation in the nucleus and the cytoplasm. It is unclear whether TBK1 binds and phosphorylates AKT3 in the cytoplasm and then both proteins are delivered into the nucleus or whether they independently translocate to the nucleus where TBK1 phosphorylates AKT3 (Fig. 8).
AKT3 is phosphorylated by its substrate mTORC2 in a positive-feedback loop (33, 34). mTORC2 and mTORC1 are direct targets of TBK1 (35, 69). Therefore, we interrogated whether mTORC1 or mTORC2 phosphorylates AKT3. We found that mTORC1 contributes to AXL-induced AKT3 phosphorylation. Considering that mTORC1 is a TBK1 and AKT substrate, it is possible that TBK1 and/or AKT3 directly activate mTORC1, and, similarly to mTORC2, it contributes to AKT3 phosphorylation in a positive-feedback loop (Fig. 8).
All AKT isoforms harbor sumoylation signals (25), and AKT sumoylation is important for cell proliferation and tumorigenesis (24). Here, we confirmed AKT3 sumoylation in Panc1 cells and showed phosphorylation of sumoylated AKT3 in response to AXL stimulation. Because sumoylation is implicated in promoting protein phosphorylation (70) and protein-protein interactions and is critical for functions of many proteins with nuclear localization (71), it will be important to examine the importance of sumoylation of AKT3 in the context of AXL signaling in future experiments.
AKT3 is implicated in various aspects of EMT, including tumor progression, DNA damage repair response, and drug resistance (62, 72–74). AKT3 directly interacts with and phosphorylates TBX3, a T-box TF. AKT3 phosphorylation of TBX3 promotes its stability and nuclear accumulation, resulting in increased EMT and cell migration (75). AKT3 may also promote cancer stemness in a Snail-dependent manner (76). In our studies, we determined that AKT3 colocalizes and interacts with Snail. AKT3:Snail complexes were coimmunoprecipitated from the nuclear but not cytoplasmic fractions of lysates. The complex also contained TBK1 protein, suggesting that all three proteins might function as a part of a multimeric protein complex. Snail is an unstable protein that was shown to degrade in the cytoplasm much faster than in the nucleus (77). Our studies revealed that AXL stimulation increased Snail half-life in AKT3-dependent manner and led to increased accumulation of Snail in the nucleus but not in the cytoplasm (Fig. 8). Nuclear accumulation of Snail is controlled by its posttranslational modifications (77). In particular, phosphorylation of Snail at T203 site by two kinases, Lats2 and STK39, is important for its nuclear retention and accumulation, as well as its functions in EMT and cell invasion (78, 79). It is plausible that AKT3 up-regulates Snail half-life by phosphorylating it at T203 site and retaining it in the nucleus. The results of synthetic peptide library profiling for substrate specificity of Ser/Thr kinases show that AKT3 can bind to Snail at T203 (80). Therefore, future experiments are needed to investigate whether these interactions happen in Gas6-stimulated Panc1 or KIC cells. It is also possible that the direct binding of AKT3 to Snail or recruitment of other proteins such as deubiquitinating enzymes (DUBs) by AKT3 protects it from proteasomal degradation. For example, the DUB USP10 promotes the stability of Snail in breast, ovarian, and lung cancer cell lines (81). Other potential candidate proteins that might be involved in the degradation of Snail are the F-box ligases, of which FBX15 and FBXO11 ubiquitinate and support the degradation of Snail (82). Further studies are needed to evaluate changes in Snail posttranslational modification and determine the mechanisms of its nuclear accumulation induced by AXL-TBK1-AKT3 signaling. Because our previous studies have shown an increase in other EMT-TFs in response to AXL-TBK1 signaling (14), we examined expression of Slug, a Snail homolog, in AXL-stimulated Panc1 cells. However, we did not detect any changes in its accumulation levels or localization. Considering that this may be a cell line–specific response, further studies are needed to examine expression of Slug and other EMT-TFs upon the AXL-TBK1-AKT3 stimulation in different cell lines.
Our in vivo studies confirmed that AKT3 expression is associated with poorly differentiated tumors and increased metastasis and suggest its potential value as a therapeutic target, the inhibition of which might enhance sensitivity to standard therapy. This is supported by other work that indicates that mutant Kras can selectively up-regulate AKT3 in lung and PDA cell lines (83). Our results were validated using pancreatic and breast cancer specimens from patients. We demonstrated that nuclear accumulation of AKT3 correlates with increased AXL expression and a less differentiated tumor cell phenotype. These results suggest that nuclear AKT3 can be used as a prognostic biomarker for predicting worse overall survival.
These studies do not rule out that AKT3 affects other cell types, such as macrophages; this is relevant given the function of TBK1 in innate immune signaling and the STING pathway. It has been reported that 7-DHC, a cholesterol precursor, stimulates type I interferon production via AKT3 activation, where AKT3 directly binds and phosphorylates interferon regulatory factor 3 on S385 (84). In addition, AKT3 (pS472) in macrophages has been shown to promote migration, proliferation, wound healing, and collagen organization (85). Furthermore, AKT3 phosphorylates RNA processing proteins that promote the alternative splicing of fibroblast growth factor receptors, highlighting the importance of nuclear AKT3 in EMT maintenance (21).
In summary, our data supported that nuclear AKT3 has utility as a potential biomarker for aggressive cancers that express AXL and that AKT3 is a specific mediator of EMT signaling downstream of AXL. In addition, because there are ongoing clinical trials targeting AXL in multiple cancer types, analyses of these tumors for AXL expression and AKT3 localization after treatment may provide clinicians with a much-needed readout for treatment efficacy. Last, we propose that selective inhibition of AKT3 may represent a therapeutic avenue for treating aggressive and recurrent cancer that avoids toxicity associated with pan-AKT inhibition.
MATERIALS AND METHODS
Drugs and Abs
The drugs used in this study are as follows: warfarin (S4545, Selleckchem), BGB324/bemcentinib (AXLi, BerGenBio), GSK8612 (TBK1i, Selleck Chemicals), BGB214 (AKT3i, BerGenBio), rapamycin (mTORC1i, MedChemExpress), JR-AB2–011 (mTORC2i, MedChemExpress), and CHX (01810, Sigma-Aldrich). All Abs and reagents used for protein staining are listed in table S2.
Plasmid constructs
pRP(Exp)-CMV-huGas6 was generated by VectorBuilder by inserting a human Gas6 coding sequence into a pRP(Exp)–cytomegalovirus (CMV) expression vector under a CMV promoter. pMFlag-TBK1 (pWZL Neo Myr Flag TBK1, Addgene plasmid # 20648; http://n2t.net/addgene:20648; RRID: Addgene_20648) and pMFlag-AKT3 (pWZL Neo Myr Flag AKT3, Addgene plasmid # 20423; http://n2t.net/addgene:20423; RRID: Addgene_20423) were gifts from W. Hahn and J. Zhao (86). pMHA-AKT3 was a gift from W. Sellers (1236 pcDNA3 Myr HA Akt3, Addgene plasmid # 9017; http://n2t.net/addgene:9017; RRID: Addgene_9017). pHA-AKT3 was created using a pMHA-AKT3 construct by removing the N-terminal myristoylation signal (Gly-2) with overlap extension polymerase chain reaction (PCR) (87) and primers set 1 (5′-CCTACTTGGCAGTACATCTACGTATTAGTCATC-3′; 5′-GGCTTGCTCTTGCTGCTCGCCATGGTGGTG-3′) and set 2 (5′-CACCACCATGGCGAGCAGCAAGAGCAAGCC-3′; 5′-CAGTGCAATTTCAGGTGCAATTGGGAATTC-3′). CRU5–internal ribosomal entry site (IRES)–GFP retroviral vectors for expression of hSLUG, AKT3, shLuc, and shAXL were prepared as described (11). CRU5-IRES-GFP retroviral vectors expressing AKT3-NLS and AKT3-NLS were generated by site-directed mutagenesis (Quick change, #2200519). Constructs expressing AKT3 targeting single-guide RNAs (sgRNAs) for CRISPR-Cas9 KO of AKT3 were generated using oligo sets “A” (5′-CACCGAATGGTAACATCGCTCATGA-3′; 5′-AAACTCATGAGCGATGTTACCATTC-3′) and “B” (5′-CACCGCCTCTGCAATCGGTCGGCTA-3′; 5′-AAACTAGCCGACCGATTGCAGAGGC-3′) that were annealed, phosphorylated, and cloned into the dual-expression vector PX458 at BbsI sites. All vectors were confirmed by DNA sequencing.
Cell culture, transfections, retroviral, and lentiviral transductions
Human cell lines PANC1 (CRL-1469), MDA-MB-231 (HTB-26), and MCF10A (CRL-10317) and murine 4T1 (CRL-2539) cells (American Type Culture Collection, Rockville, MD) were cultured as described (11). HMLE and HMLER (CVCL_DG85) cells (a gift from R. Weinberg) were maintained as previously described (88). TBK1 WT and deficient KIC and KPfC murine cell lines were established from single-cell clones isolated from minced and digested tumors. Cells were maintained in Dulbecco’s modified Eagle’s medium (DMEM) supplemented with 10% fetal bovine serum (FBS) (14). Retroviruses were harvested 48 hours after viral vector plasmid transfection of Phoenix A retroviral packaging cells (89). Lentiviruses were collected 48 hours after cotransfection of viral vector and lentiviral packaging plasmids in HEK293 T cells (14). Expi293F cells (A14527, Thermo Fisher Scientific), suspension-adapted HEK 293 cells, were maintained in Expi293 expression medium (A1435102, Thermo Fisher Scientific).
CRISPR KO
Plasmid constructs expressing AKT3 sgRNA or empty vector control were transfected into KPfC cells with Lipofectamine 2000 (11668027, Thermo Fisher Scientific). Positive cells expressing GFP were sorted as single clones and expanded. Each expanded clone was subjected to validation through PCR (AKT3 KO A: 5′-AAACCCTAAAACTGACCTGCAA-3′; 5′-AGGAAAGACCAACTCTCAGCAC-3′; AKT3 KOB: 5′-GACATTATTTGCATTCATCCCA-3′; 5′-GACGCATCCATCTCTTCTTCTC-3′) and immunoblotting.
Human Gas6 preparation and cell stimulation
Human Gas6 from conditioned media was prepared by transfecting Expi293F cells with pRP(Exp)–CMV-huGas6 using ExpiFectamine 293 transfection kit (A14525, Thermo Fisher Scientific). Gas6 expression and carboxylation were tested by immunoblotting. Gas6 concentration was measured by ELISA. For stimulation with Gas6, Panc1 cells were plated in 2 μM warfarin–1% FBS–DMEM, and KIC and KPfC cells were plated in 2 μM warfarin–10% FBS–DMEM 24 hours before stimulation. The cells were preincubated with indicated drugs (2 μM AXL, 10 μM TBK1i, or 10 μM AKT3i for 2 hours; 10 μM mTORC1i or 10 μM mTORC2i for 24 hours before stimulation) and then stimulated with Gas6 (200 ng/ml) in the presence or absence of the inhibitors for 30 min. After stimulation, the cells were washed with phosphate buffered saline (PBS), scraped, spun down, and either immediately lysed and fractionated or stored at −80°C.
Cell fractionation
Cells plated in 10-cm dishes were treated as described, washed with ice-cold PBS, and harvested. Cell fractionation was done as described elsewhere (90). Briefly, cells resuspended in 1 ml of ice-cold 0.1% NP-40–PBS–supplemented Halt protease and phosphatase inhibitor cocktail (PI78442, Thermo Fisher Scientific) and triturated five times with P1000 pipette and transferred to 1.5-ml microcentrifuge tube. An aliquot of the lysate was transferred to a fresh tube, resuspended in Laemmli sample buffer, and labeled as “whole cell lysate (WCL).” The remaining lysate was spun down at 9600g for 10 s. The supernatant was transferred to a fresh tube and labeled as “cytoplasmic fraction.” The pellet containing nuclei was briefly washed with 1 ml of ice-cold 0.1% NP-40–PBS–supplemented Halt protease and phosphatase inhibitor cocktail, spun down at 9600g for 10 s, resuspended in 1 ml of 0.1% NP-40–PBS–supplemented Halt protease and phosphatase inhibitor cocktail and Benzonase nuclease (E1014–5KU, MilliporeSigma), and labeled as “nuclear fraction.”
Immunoprecipitation
Cells plated in 10-cm dishes were treated as described and harvested. To determine phosphorylation status of AXL and AKT isoforms, the cells were lysed in 50 mM tris (pH7.4)–150 mM NaCl–5 mM EDTA–0.1% SDS–0.5% sodium deoxycholate–0.5% NP-40 (RIPA) buffer supplemented with Halt protease and phosphatase inhibitor cocktail and Benzonase nuclease. The lysates were immunoprecipitated with indicated Ab overnight at 4°C. To capture ~90-kDa pAKT3, the lysates were immunoprecipitated for 2 hours at 4°C. For pull-down TBK1:AKT3 complexes, the cells were lysed in RIPA buffer, and the lysates were subjected to immunoprecipitation overnight at 4°C. AKT3:Snail complexes were immunoprecipitated in 1% NP-40–PBS supplemented with Halt protease and phosphatase inhibitor cocktail and Benzonase nuclease. mTORC1/mTORC2 complexes were immunoprecipitated in 50 mM tris (pH7.4)–5 mM EDTA–0.3% CHAPS–150 mM NaCl (TECN) buffer supplemented with Halt protease and phosphatase inhibitor cocktail and Benzonase nuclease. The lysates (250 to 500 μg of protein) were normalized on the basis of total protein concentrations measured with bicinchoninic acid (BCA) protein assay kit (23225, Pierce). The samples were precleared by incubation with protein A/G plus agarose beads (Santa Cruz Biotechnology, sc-2003) and coimmunoprecipitated with indicated Ab and protein A/G plus agarose beads or Ab conjugated to agarose or Sepharose beads (table S2). The samples were washed with the lysis buffer and analyzed by immunoblotting with indicated Ab (table S2).
Immunoblotting
For protein accumulation assays, the cells were lysed in RIPA buffer supplemented with Benzonase nuclease and Halt protease and phosphatase inhibitor cocktail. Lysates were normalized on the basis of total protein concentrations measured with BCA protein assay kit. Otherwise, the samples were prepared as indicated. The samples were separated on 10% SDS-PAGE, transferred to a nitrocellulose membrane, blocked with 3% bovine serum albumin (BSA, A30075, Research Products International) diluted in 20 mM tris base–0.5 M NaCl–0.05% Tween 20 (TBST) buffer, and blotted with the indicated Ab diluted in 3% BSA–or 5% skim milk–TBST buffer.
Enzyme-linked immunosorbent assay
Measuring pAXL protein concentration
Gas6-stimulated and unstimulated Panc1 cells were lysed in RIPA buffer. Lysates were normalized on the basis of total protein concentrations measured with BCA protein assay kit and applied to Nunc Maxi-Sorp 96C white plates (437796, Thermo Fisher Scientific) coated with α-HsAXL Ab 5F11 (BerGenBio). pAXL was detected with α-pAXL-Y866 (BerGenBio) and horseradish peroxidase (HRP)–conjugated goat anti-rabbit (the Jackson Laboratory) Abs. The signal was developed with Supersignal ELISA Femto Substrate (37374, Thermo Fisher Scientific).
Measuring Gas6 protein concentration
Gas6 protein concentration was measured by ELISA with Abs listed in table S2.
In vitro kinase assay
Recombinant GST-AKT3 (1.5 μg, BML-SE369–0005, Enzo Life Sciences) or His-AKT3 (NBP3–13999, Novus Biologicals), 0.1 μg of recombinant active human GST-TBK1 (T02–10G-05, SignalChem), and 1 μl of 10 mM ATP were combined in kinase reaction buffer [20 mM tris-HCl (pH 7.4), 500 mM β-glycerol phosphate, and 12 mM magnesium acetate] up to a total of 30 μl. The kinase reaction was carried out at 30°C, 500 rpm for 1 hour. After the reaction, AKT3 protein was resolved by SDS-PAGE and stained by Coomassie brilliant blue. AKT3 bands were cut out for MS analysis to identify phosphorylation site by the UT Southwestern Proteomics core. The purity of GST-TBK1 preparation was confirmed using MS analysis of complex mixtures.
Confocal microscopy
The cells were plated on coverslips or chambered coverglass and incubated overnight as described. The cells were fixed/permeabilized with 2% formalin or fixed with 4% formaldehyde and permeabilized with 0.2% Triton X-100, stained with indicated Abs, and imaged with Zeiss LSM780 or Zeiss LSM880 inverted microscopes.
Immunohistochemistry
AXL and AKT3 IHC in PDA
Paraffin-embedded human PDA samples were provided by the Tissue Management Shared Resource within the Simmons Comprehensive Cancer Center at UT Southwestern. AXL and AKT3 Abs were optimized and stained using a Leica Autostainer. For Snail and AKT3 costaining in human PDA, IHC was performed as described (91). Briefly, slides were deparaffinized, rehydrated, and then fixed in 10% neutral-buffered formalin for 30 min. Antigen retrieval was performed in antigen retrieval buffer (10 mM tris-HCl and 1 mM EDTA with 10% glycerol) at 110°C for 20 min. Tissue sections were blocked with 2.5% goat serum (S-1012–50; Vector Laboratories), followed by overnight incubation with primary Ab. The slides were washed and incubated with HRP-conjugated secondary Ab for 30 min. Opal substrates were used to develop fluorescence signal (520, FP1487001KT; 570, FP1488001KT; Akoya Biosciences). When performing multiplex IHC, the Abs were stripped after each round of staining by incubating in 10 mM citrate buffer (pH 6.2, 10% glycerol) in a pressure cooker at 110°C for 2 min. The slides were counterstained with 4′,6-diamidino-2-phenylindole and then coverslipped using Prolong Gold (# P36931, Life Technologies). Slides were scanned at 20× using the Zeiss Axioscan.Z1 in the Whole Brain Microscopy Facility of UT Southwestern.
AKT3 IHC in breast cancer
Paraffin-embedded human breast cancer samples were obtained from the Regional Institute of Oncology in Iasi, Romania and assayed for quality control by a pathologist. IHC staining was carried out using DAKO, EnVision FLEX kit with 3,3′-diaminobenzidine (DAB) before counterstaining with hematoxylin (DAKO, EnVision FLEX Hematoxylin K8008). Stained samples were acquired with a Zeiss Axio Observer Z1 microscope and analyzed with TissueGnostics software for acquisition and analysis. Representative regions were analyzed from each sample slide, and mean intensity of DAB-AKT3 staining from nuclei and cytoplasm was used to separate nuclear AKT3 cases from cytoplasmic AKT3 cases.
For AXL, N-cadherin, and Twist-2 IHC in human breast cancer
IHC was performed on 5-μm-thick sections of formalin-fixed, paraffin-embedded biopsy tissues. For AXL analysis, the sections were boiled in tris buffer (pH 6.0) (Dako) for 20 min, followed by incubation overnight at room temperature with goat immunoglobulin G Ab AXL (dilution 1:50, R&D, AF154). For N-cadherin analysis, antigen retrieval was performed by boiling for 20 min in tris EDTA buffer (pH 9) in a microwave oven. A Dako Autostainer was used for staining. The slides were incubated for 60 min at room temperature with a monoclonal Ab against N-cadherin (M3613), dilution 1:25 (Dako). For analysis of Twist-2, antigen retrieval was performed by boiling in TRS buffer (pH 6.0) (Dako) for 25 min and incubated for 1 hour in room temperature with the rabbit polyclonal Ab Twist-2 diluted to 1:500. All reactions were developed with HRP EnVision (Dako) for 30 min at room temperature. The peroxidase was localized by the diaminobenzidine tetrachloride peroxidase reaction and counterstained with Mayer’s hematoxylin.
3D culture experiments
Growth factor–reduced Matrigel (354230, Corning, 10 to 12 mg/ml stock concentration) and bovine (354231, Corning) or rat tail (354236, Corning) collagen I were used for organotypic culture experiments. Vertical invasion assays and experiments in 3D culture were performed and quantified as described previously (92) using a Matrigel/collagen I matrix (3 to 5 mg/ml Matrigel and 1.8 to 2.1 mg/ml collagen I). A 120-μm span on the z axis is shown for the vertical invasion assays.
CHX chase
Parental or AKT3 shRNA (shAKT3)–transduced Panc1 cells were treated with CHX (0.5 μg/ml) in the presence or absence of Gas6 (200 ng/ml) and harvested at indicated time points. Snail accumulation was tested by immunoblotting.
Biochemical assays for AKT activity
Akt isoforms were activated by phosphorylation with PDK1 kinase in the presence of indicated concentrations of AKT3i. The output was measured using the GSK3α-derived LANCE Ultra Ulight–labeled crosstide substrate peptide (TRF0106-M, PerkenElmer) and europium-labeled LANCE Ultra Anti-Phospho-GSK3a (Ser21) Ab (TRF0202-M, PerkinElmer) according to manufacturer instructions. Plates were read with an EnVision multilabel plate reader, excitation at 320 nm and emission at 665 and 615 nm. Results were converted to percent inhibition of phosphorylation by normalizing to positive and negative controls, and compound IC50 was determined using a three-parameter equation (Prism, GraphPad).
Cell viability assay
Panc1 or MDA-MB-231 cells were plated on day 0. On day 1, the medium was replaced with a fresh one containing AKT3i in fourfold dilutions. Eight different drug concentrations were tested with eight replicates per concentration. Relative cell number was determined by an MTS assay: 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymet hoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium was added, and the cells were incubated for 1 to 3 hours at 37°C. Drug sensitivity curves and IC50s were calculated using in-house software. Response was validated in replicate plates (n ≥ 4). Assays were repeated in four biological replicates, with 16 technical replicates each in total.
Gene expression analysis and RNA sequencing
The expression analysis of the breast cancer cell lines and human samples (cancer, normal) was performed from published and GEO-submitted Affymetrix data as described (93). Total RNA from fluorescence-activated cell sorting–enriched primary culture cells were isolated with TRIzol (Invitrogen) and RNeasy Mini column (Qiagen) and evaluated using Bioanalyzer (Agilent Technologies). Gene expression levels were measured using the Illumina HumanHT-12 v4 Expression BeadChip whole-genome expression array. The Illumina Bead Array data were quality controlled in Genome Studio, and both probe level and gene level data were imported into JExpress Pro (http://jexpress.bioinfo.no) for analysis. After quantile normalization, both datasets were log2 transformed. Correspondence analysis (94) was performed on the datasets, together with hierarchical clustering of the samples using a Pearson correlation measure on a per gene mean-centered version of the data. Differentially expressed genes between AXL+ and AXL− groups were identified using the rank product method on both datasets (95). The resulting lists of differentially expressed genes with a false discovery rate value q = 10% from these two analyses were considered differentially expressed between the two groups. The cells were plated on 10-cm dishes until cell densities of 70% were achieved. Total RNA was extracted from cells using QIAGEN RNeasy Mini kit and stored at −80°C. One microgram of total RNA per sample was subjected to library generation using the TruSeq stranded total RNA sample preparation kit, according to the manufacturer’s protocol (Illumina). The libraries were pooled and sequenced on a NextSeq 500 instrument (high output flowcell) at 1 × 75–base pair single-end reads (Illumina). Raw RNA sequencing reads were aligned against to the human genome release GRCh38/hg38 using HISAT2 (96), and exons were counted using RSubread.featureCounts (97). Libraries were filtered to remove gene counts of less than 1 CPM across all libraries and normalized. Differentially expressed genes between GFP control group and AKT3 overexpressing MCF10A cells were calculated using edgeR (98, 99). Genes were considered differentially expressed with a fold change > 2 and P < 0.05.
AKT3 score and METABRIC dataset
To assess the influence of AKT3 signaling and its downstream targets on survival of patients with breast cancer, genes that were found to be differentially expressed after AKT3 overexpression in MCF10A cells were used to generate an AKT3 score. The score essentially represented the sum of expression of 42 differentially expressed genes, adjusted for expected directionality. Initially, we examined 46 different genes, but only 42 of them were represented with probes on the expression array. For genes represented by multiple probes (the 42 genes mapped to 71 different probes), mean signal intensity was used. The influence on breast cancer–specific survival and the putative difference between molecular subtypes was investigated in the Metabric cohort, composed of 1980 patients with breast cancer enrolled at five different hospitals in the UK and Canada (100). Gene expression was assessed using the Illumina HT-12 v3 microarray, and normalized data were downloaded from the European Genome-phenome Archive (EGA) data portal. Missing values were imputed using the impute.knn function as implemented in the R library “impute” with default settings (101). The data were batch adjusted for hospital effect using the pamr.batchadjust function in the “pamr” library with default settings (102). Association between the score and molecular subtypes (100, 103) was tested using Kruskal-Wallis rank test, and correlations were estimated with Spearman’s rank correlation. Survival analyses were performed using a Cox proportional hazards regression model as implemented in the R library “rms” (104). Survival plots were generated using the survplot function, as implemented in the rms library. All analyses were performed using R version 3.3.1.
Animal studies
Syngenic PDA cells (KPfC: CAS9-EV, AKT3 KO, Rescue) were injected orthotopically (2.5 × 105 cells) in 6- to 8-week-old female C57BL/6 mice (stock # 000664, the Jackson Laboratory). Nineteen days after tumor cell injection, the mice were euthanized and organs were harvested for analysis. All animals were housed in a pathogen-free facility with 24-hour access to food and water. Animal experiments in this study were approved by and performed in accordance with the institutional animal care and use committee at the UTSW Medical Center at Dallas. Before implantation, the cells were confirmed to be pathogen free.
Statistical analysis
Comparisons of histological staining index (SI) groups were performed by Pearson χ2 test using cut-off values for SI categories based on median values. Fisher’s exact tests were used to investigate the association between nuclear and cytoplasmic AKT3 and AXL in human PDA. Kruskal-Wallis tests were used to determine whether there was a significant difference in continuous outcomes among more than two groups. If a Kruskal-Wallis test showed a significant difference, Dunn’s multiple comparisons tests were conducted to examine which pairs of groups yielded significant differences. GraphPad Prism 5.0 for PC and MATLAB were used for statistical analysis.
Supplementary Material
Acknowledgments:
We dedicate this work to our wonderful colleague, Crina Trion, who passed away on 21 May 2024. We thank S. V. Berge, K. Sputova, E. Verwayen, H. Haugen, M. Enger, M. Scottn, A. Boniecka, and E. Milde for excellent technical support; M. LaBarge for conceptual support and resources; K. Petersen; the Computational Biology Unit; the University of Bergen; and the Norwegian Bioinformatics platform for microarray analysis support. In addition, we thank our UT Southwestern colleagues L. Hu and Q. Zhang for input and technical advice. We would also like to thank the UT Southwestern Proteomics core, C. Lewis, director of the Shared Tissue Management Resource Core (supported by P30 CA142543), and A. Mobley, manager of the Flow Cytometry core.
Funding:
The work was supported by NIH grants R01 CA192381, R01 CA243577, and U54 CA210181 Project 2 to R.A.B.; the Effie Marie Cain Scholarship in Angiogenesis Research and the Gillson Longenbaugh Foundation to R.A.B.; grants from the Norwegian Cancer Society; Norwegian Research Council; Bergen Health Authority and BerGenBio ASA to J.B.L.; American Cancer Society – Fred Ross Desert Spirit Postdoctoral Fellowship (PF-21–184-01-CSM) to S.H.; the Sigrid Juselius Foundation and Finnish Cancer Institute to J.I. and R.V.; as well as CPRIT RP160157 (principal investigator: M. Cobb, UT Southwestern, Dallas, Texas) and NCI F99 CA253718 to E.N.A. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
Footnotes
Competing interests: R.A.B. received research support from BerGenBio ASA for unrelated work; S.H., A.M., K.Y.A., G.G., D.M., M.B., and J.B.L. are or were employees of BerGenBio ASA. J.B.L. and D.M. have ownership interest in BerGenBio ASA. R.V. is a current employee at Orion Corporation. The other authors declare that they have no competing interests.
Data and materials availability:
All data needed to evaluate the conclusions in the paper are present in the paper or the Supplementary Materials. RNA sequencing data are accessible through the gene expression omnibus (accession number GSE274874). Requests for reagents described in this study will be fulfilled by J.B.L. and R.A.B., except for the AKT3i (BGB214), request for which should be directed to BerGenBio ASA.
REFERENCES AND NOTES
- 1.Wang Z, Li Y, Ahmad A, Banerjee S, Azmi AS, Kong D, Sarkar FH, Pancreatic cancer: Understanding and overcoming chemoresistance. Nat. Rev. Gastroenterol. Hepatol. 8, 27–33 (2011). [DOI] [PubMed] [Google Scholar]
- 2.Tran DD, Corsa CAS, Biswas H, Aft RL, Longmore GD, Temporal and spatial cooperation of Snail1 and Twist1 during epithelial-mesenchymal transition predicts for human breast cancer recurrence. Mol. Cancer Res. 9, 1644–1657 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Seyfried TN, Huysentruyt LC, On the origin of cancer metastasis. Crit. Rev. Oncog. 18, 43–73 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Cates JM, Byrd RH, Fohn LE, Tatsas AD, Washington MK, Black CC, Epithelial-mesenchymal transition markers in pancreatic ductal adenocarcinoma. Pancreas 38, e1–e6 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kudo-Saito C, Shirako H, Takeuchi T, Kawakami Y, Cancer metastasis is accelerated through immunosuppression during Snail-induced EMT of cancer cells. Cancer Cell 15, 195–206 (2009). [DOI] [PubMed] [Google Scholar]
- 6.Puls TJ, Tan X, Whittington CF, Voytik-Harbin SL, 3D collagen fibrillar microstructure guides pancreatic cancer cell phenotype and serves as a critical design parameter for phenotypic models of EMT. PLOS ONE 12, e0188870 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Larue L, Bellacosa A, Epithelial-mesenchymal transition in development and cancer: Role of phosphatidylinositol 3′ kinase/AKT pathways. Oncogene 24, 7443–7454 (2005). [DOI] [PubMed] [Google Scholar]
- 8.Wasif N, Ko CY, Farrell J, Wainberg Z, Hines OJ, Reber H, Tomlinson JS, Impact of tumor grade on prognosis in pancreatic cancer: Should we include grade in AJCC staging? Ann. Surg. Oncol. 17, 2312–2320 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Kirane A, Ludwig KF, Sorrelle N, Haaland G, Sandal T, Ranaweera R, Toombs JE, Wang M, Dineen SP, Micklem D, Dellinger MT, Lorens JB, Brekken RA, Warfarin blocks Gas6-mediated Axl activation required for pancreatic cancer epithelial plasticity and metastasis. Cancer Res. 75, 3699–3705 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Koorstra J-BM, Karikari CA, Feldmann G, Bisht S, Rojas PL, Offerhaus GJA, Alvarez H, Maitra A, The Axl receptor tyrosine kinase confers an adverse prognostic influence in pancreatic cancer and represents a new therapeutic target. Cancer Biol. Ther. 8, 618–626 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Gjerdrum C, Tiron C, Hoiby T, Stefansson I, Haugen H, Sandal T, Collett K, Li S, McCormack E, Gjertsen BT, Micklem DR, Akslen LA, Glackin C, Lorens JB, Axl is an essential epithelial-to-mesenchymal transition-induced regulator of breast cancer metastasis and patient survival. Proc. Natl. Acad. Sci. U.S.A. 107, 1124–1129 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Ludwig KF, Du W, Sorrelle NB, Wnuk-Lipinska K, Topalovski M, Toombs JE, Cruz VH, Yabuuchi S, Rajeshkumar NV, Maitra A, Lorens JB, Brekken RA, Small-molecule inhibition of Axl targets tumor immune suppression and enhances chemotherapy in pancreatic cancer. Cancer Res. 78, 246–255 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Du W, Brekken RA, Does Axl have potential as a therapeutic target in pancreatic cancer? Expert Opin. Ther. Targets 22, 955–966 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Cruz VH, Arner EN, Du W, Bremauntz AE, Brekken RA, Axl-mediated activation of TBK1 drives epithelial plasticity in pancreatic cancer. JCI Insight 5, e126117 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ou Y-H, Torres M, Ram R, Formstecher E, Roland C, Cheng T, Brekken R, Wurz R, Tasker A, Polverino T, Tan S-L, White MA, TBK1 directly engages Akt/PKB survival signaling to support oncogenic transformation. Mol. Cell 41, 458–470 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bellacosa A, Testa JR, Staal SP, Tsichlis PN, A retroviral oncogene, akt, encoding a serine-threonine kinase containing an SH2-like region. Science 254, 274–277 (1991). [DOI] [PubMed] [Google Scholar]
- 17.Grille SJ, Bellacosa A, Upson J, Klein-Szanto AJ, van Roy F, Lee-Kwon W, Donowitz M, Tsichlis PN, Larue L, The protein kinase Akt induces epithelial mesenchymal transition and promotes enhanced motility and invasiveness of squamous cell carcinoma lines. Cancer Res. 63, 2172–2178 (2003). [PubMed] [Google Scholar]
- 18.Xu W, Yang Z, Lu N, A new role for the PI3K/Akt signaling pathway in the epithelial-mesenchymal transition. Cell Adh. Migr. 9, 317–324 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Woodgett JR, Recent advances in the protein kinase B signaling pathway. Curr. Opin. Cell Biol. 17, 150–157 (2005). [DOI] [PubMed] [Google Scholar]
- 20.Irie HY, Pearline RV, Grueneberg D, Hsia M, Ravichandran P, Kothari N, Natesan S, Brugge JS, Distinct roles of Akt1 and Akt2 in regulating cell migration and epithelial-mesenchymal transition. J. Cell Biol. 171, 1023–1034 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Sanidas I, Polytarchou C, Hatziapostolou M, Ezell SA, Kottakis F, Hu L, Guo A, Xie J, Comb MJ, Iliopoulos D, Tsichlis PN, Phosphoproteomics screen reveals akt isoform-specific signals linking RNA processing to lung cancer. Mol. Cell 53, 577–590 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Lee H-J, Jeng Y-M, Chen Y-L, Chung L, Yuan R-H, Gas6/Axl pathway promotes tumor invasion through the transcriptional activation of Slug in hepatocellular carcinoma. Carcinogenesis 35, 769–775 (2014). [DOI] [PubMed] [Google Scholar]
- 23.Tang Z, Li C, Kang B, Gao G, Li C, Zhang Z, GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45, W98–W102 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Li R, Wei J, Jiang C, Liu D, Deng L, Zhang K, Wang P, Akt SUMOylation regulates cell proliferation and tumorigenesis. Cancer Res. 73, 5742–5753 (2013). [DOI] [PubMed] [Google Scholar]
- 25.Risso G, Pelisch F, Pozzi B, Mammi P, Blaustein M, Colman-Lerner A, Srebrow A, Modification of Akt by SUMO conjugation regulates alternative splicing and cell cycle. Cell Cycle 12, 3165–3174 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Marchlik E, Thakker P, Carlson T, Jiang Z, Ryan M, Marusic S, Goutagny N, Kuang W, Askew GR, Roberts V, Benoit S, Zhou T, Ling V, Pfeifer R, Stedman N, Fitzgerald KA, Lin L-L, Hall JP, Mice lacking Tbk1 activity exhibit immune cell infiltrates in multiple tissues and increased susceptibility to LPS-induced lethality. J. Leukoc. Biol. 88, 1171–1180 (2010). [DOI] [PubMed] [Google Scholar]
- 27.Mende I, Malstrom S, Tsichlis PN, Vogt PK, Aoki M, Oncogenic transformation induced by membrane-targeted Akt2 and Akt3. Oncogene 20, 4419–4423 (2001). [DOI] [PubMed] [Google Scholar]
- 28.Alessi DR, James SR, Downes CP, Holmes AB, Gaffney PRJ, Reese CB, Cohen P, Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Bα. Curr. Biol. 7, 261–269 (1997). [DOI] [PubMed] [Google Scholar]
- 29.Girardi C, James P, Zanin S, Pinna LA, Ruzzene M, Differential phosphorylation of Akt1 and Akt2 by protein kinase CK2 may account for isoform specific functions. Biochim. Biophys. Acta 1843, 1865–1874 (2014). [DOI] [PubMed] [Google Scholar]
- 30.Guo H, Gao M, Lu Y, Liang J, Lorenzi PL, Bai S, Hawke DH, Li J, Dogruluk T, Scott KL, Jonasch E, Mills GB, Ding Z, Coordinate phosphorylation of multiple residues on single AKT1 and AKT2 molecules. Oncogene 33, 3463–3472 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Sarbassov DD, Guertin DA, Ali SM, Sabatini DM, Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 307, 1098–1101 (2005). [DOI] [PubMed] [Google Scholar]
- 32.Degan SE, Gelman IH, Emerging roles for AKT isoform preference in cancer progression pathways. Mol. Cancer Res. 19, 1251–1257 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Yang G, Murashige DS, Humphrey SJ, James DE, A positive feedback loop between Akt and mTORC2 via SIN1 phosphorylation. Cell Rep. 12, 937–943 (2015). [DOI] [PubMed] [Google Scholar]
- 34.Yu Z, Chen J, Takagi E, Wang F, Saha B, Liu X, Joubert L-M, Gleason CE, Jin M, Li C, Nowotny C, Agard D, Cheng Y, Pearce D, Interactions between mTORC2 core subunits Rictor and mSin1 dictate selective and context-dependent phosphorylation of substrate kinases SGK1 and Akt. J. Biol. Chem. 298, 102288 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bodur C, Kazyken D, Huang K, Ekim Ustunel B, Siroky KA, Tooley AS, Gonzalez IE, Foley DH, Acosta-Jaquez HA, Barnes TM, Steinl GK, Cho K-W, Lumeng CN, Riddle SM, Myers MG Jr., D. C. Fingar, The IKK-related kinase TBK1 activates mTORC1 directly in response to growth factors and innate immune agonists. EMBO J. 37, 19–38 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Saji M, Vasko V, Kada F, Allbritton EH, Burman KD, Ringel MD, Akt1 contains a functional leucine-rich nuclear export sequence. Biochem. Biophys. Res. Commun. 332, 167–173 (2005). [DOI] [PubMed] [Google Scholar]
- 37.Santi SA, Lee H, The Akt isoforms are present at distinct subcellular locations. Am. J. Physiol. Cell Physiol. 298, C580–C591 (2010). [DOI] [PubMed] [Google Scholar]
- 38.S. R. Wente, M. P. Rout, The nuclear pore complex and nuclear transport. Cold Spring Harb. Perspect. Biol. 2, a000562 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Kosugi S, Hasebe M, Tomita M, Yanagawa H, Systematic identification of cell cycle-dependent yeast nucleocytoplasmic shuttling proteins by prediction of composite motifs. Proc. Natl. Acad. Sci. U.S.A. 106, 10171–10176 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Elenbaas B, Spirio L, Koerner F, Fleming MD, Zimonjic DB, Donaher JL, Popescu NC, Hahn WC, Weinberg RA, Human breast cancer cells generated by oncogenic transformation of primary mammary epithelial cells. Genes Dev. 15, 50–65 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Zhou BP, Deng J, Xia W, Xu J, Li YM, Gunduz M, Hung M-C, Dual regulation of Snail by GSK-3beta-mediated phosphorylation in control of epithelial-mesenchymal transition. Nat. Cell Biol. 6, 931–940 (2004). [DOI] [PubMed] [Google Scholar]
- 42.Hudis C, Swanton C, Janjigian YY, Lee R, Sutherland S, Lehman R, Chandarlapaty S, Hamilton N, Gajria D, Knowles J, Shah J, Shannon K, Tetteh E, Sullivan DM, Moreno C, Yan L, Han HS, A phase 1 study evaluating the combination of an allosteric AKT inhibitor (MK-2206) and trastuzumab in patients with HER2-positive solid tumors. Breast Cancer Res. 15, R110 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Yap TA, Yan L, Patnaik A, Tunariu N, Biondo A, Fearen I, Papadopoulos KP, Olmos D, Baird R, Delgado L, Tetteh E, Beckman RA, Lupinacci L, Riisnaes R, Decordova S, Heaton SP, Swales K, deSouza NM, Leach MO, Garrett MD, Sullivan DM, de Bono JS, Tolcher AW, Interrogating two schedules of the AKT inhibitor MK-2206 in patients with advanced solid tumors incorporating novel pharmacodynamic and functional imaging biomarkers. Clin. Cancer Res. 20, 5672–5685 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ma BBY, Goh BC, Lim WT, Hui EP, Tan EH, de Lima Lopes G, Lo KW, Li L, Loong H, Foster NR, Erlichman C, King AD, Kam MKM, Leung SF, Chan KC, Chan ATC, Multicenter phase II study of the AKT inhibitor MK-2206 in recurrent or metastatic nasopharyngeal carcinoma from patients in the mayo phase II consortium and the cancer therapeutics research group (MC1079). Invest. New Drugs 33, 985–991 (2015). [DOI] [PubMed] [Google Scholar]
- 45.Jansen VM, Mayer IA, Arteaga CL, Is there a future for AKT inhibitors in the treatment of cancer? Clin. Cancer Res. 22, 2599–2601 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Barnett SF, Defeo-Jones D, Fu S, Hancock PJ, Haskell KM, Jones RE, Kahana JA, Kral AM, Leander K, Lee LL, Malinowski J, McAvoy EM, Nahas DD, Robinson RG, Huber HE, Identification and characterization of pleckstrin-homology-domain-dependent and isoenzyme-specific Akt inhibitors. Biochem. J. 385, 399–408 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Lindsley CW, Zhao Z, Leister WH, Robinson RG, Barnett SF, Defeo-Jones D, Jones RE, Hartman GD, Huff JR, Huber HE, Duggan ME, Allosteric Akt (PKB) inhibitors: Discovery and SAR of isozyme selective inhibitors. Bioorg. Med. Chem. Lett. 15, 761–764 (2005). [DOI] [PubMed] [Google Scholar]
- 48.Wu W-I, Voegtli WC, Sturgis HL, Dizon FP, Vigers GPA, Brandhuber BJ, Crystal structure of human AKT1 with an allosteric inhibitor reveals a new mode of kinase inhibition. PLOS ONE 5, e12913 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Cao D, Maitra A, Saavedra J-A, Klimstra DS, Adsay NV, Hruban RH, Expression of novel markers of pancreatic ductal adenocarcinoma in pancreatic nonductal neoplasms: Additional evidence of different genetic pathways. Mod. Pathol. 18, 752–761 (2005). [DOI] [PubMed] [Google Scholar]
- 50.Anwar T, Rufail ML, Djomehri SI, Gonzalez ME, de la Vega LL, Tomlins SA, Newman LA, Kleer CG, Next-generation sequencing identifies recurrent copy number variations in invasive breast carcinomas from Ghana. Mod. Pathol. 33, 1537–1545 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Meric-Bernstam F, Frampton GM, Ferrer-Lozano J, Yelensky R, Perez-Fidalgo JA, Wang Y, Palmer GA, Ross JS, Miller VA, Su X, Eroles P, Barrera JA, Burgues O, Lluch AM, Zheng X, Sahin A, Stephens PJ, Mills GB, Cronin MT, Gonzalez-Angulo AM, Concordance of genomic alterations between primary and recurrent breast cancer. Mol. Cancer Ther. 13, 1382–1389 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Bonin S, Pracella D, Barbazza R, Dotti I, Boffo S, Stanta G, PI3K/AKT signaling in breast cancer molecular subtyping and lymph node involvement. Dis. Markers 2019, 7832376 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Tanaka M, Siemann DW, Gas6/Axl signaling pathway in the tumor immune microenvironment. Cancer 12, 1850 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zhu C, Wei Y, Wei X, AXL receptor tyrosine kinase as a promising anti-cancer approach: Functions, molecular mechanisms and clinical applications. Mol. Cancer 18, 153 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hinz N, Jucker M, Distinct functions of AKT isoforms in breast cancer: A comprehensive review. Cell Commun. Signal 17, 154 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Tsai P-J, Lai Y-H, Manne RK, Tsai Y-S, Sarbassov D, Lin H-K, Akt: A key transducer in cancer. J. Biomed. Sci. 29, 76 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Konishi H, Kuroda S, Tanaka M, Matsuzaki H, Ono Y, Kameyama K, Haga T, Kikkawa U, Molecular cloning and characterization of a new member of the RAC protein kinase family: Association of the pleckstrin homology domain of three types of RAC protein kinase with protein kinase C subspecies and beta gamma subunits of G proteins. Biochem. Biophys. Res. Commun. 216, 526–534 (1995). [DOI] [PubMed] [Google Scholar]
- 58.Santi SA, Lee H, Ablation of Akt2 induces autophagy through cell cycle arrest, the downregulation of p70S6K, and the deregulation of mitochondria in MDA-MB231 cells. PLOS ONE 6, e14614 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Stottrup C, Tsang T, Chin YR, Upregulation of AKT3 confers resistance to the AKT inhibitor MK2206 in breast cancer. Mol. Cancer Ther. 15, 1964–1974 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Li Y, Cai B, Shen L, Dong Y, Lu Q, Sun S, Liu S, Ma S, Ma PX, Chen J, MiRNA-29b suppresses tumor growth through simultaneously inhibiting angiogenesis and tumorigenesis by targeting Akt3. Cancer Lett. 397, 111–119 (2017). [DOI] [PubMed] [Google Scholar]
- 61.Grottke A, Ewald F, Lange T, Norz D, Herzberger C, Bach J, Grabinski N, Graser L, Hoppner F, Nashan B, Schumacher U, Jucker M, Downregulation of AKT3 increases migration and metastasis in triple negative breast cancer cells by upregulating S100A4. PLOS ONE 11, e0146370 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Chin YR, Yoshida T, Marusyk A, Beck AH, Polyak K, Toker A, Targeting Akt3 signaling in triple-negative breast cancer. Cancer Res. 74, 964–973 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Chung S, Yao J, Suyama K, Bajaj S, Qian X, Loudig OD, Eugenin EA, Phillips GR, Hazan RB, N-cadherin regulates mammary tumor cell migration through Akt3 suppression. Oncogene 32, 422–430 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Mure H, Matsuzaki K, Kitazato KT, Mizobuchi Y, Kuwayama K, Kageji T, Nagahiro S, Akt2 and Akt3 play a pivotal role in malignant gliomas. Neuro Oncol. 12, 221–232 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Nakatani K, Sakaue H, Thompson DA, Weigel RJ, Roth RA, Identification of a human Akt3 (protein kinase B γ) which contains the regulatory serine phosphorylation site. Biochem. Biophys. Res. Commun. 257, 906–910 (1999). [DOI] [PubMed] [Google Scholar]
- 66.Nakatani K, Thompson DA, Barthel A, Sakaue H, Liu W, Weigel RJ, Roth RA, Up-regulation of Akt3 in estrogen receptor-deficient breast cancers and androgen-independent prostate cancer lines. J. Biol. Chem. 274, 21528–21532 (1999). [DOI] [PubMed] [Google Scholar]
- 67.Stahl JM, Sharma A, Cheung M, Zimmerman M, Cheng JQ, Bosenberg MW, Kester M, Sandirasegarane L, Robertson GP, Deregulated Akt3 activity promotes development of malignant melanoma. Cancer Res. 64, 7002–7010 (2004). [DOI] [PubMed] [Google Scholar]
- 68.Suyama K, Yao J, Liang H, Benard O, Loudig OD, Amgalan D, McKimpson WM, Phillips GR, Segall J, Wang Y, Fineberg S, Norton L, Kitsis RN, Hazan RB, An Akt3 splice variant lacking the serine 472 phosphorylation site promotes apoptosis and suppresses mammary tumorigenesis. Cancer Res. 78, 103–114 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Tooley AS, Kazyken D, Bodur C, Gonzalez IE, Fingar DC, The innate immune kinase TBK1 directly increases mTORC2 activity and downstream signaling to Akt. J. Biol. Chem. 297, 100942 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Yao Q, Li H, Liu B-Q, Huang X-Y, Guo L, SUMOylation-regulated protein phosphorylation, evidence from quantitative phosphoproteomics analyses. J. Biol. Chem. 286, 27342–27349 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Zhao X, SUMO-mediated regulation of nuclear functions and signaling processes. Mol. Cell 71, 409–418 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Lin F-M, Yost SE, Wen W, Frankel PH, Schmolze D, Chu P-G, Yuan Y-C, Liu Z, Yim J, Chen Z, Yuan Y, Differential gene expression and AKT targeting in triple negative breast cancer. Oncotarget 10, 4356–4368 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Turner KM, Sun Y, Ji P, Granberg KJ, Bernard B, Hu L, Cogdell DE, Zhou X, Yli-Harja O, Nykter M, Shmulevich I, Yung WKA, Fuller GN, Zhang W, Genomically amplified Akt3 activates DNA repair pathway and promotes glioma progression. Proc. Natl. Acad. Sci. U.S.A. 112, 3421–3426 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Toulany M, Maier J, Iida M, Rebholz S, Holler M, Grottke A, Juker M, Wheeler DL, Rothbauer U, Rodemann HP, Akt1 and Akt3 but not Akt2 through interaction with DNA-PKcs stimulate proliferation and post-irradiation cell survival of K-RAS-mutated cancer cells. Cell Death Discov. 3, 17072 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Peres J, Mowla S, Prince S, The T-box transcription factor, TBX3, is a key substrate of AKT3 in melanomagenesis. Oncotarget 6, 1821–1833 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Tian Y, Li J, Cheung TC, Tam V, Lau CG, Wang X, Chin YR, Akt3 promotes cancer stemness in triple-negative breast cancer through YB1-Snail/Slug signaling axis. Genes Dis. 10, 301–306 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Muqbil I, Wu J, Aboukameel A, Mohammad RM, Azmi AS, Snail nuclear transport: The gateways regulating epithelial-to-mesenchymal transition? Semin. Cancer Biol. 27, 39–45 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Qiu Z, Dong B, Guo W, Piotr R, Longmore G, Yang X, Yu Z, Deng J, Evers BM, Wu Y, STK39 promotes breast cancer invasion and metastasis by increasing SNAI1 activity upon phosphorylation. Theranostics 11, 7658–7670 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Zhang K, Rodriguez-Aznar E, Yabuta N, Owen RJ, Mingot JM, Nojima H, Nieto MA, Longmore GD, Lats2 kinase potentiates Snail1 activity by promoting nuclear retention upon phosphorylation. EMBO J. 31, 29–43 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Johnson JL, Yaron TM, Huntsman EM, Kerelsky A, Song J, Regev A, Lin T-Y, Liberatore K, Cizin DM, Cohen BM, Vasan N, Ma Y, Krismer K, Robles JT, van de Kooij B, van Vlimmeren AE, Andree-Busch N, Kaufer NF, Dorovkov MV, Ryazanov AG, Takagi Y, Kastenhuber ER, Goncalves MD, Hopkins BD, Elemento O, Taatjes DJ, Maucuer A, Yamashita A, Degterev A, Uduman M, Lu J, Landry SD, Zhang B, Cossentino I, Linding R, Blenis J, Hornbeck PV, Turk BE, Yaffe MB, Cantley LC, An atlas of substrate specificities for the human serine/threonine kinome. Nature 613, 759–766 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ouchida AT, Kacal M, Zheng A, Ambroise G, Zhang B, Norberg E, Vakifahmetoglu-Norberg H, USP10 regulates the stability of the EMT-transcription factor Slug/SNAI2. Biochem. Biophys. Res. Commun. 502, 429–434 (2018). [DOI] [PubMed] [Google Scholar]
- 82.Yu Q, Zhou BP, Wu Y, The regulation of snail: On the ubiquitin edge. Cancer Cell Microenviron. 4, e1567 (2017). [PMC free article] [PubMed] [Google Scholar]
- 83.Geissert R, Lammert A, Wirth S, Honig R, Lohfink D, Unger M, Pek D, Schluter K, Scheftschik T, Smit DJ, Jucker M, Menke A, Giehl K, K-Ras(V12) differentially affects the three Akt isoforms in lung and pancreatic carcinoma cells and upregulates E-cadherin and NCAM via Akt3. Cell Commun. Signal 22, 85 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Xiao J, Li W, Zheng X, Qi L, Wang H, Zhang C, Wan X, Zheng Y, Zhong R, Zhou X, Lu Y, Li Z, Qiu Y, Liu C, Zhang F, Zhang Y, Xu X, Yang Z, Chen H, Zhai Q, Wei B, Wang H, Targeting 7-dehydrocholesterol reductase integrates cholesterol metabolism and IRF3 activation to eliminate infection. Immunity 52, 109–122 e106 (2020). [DOI] [PubMed] [Google Scholar]
- 85.Gu S, Dai H, Zhao X, Gui C, Gui J, AKT3 deficiency in M2 macrophages impairs cutaneous wound healing by disrupting tissue remodeling. Aging 12, 6928–6946 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Boehm JS, Zhao JJ, Yao J, Kim SY, Firestein R, Dunn IF, Sjostrom SK, Garraway LA, Weremowicz S, Richardson AL, Greulich H, Stewart CJ, Mulvey LA, Shen RR, Ambrogio L, Hirozane-Kishikawa T, Hill DE, Vidal M, Meyerson M, Grenier JK, Hinkle G, Root DE, Roberts TM, Lander ES, Polyak K, Hahn WC, Integrative genomic approaches identify IKBKE as a breast cancer oncogene. Cell 129, 1065–1079 (2007). [DOI] [PubMed] [Google Scholar]
- 87.Horton RM, Hunt HD, Ho SN, Pullen JK, Pease LR, Engineering hybrid genes without the use of restriction enzymes: Gene splicing by overlap extension. Gene 77, 61–68 (1989). [DOI] [PubMed] [Google Scholar]
- 88.Mani SA, Guo W, Liao M-J, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, Campbell LL, Polyak K, Brisken C, Yang J, Weinberg RA, The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133, 704–715 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Swift S, Lorens J, Achacoso P, Nolan GP, Rapid production of retroviruses for efficient gene delivery to mammalian cells using 293T cell-based systems. Curr. Protoc. Immunol. 10, 10.1002/0471142735.im1017cs31 (2021). [DOI] [PubMed] [Google Scholar]
- 90.Nabbi A, Riabowol K, Rapid isolation of nuclei from cells in vitro. Cold Spring Harb. Protoc. 2015, 769–772 (2015). [DOI] [PubMed] [Google Scholar]
- 91.Sorrelle N, Ganguly D, Dominguez ATA, Zhang Y, Huang H, Dahal LN, Burton N, Ziemys A, Brekken RA, Improved multiplex immunohistochemistry for immune microenvironment evaluation of mouse formalin-fixed, paraffin-embedded tissues. J. Immunol. 202, 292–299 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Westcott JM, Prechtl AM, Maine EA, Dang TT, Esparza MA, Sun H, Zhou Y, Xie Y, Pearson GW, An epigenetically distinct breast cancer cell subpopulation promotes collective invasion. J. Clin. Invest. 125, 1927–1943 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Kilpinen S, Autio R, Ojala K, Iljin K, Bucher E, Sara H, Pisto T, Saarela M, Skotheim RI, Bjorkman M, Mpindi J-P, Haapa-Paananen S, Vainio P, Edgren H, Wolf M, Astola J, Nees M, Hautaniemi S, Kallioniemi O, Systematic bioinformatic analysis of expression levels of 17,330 human genes across 9,783 samples from 175 types of healthy and pathological tissues. Genome Biol. 9, R139 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Fellenberg K, Hauser NC, Brors B, Neutzner A, Hoheisel JD, Vingron M, Correspondence analysis applied to microarray data. Proc. Natl. Acad. Sci. U.S.A. 98, 10781–10786 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Breitling R, Armengaud P, Amtmann A, Herzyk P, Rank products: A simple, yet powerful, new method to detect differentially regulated genes in replicated microarray experiments. FEBS Lett. 573, 83–92 (2004). [DOI] [PubMed] [Google Scholar]
- 96.Kim D, Paggi JM, Park C, Bennett C, Salzberg SL, Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 37, 907–915 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Liao Y, Smyth GK, Shi W, featureCounts: An efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014). [DOI] [PubMed] [Google Scholar]
- 98.McCarthy DJ, Chen Y, Smyth GK, Differential expression analysis of multifactor RNA-seq experiments with respect to biological variation. Nucleic Acids Res. 40, 4288–4297 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Robinson MD, McCarthy DJ, Smyth GK, edgeR: A bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Curtis C, Shah SP, Chin S-F, Turashvili G, Rueda OM, Dunning MJ, Speed D, Lynch AG, Samarajiwa S, Yuan Y, Gräf S, Ha G, Haffari G, Bashashati A, Russell R, McKinney S, METABRIC Group A Green Langerød, A., Provenzano E, Wishart G, Pinder S, Watson P, Markowetz F, Murphy L, Ellis I, Purushotham A, Børresen-Dale A-L, Brenton JD, Tavare S, Caldas C, Aparicio S, The genomic and transcriptomic architecture of 2,000 breast tumours reveals novel subgroups. Nature 486, 346–352 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Hastie T, Tibshirani R, Narasimhan B, Chu G, impute: Impute: Imputation for microarray data. R package version 1.68.0 (2021); 10.18129/B9.bioc.impute. [DOI] [Google Scholar]
- 102.Hastie T, Tibshirani R, Narasimhan B, Chu G, pamr: Pam: Prediction analysis for microarrays, version 1.57 (2024); 10.32614/CRAN.package.pamr. [DOI] [Google Scholar]
- 103.Parker JS, Mullins M, Cheang MCU, Leung S, Voduc D, Vickery T, Davies S, Fauron C, He X, Hu Z, Quackenbush JF, Stijleman IJ, Palazzo J, Marron JS, Nobel AB, Mardis E, Nielsen TO, Ellis MJ, Perou CM, Bernard PS, Supervised risk predictor of breast cancer based on intrinsic subtypes. J. Clin. Oncol. 27, 1160–1167 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Harrell FE Jr., Regression Modeling Strategies: With Applications to Linear Models, Logistic and Ordinal Regression, and Survival Analysis (Springer, 2015). [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Data Availability Statement
All data needed to evaluate the conclusions in the paper are present in the paper or the Supplementary Materials. RNA sequencing data are accessible through the gene expression omnibus (accession number GSE274874). Requests for reagents described in this study will be fulfilled by J.B.L. and R.A.B., except for the AKT3i (BGB214), request for which should be directed to BerGenBio ASA.