Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Therapeutics, Targets, and Chemical Biology

Elevation of Receptor Tyrosine Kinases by Small Molecule AKT Inhibitors in Prostate Cancer Is Mediated by Pim-1

Bo Cen, Sandeep Mahajan, Wenxue Wang and Andrew S. Kraft
Bo Cen
1Department of Medicine; and 2The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Sandeep Mahajan
1Department of Medicine; and 2The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Wenxue Wang
1Department of Medicine; and 2The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Andrew S. Kraft
1Department of Medicine; and 2The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
1Department of Medicine; and 2The Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI: 10.1158/0008-5472.CAN-12-4619 Published June 2013
  • Article
  • Figures & Data
  • Info & Metrics
  • PDF
Loading

Abstract

The PI3K/AKT pathway is hyperactivated in prostate cancer but its effective therapeutic targeting has proven difficult. In particular, the antitumor activity of AKT inhibitors is attenuated by upregulation of receptor tyrosine kinases (RTK) through an uncharacterized feedback mechanism. In this report, we show that RNA interference-mediated silencing or pharmacologic inhibition of Pim-1 activity curtails AKT inhibitor-induced upregulation of RTKs in prostate cancer cells. Although Pim kinases have been implicated in cap-dependent translational control, we find that in the context of AKT inhibition, the expression of RTKs is controlled by Pim-1 in a cap-independent manner by controlling internal ribosome entry. Combination of Pim and AKT inhibitors resulted in synergistic inhibition of prostate tumor growth in vitro and in vivo. Together, our results show that Pim-1 mediates resistance to AKT inhibition and suggest its targeting to improve the efficacy of AKT inhibitors in anticancer therapy. Cancer Res; 73(11); 3402–11. ©2013 AACR.

Introduction

The PI3K/AKT pathway is commonly activated in human cancer and controls cellular processes that contribute to the initiation and maintenance of cancer (1). It is activated in 40% of primary and 70% of metastatic prostate cancers secondary to mutations or deletions in PTEN (1–3). Activation of the pathway can be associated with mutations in the phosphoinositide 3-kinase (PI3K) catalytic subunit P110 α and regulatory subunit (1), mutations in each of the 3 AKT isoforms (1, 4), and activation of receptor tyrosine kinases (RTK) by mutation (e.g., EGF receptor; EGFR) or gene amplification (e.g., HER2), which can result in activation of downstream PI3K/AKT (1, 5). Multiple small-molecule inhibitors have been developed to target PI3K/mTOR or AKT (6), but the efficacy of these drugs is compromised by the stimulation of compensatory signaling pathways that have the potential to enhance tumor growth (7–9). There is accumulating evidence that inhibition of the PI3K/AKT pathway can lead to adaptive resistance due to upregulation and activation of RTKs (7–9). The mechanism underlying the AKT inhibition-induced upregulation of some of these RTKs, including HER3, INSR, and insulin-like growth factor-1 receptor (IGF-IR), has been shown to, in part, involve FOXO transcription factors (7); however, these transcription factors do not seem to be involved in the AKT inhibition-induced upregulation of other RTKs, including MET, HER2, and RET (7).

The Pim family of serine/threonine kinases regulates cell survival pathways and has been implicated in the progression of several human cancers, including prostate cancer (10). Clinically, the expression of the Pim kinases is elevated in human prostate cancer (10), in which the PI3K/AKT pathway is activated, and the levels of Pim correlate with survival of patients with certain subtypes of human lymphoma (11), suggesting that the Pim kinases could play an important role in regulating tumor growth and, potentially, patient survival. As the Pim kinases have overlapping activity with AKT with both regulating apoptosis, cell-cycle progression, and cellular metabolism (12–13), and AKT and the Pim kinases share substrates in common (12–13), it has been suggested that Pim could play an important role in the activation of AKT (14). Reciprocal regulation of AKT and Pim-1 levels is suggested by the report that forced expression of nuclear-targeted AKT induces Pim-1 and either expression of a dominant-negative Pim-1 or genetic deletion of the enzyme increased AKT expression and phospho-AKT levels in cardiomyocytes (14).

Here, we show that inhibition of AKT leads to transcriptional induction of the Pim-1 protein kinase, and in turn, Pim-1 regulates the expression of RTKs. The anticancer activity of small-molecule AKT and Pim kinase inhibitors has been investigated.

Materials and Methods

Reagents and antibodies

GSK690693 was provided by GlaxoSmithKline for in vitro and in vivo studies. MK2206, PP242, AZD8055, and BEZ235 were purchased from Selleck Biochemicals. Antibodies are listed in the Supplementary Data.

Plasmids

The 5′-untranslated region (UTR) of human Met (15) was amplified by PCR using genomic DNA extracted from PC3-LN4 cells as template with the following 2 primers: 5′-ATACTAGTGCTGCAGCGGCCGCGGTGGCTGA-3′ and 5′-AACCATGGCCCAACCTCCAGGATGTCGGCGCA-3′. The PCR product was sequenced and cloned into the EcoRI and NcoI sites of the plasmid of pRF to create pR-MET-F.

Immunoblotting

Cells were harvested in lysis buffer A consisting of 50 mmol/L Tris, pH 7.4, 150 mmol/L NaCl, 1% NP-40, and 5 mmol/L EDTA. Protein concentrations were determined by DC Protein Assay (Bio-Rad).

Cell culture and transfections

Cell lines were grown in RPMI (PC3-LN4, DU145, 22RV1, VCAP, and BT474) or Dulbecco's Modified Eagle Medium [HeLa, mouse embryo fibroblasts (MEF)] in 5% CO2. DU145, 22RV1, VCAP, BT474, and HeLa cells were supplied by American Type Culture Collection and passaged in our laboratory for less than 6 months after receipt. PC3-LN4 cells were described before (16). The MEFs, which were triple knockout (TKO) for all Pim genes, were previously described (17). Cells were transfected with Lipofectamine 2000 reagent according to manufacturer's instructions.

Real-time PCR analyses

SYBR Green reactions were done using a BioRad iQ5 quantitative real-time PCR (qRT-PCR) system. For data analysis, raw counts were normalized to the housekeeping gene averaged for the same timepoint and condition (ΔCt). Counts are reported as fold change relative to the untreated control (2−ΔΔCt). All primers were designed and synthesized by Integrated DNA Technologies. Primers are listed in the Supplementary Data.

Luciferase assays

Firefly luciferase and Renilla luciferase activities were measured in a luminometer (Model TD 20/20; Turner Designs) using the reagents provided with the Dual Luciferase Reporter kit (Promega).

Soft-agar colony formation assays

The soft-agar assay was conducted on 6-well plates in duplicate. For each well, 5,000 cells were mixed in growth medium containing 0.7% agarose and GSK690693 or SMI-4a. Cells were then layered over 1% agarose in regular medium. Medium containing GSK690693 or SMI-4a was added to each well every 4 days. The assays were terminated after 21 days, and colonies were stained with crystal violet and counted under a microscope.

Cell proliferation measurement

Cells were plated in 96-well plates at 3,000 cells per well in 100 μL of 10% FBS-containing medium. After 24-hour incubation, the medium was replaced with 0.2% FBS medium with GSK690693, SMI-4a, or dimethyl sulfoxide (DMSO) for 72 hours. Cell viability was measured using a MTT assay. The absorbance was read at 590 nm with a reference filter of 620 nm.

In vitro transcription and RNA transfection

The mRNAs were purified with MEGA Clear Kit (Ambion), quantified spectrophotometrically, and their qualities were verified on a denaturing agarose gel. RNA transfection was conducted with TransIT-mRNA Transfection Kit (Mirus) according to the manufacturer's suggestion. An aliquot of 1 μg of capped mRNAs and 2 μL of TransIT-mRNA reagent together with 1 μL of mRNA boost reagent was used to transfect 80% confluent cells grown in 12-well plates. At 16 hours after transfection, cells were harvested and lysed for luciferase assay.

Animal experiments

Four- to six-week-old nu/nu nude male mice were obtained from Charles River Laboratories and maintained in pressurized ventilated caging. All studies were conducted in compliance with Institutional guidelines under an Institutional Animal Care and Use Committee-approved protocol (MUSC#3081). For efficacy studies, mice with well-established tumors were selected and randomized 14 days after implantation (size > 150 mm3); PC3-LN4 xenograft tumors were established in nude mice by subcutaneously injecting 5 × 106 cells suspended in PBS into the right flank. Mice were treated with vehicle, GSK690693, or SMI-4a, or GSK690693 + SMI-4a at the indicated doses. GSK690693 was dissolved in 30% propylene glycol, 5% Tween-80, 65% of 5% dextrose in water (pH4–5), and administered intraperitoneally daily, whereas SMI-4a was dissolved in the same solvent and administered by oral gavage twice daily. Tumor dimensions were measured with a caliper and tumor volumes were calculated [tumor volume (mm3) = (length × width2)/2].

Statistical analysis

The results of quantitative studies are reported as mean ± SD or mean ± SEM (for animal experiments). Differences were analyzed by Student t test. P < 0.05 was regarded as significant.

Results

AKT inhibition induces Pim-1 expression in prostate cancer cells

Treatment of the prostate cancer PC3-LN4 cells with the pan-AKT inhibitor GSK690693 markedly increased the levels of Pim-1 protein in a time- and concentration-dependent fashion (Fig. 1A and B) but had a minimal effect on the expression of Pim-3 protein and reduced the levels of Pim-2 (Fig. 1C). Similar results were obtained using another AKT inhibitor, MK2206, and a PI3K/mTOR dual inhibitor, BEZ235 (Fig. 1C). The induction of Pim-1 was also observed with GSK690693 treatment of human prostate cancer cell lines DU145, 22RV1, and VCAP (Supplementary Fig. S1A). The effect of GSK690693 on Pim-1 was not secondary to an off-target effect as knockdown in PC3-LN4 cells of all 3 AKTs with siRNAs increased the levels of Pim-1 protein (Fig. 1D). Treatment of PC3-LN4 cells with GSK690693 or MK2206 resulted in elevations in the level of Pim-1 mRNA, but not Pim-2 or Pim-3 (Fig. 1E). Similarly, treatment of PC3-LN4 cells with siRNAs directed at AKT1, AKT2, and AKT3 also resulted in the elevation of Pim-1 mRNA (Fig. 1F). To further determine whether GSK690693 regulates the transcription of the Pim-1 gene, a 3.0 kb promoter fragment of the Pim-1 promoter was cloned upstream of a luciferase reporter. Addition of GSK690693 increased the activity of this promoter in PC3-LN4 cells (Fig. 1G).

Figure 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 1.

AKT inhibition induces expression of Pim-1. PC3-LN4 cells were treated with 5 μmol/L GSK690693 (A) for the times indicated, increasing doses of GSK690693 (B) as indicated for 24 hours, 5 μmol/L GSK690693, 2 μmol/L MK2206, or 0.5 μmol/L BEZ235 (C) for 24 hours, and siRNAs against AKT1, AKT2, and AKT3 or a negative control siRNA (D) for 72 hours. Whole-cell lysates were subjected to immunoblot analyses with the indicated antibodies. E, cells as in C were harvested and total RNA was isolated. Real-time qPCR analyses were conducted with Pim-1, Pim-2, Pim-3-specific primers. Results were normalized to the expression of β-actin. F, PC3-LN4 cells were treated with siRNAs against AKT1, 2 (siA1,2), AKT3 (siA3), AKT1, 2, 3 (siA1,2,3), or a nontargeting control siRNA (siC) for 72 hours and then RNA isolated, and real-time qPCR with indicated primers conducted. G, PC3-LN4 cells were transfected with a luciferase reporter containing a 3.0 kb human Pim-1 promoter. After 24 hours, cells were treated with DMSO or 2 different doses of GSK690693 (GSK) as indicated for additional 24 hours before harvesting for luciferase assays. Results were normalized to Renilla luciferase activity by a cotransfected plasmid carrying this enzyme. Data in E, F, and G are mean ± SD of 3 independent experiments. *, P < 0.05 compared with the corresponding negative control. GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

Upregulation of Pim-1 is required for AKT inhibitor-associated induction of RTKs

Consistent with previous reports of upregulation of these RTKs in response to AKT inhibition (7), treatment of PC3-LN4 cells with GSK690693 increased the protein levels of multiple RTKs, including MET, EPHA2, RON, EGFR, HER2, HER3, INSR, and IGF-IR (Supplementary Fig. S1B). In addition, we observed increased extracellular signal-regulated kinase (ERK) phosphorylation resulting from treatment with GSK690693 (Supplementary Fig. S1B). This is in keeping with previous finding that PI3K inhibition leads to mitogen-activated protein kinase pathway activation (18–19).

GSK690693 also completely blocked the phosphorylation of 2 well-known AKT substrates, GSK3β and PRAS40, showing the effectiveness of this compound (Supplementary Fig. S1C) and caused the paradoxical hyperphosphorylation of AKT at its 2 regulatory sites (Thr308 and Ser473; Supplementary Fig. S1C), a common property of ATP-competitive AKT inhibitors (20).

To determine whether Pim-1 plays an important regulatory role in the ability of AKT inhibitors to modulate RTKs, we first determined the effects of Pim-directed siRNAs and small-molecule inhibitors. The use of siRNA directed at Pim-1 showed that a forced reduction in Pim-1 levels markedly reduced the ability of GSK690693 to elevate the protein levels of multiple RTKs, including MET and EPHA2, HER3, HER2, INSR, and IGF-IR, as well as the phosphorylation of ERK (Fig. 2A). The addition of SMI-4a, a small-molecule Pim kinase inhibitor (21), reduced GSK690693-induced upregulation of RTK protein levels in PC3-LN4 (Fig. 2B), DU145, 22RV1, and VCAP cells (Supplementary Fig. S2)

Figure 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 2.

Pim-1 is required for elevated expression of RTKs induced by AKT inhibition. Immunoblot analyses were carried out with the indicated antibodies in PC3-LN4 cells that were treated with 2 different siRNAs (1 and 2) against Pim-1 as well as a nontargeting control siRNA (2 left lanes) for 48 hours followed by the addition of GSK690693 (5 μmol/L) for an additional 24 hours (A). B, PC3-LN4 cells were treated with GSK690693 (GSK, 5 μmol/L) or SMI-4a (4a, 10 μmol/L) or the combination of the 2 compounds for 24 hours. C–E, wild-type (WT; C), Pim kinase TKO murine embryonic fibroblast cells were treated with 5 μmol/L GSK690693 for 24 hours; PC3-LN4 cells were treated with 3 different Pim inhibitors, SMI-4a (4a, 10 μmol/L), SMI-16a (16a, 10 μmol/L), or K00135 (K, 5 μmol/L) for 24 hours (D); PC3-LN4 cells were transfected with a nontargeting control siRNA, siRNA against Pim-1, an empty vector, or a Pim-1–expressing plasmid for 72 hours (E).

The results of phospho-RTK antibody array (reverse-phase protein array; RPPA) analysis revealed that treatment of PC3-LN4 cells with GSK690693 increased the tyrosine phosphorylation of a number of RTKs tested in the assay, that is, MET, EPHA2, HER2, INSR, and EGFR (Supplementary Fig. S3). The lack of complete correlation in these assays may arise from the differing specificity of the antibodies used in the RPPA analysis. This change in RTK phosphorylation is consistent with the AKT inhibitor-induced increases in the protein levels of the RTKs; however, it cannot be ruled out that GSK690693 stimulates RTK phosphorylation through an alternative mechanism (7). Treatment with SMI-4a blocked the GSK690693-induced RTK phosphorylation (Supplementary Fig. S3), showing that the inhibition of Pim reverses the activity of this AKT inhibitor. To further evaluate the role of Pim-1 in regulating AKT inhibitor-induced upregulation of RTKs, MEFs were treated with GSK690693. In wild-type cells, but not in the Pim kinase-deficient (TKO) cells, GSK690693 treatment of the cells increased the levels of the RTKs tested, that is, MET, HER3, IGF-IR, and EPHA2 protein, as well as the phosphorylation of ERK (Fig. 2C). We treated PC3-LN4 (Fig. 2D) and VCAP (Supplementary Fig. S4) cells with 3 different Pim kinase inhibitors, SMI-4a, SMI-16a (21), and K00135 (22), to test whether Pim-1 activity affects the baseline level of RTK proteins in tumor cells. Treatment decreased the protein levels of the RTKs, for example, MET, EPHA2, and HER3, in both cell lines. Similarly, siRNA targeting of Pim-1 decreased the levels of MET, HER3, HER2, and EGFR protein in PC3-LN4 cells (Fig. 2E). Conversely, overexpression of human Pim-1 in PC3-LN4 increased the levels of the RTKs, MET, HER3, EPHA2, HER2, and EGFR (Fig. 2E).

AKT inhibition increases cap-independent translation

AKT protein kinase activity controls protein synthesis by regulating the multistep process of mRNA translation at multiple stages from ribosome biogenesis to translation initiation and elongation (23). Although GSK690693 treatment of prostate cancer cells did not modify phosphorylation of 4E-BP1, this compound increased phosphorylation of eIF2α and eliminated phosphorylation of ribosomal protein S6 (Fig. 3A). To further define the role of cap-dependent translation in the mechanism of action of this agent, GSK690693 was combined with 2 potent inhibitors of mTOR complex (mTORC)-1/mTORC2 and thus cap-dependent translation, PP242 and AZD8055 (24–25). These inhibitors in combination with GSK690693 resulted in reduced phosphorylation of 4E-BP1 and increased eIF2α phosphorylation compared with GSK690693 alone (Fig. 3A), suggesting inhibition of 5′-cap–dependent translation. We measured the binding of eIF4G and 4E-BP1 to the 5′ mRNA cap by using m7GTP-sepharose. The structure of these beads mimics the 5′ mRNA cap and precipitates cap-interacting proteins. In agreement with the effect on phosphorylation of 4E-BP1, PP242, or AZD8055 in combination with GSK690693 strongly reduced eIF4G and increased 4E-BP1, binding to m7GTP-sepharose, whereas GSK690693 alone did not have a significant effect (Supplementary Fig. S5A). However, the treatment of prostate cancer cells with these mTORC1 inhibitors did not reduce the GSK690693-induced elevation of MET, EPHA2, HER3, and IGF-IR (Fig. 3A). A recent study (26) using Torin 1, an ATP-competitive mTOR inhibitor, showed that Torin 1-resistant mRNAs are enriched for RTKs such as MET, IGF-IR, and INSR, indicating that the translation initiation of these mRNAs do not depend on mTOR activity (27). We found that treatment of PC3-LN4 cells with PP242 or AZD8055 indeed did not inhibit the expression of MET, EPHA2, HER3, IGF1R, or INSR (Supplementary Fig. S5B). In addition, the expression of Bcl-2 whose translation under cellular stress (28) has been shown to be controlled by a cap-independent mechanism was not suppressed by treatment with mTOR inhibitors, whereas proteins known to be sensitive to mTOR inhibiton, YB-1, HSP90, RPS7 (26, 29), were reduced (Supplementary Fig. S5B). Reduced eIF4G and increased 4EBP1 binding to m7GTP-sepharose, and increased eIF2α phosphorylation (Supplementary Fig. S5C) confirmed that cap-dependent translation was efficiently inhibited. Together these data suggest that upregulation of RTKs is not controlled by cap-dependent mechanisms.

Figure 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 3.

AKT inhibition increases cap-independent translation. A, PC3-LN4 cells were treated with GSK690693 (5 μmol/L) alone or in combination with PP242 (2 μmol/L) or AZD8055 (1 μmol/L) for 24 hours and immunoblotting conducted. B, dicistronic luciferase plasmids containing viral (CrPV and HCV) or cellular (HIF1α, VEGF, and Myc) IRESs were transfected into PC3-LN4 cells. GSK690693 (5 μmol/L) was added 6 hours after transfection for additional 24 hours and luciferase activities were determined. Data are mean ± SD of 4 independent experiments. *, P < 0.05 compared with the corresponding DMSO control. C, PC3-LN4 cells were treated with increasing doses of GSK690693 as indicated for 24 hours and lysates examined by Western blotting. GAPDH, glyceraldehyde-3-phosphate dehydrogenase.

Under conditions of decreased cap-dependent translation, the internal ribosome entry site (IRES)-mediated translation can play a larger role in regulating protein synthesis (30). Recently, it has been shown that inhibition of PI3K/mTOR leads to increased IRES-mediated translation (8). Inhibition of AKT by GSK690693 resulted in increased IRES activity measured by ratio of firefly to Renilla luciferase activities in constructs containing either cellular [hypoxia-inducible factor-1α (HIF1α), Myc, and VEGF; ref. 31] or viral [cricket paralysis virus (CrPV) and hepatitis C virus (HCV); ref. 32] IRES sequences (Fig. 3B). In agreement with these findings, GSK690693 induced expression of Bcl-2, Myc, VEGF, and HIF1α, all of which can be translated in a cap-independent manner under cellular stress (28, 31, 33–34), further suggesting the possibility that cap-independent translation is upregulated (Fig. 3C).

Pim-1 regulates RTK expression through cap-independent translation

Expression of human Pim-1 in PC3-LN4 cells did not affect the levels of RTK mRNAs (Supplementary Fig. S6A and B) or the half life of the RTKs (Supplementary Fig. S6C), suggesting that Pim-1 may control the levels of these proteins through a translational mechanism. Plus, GSK690693 increased cap-independent translation (Fig. 3). Taken together, we speculated that the upregulation of the RTKs induced by AKT inhibitors could be controlled, at least in part, by a cap-independent mechanism. We first determined whether the MET 5′-UTR contains an IRES that could be stimulated by either GSK690693 or Pim-1. The MET 5′-UTR is relatively long (408 nt) and is guanine-cytosine (GC)-rich (15), which are 2 common properties of IRES-containing 5′-UTRs. The 5′-UTR of MET was cloned and inserted in front of firefly luciferase in the dicistronic vector pRF (35). The presence of the MET 5′-UTR sequence increased the expression of downstream firefly luciferase relative to Renilla by 38-fold compared with the vector control (Fig. 4A), suggesting that it could function as an IRES. In comparison, the IRESs of encephalomyocarditis virus (EMCV), HIF1α, and VEGF produced 18-, 9-, and 13-fold increases, respectively. In PC3-LN4 cells transfected with the pRF vector containing the MET IRES, overexpression of Pim-1 or treatment of GSK690693 resulted in an increase in ratio of firefly to Renilla luciferase activities as compared with control treatment (Fig. 4A). Knockdown of Pim-1 suppressed GSK690603-induced MET IRES activities (Supplementary Fig. S7). Collectively, these results indicated that Pim-1 can potentially regulate translation of MET in a cap-independent fashion.

Figure 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 4.

Pim-1 regulates RTK translation by controlling IRES activity. A, dicistronic plasmids pRF, pR-EMCV-F, pR-HIF-F, pR-VEGF-F, and pR-MET-F were transfected into PC3-LN4 cells. A Pim-1–expressing plasmid was cotransfected with pR-MET-F as indicated. GSK690693 (GSK, 5 μmol/L) was added 6 hours after transfection and luciferase activities were determined 24 hours after transfection. Data are mean ± SD of 4 independent experiments. *, P < 0.05 compared with the MET. B, capped, polyadenylated dicistronic mRNAs were transfected into PC3-LN4 cells. The ratios of firefly/Renilla activities are shown relative to the ratio for RF, which was given a value of 1. C, a dicistronic plasmid containing IGF-IR IRES was transfected into PC3-LN4 cells with or without either a Pim-1–expressing plasmid or siRNA targeted at Pim-1. GSK690693 (GSK, 5 μmol/L) was added 6 hours after transfection and at 48 hours luciferase activities were determined. D, dicistronic luciferase plasmids containing viral (CrPV and HCV) or cellular (HIF1α and Myc) IRESs were transfected into PC3-LN4 cells together with siRNA against Pim-1 or a nontargeting control siRNA, and luciferase activities were determined 48 hours after transfection. E, PC3-LN4 cells were treated with 2 different siRNAs (1 and 2) against Pim-1 as well as a nontargeting control siRNA (2 left lanes) for 48 hours followed by adding GSK690693 (5 μmol/L) for additional 24 hours as indicated. Whole-cell lysates were subjected to immunoblot analyses with the indicated antibodies. Data in B, C, D, and E reflect the mean ± SD of 4 independent experiments. *, P < 0.05 compared with corresponding negative controls.

To determine whether the MET 5′-UTR is sufficient to drive translation by acting as an IRES and to rule out the possibility of a cryptic promoter in the 5′-UTR of MET, we in vitro transcribed the pRF vector containing the MET IRES yielding a capped dicistronic mRNA, and then transfected this mRNA directly into PC3-LN4 cells. Insertion of the MET or VEGF 5′-UTR resulted in a 7- or 5-fold increase in the firefly/Renilla ratio, respectively. In comparison, when the pRF vector containing the viral EMCV IRES was transcribed and transduced into these cells, the firefly/Renilla ratio increased by 114-fold (Fig. 4B). Thus, in comparison with a viral IRES, both the MET and VEGF sequences have relatively weak IRES activities. Besides MET, other RTKs including IGF-IR have been reported to have IRES elements in their 5′-UTRs (36). As shown in Fig. 4C, the IRES activity of the 5′-UTR of IGF-IR was increased on treatment of the cells with GSK690693 or Pim-1 overexpression and, conversely, was decreased on knockdown of endogenous Pim-1 protein levels. Furthermore, knockdown of Pim-1 suppressed GSK690603-induced IGF-IR IRES activities (Supplementary Fig. S7). It is possible that this mechanism is important for the control of other RTKs because in general these genes have long 5′-UTRs. In addition, knockdown of Pim-1 in PC3-LN4 cells led to a reduction of IRES activities of viral, CrPV and HCV, and cellular, HIF1α and Myc, IRESs (Fig. 4D). These data suggest that Pim-1 could be a more general regulator of IRES-mediated translation. This concept is further supported by our finding that the upregulation of proteins whose translation can be controlled by an IRES-mediated mechanism under cellular stress, Bcl-2, Myc, VEGF, and HIF1α, is stimulated by GSK690693 and requires Pim-1 expression (Fig. 4E).

Ribosomal stress abrogates AKT inhibition-induced upregulation of RTK expression

Pim-1 has been shown to physically interact with ribosomal protein S19 and to cosediment with ribosomes (37–38). Knockdown of ribosomal protein S19 or S6 abolished upregulation of MET, EPHA2, HER3, and IGF-IR induced by GSK690693 without affecting Pim-1 induction (Fig. 5A). Consistent with findings from other laboratories (38–40), reduced protein expression of ribosomal protein S6 was seen when S19 was decreased by siRNA and vice versa (Fig. 5A). To test the effect of ribosomal stress on RTK upregulation independent of ribosomal protein knockdowns, low concentrations of actinomycin D (ActD) were used to inhibit RNA polymerase I, and thus induce ribosomal stress (41–42). Similar to S19 and S6 knockdowns, ActD treatment blocked upregulation of MET, EPHA2, HER3, and IGF-IR induced by GSK690693 (Fig. 5B). ActD treatment also inhibited upregulation of MET, EPHA2, and HER3 resulting from direct Pim-1 overexpression in PC3-LN4 cells (Fig. 5C). Ribosomal stress did not seem to affect global translation as the expression of Src and ERK1/2 proteins was not altered (Fig. 5A and B). These data suggest that Pim-1 may work through intact ribosomes to control RTK expression.

Figure 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 5.

Ribosomal stress abrogates RTK upregulation induced by GSK690693. A, PC3-LN4 cells were treated for 48 hours with siRNAs against Pim-1, ribosomal protein S19, S6 as well as a nontargeting control siRNA (2 left lanes) followed by adding GSK690693 (5 μmol/L) for an additional 24 hours. B, PC3-LN4 cells were treated with increasing dose of ActD with and without 5 μmol/L GSK690693 for 24 hours. C, PC3-LN4 cells were transfected with a Pim-1–expressing plasmid or a control vector. ActD (5 nmol/L) was added 24 hours after transfection for an additional 16 hours. Whole-cell lysates were subjected to immunoblot analyses with the indicated antibodies.

Combination treatment with an AKT and a Pim inhibitor synergistically blocks prostate tumor growth in vitro and in vivo

As a preliminary test of whether combined inhibition of AKT and Pim kinases might provide synergistic antitumor efficacy, we tested the effects of the inhibitors on the proliferation of PC3-LN4 cells in vitro. Treatment of PC3-LN4 cells with the Pim inhibitor SMI-4a in combination with the AKT inhibitor GSK690693 resulted in a synergistic enhancement of the inhibition of proliferation as shown by combination index of less than 0.5 (Fig. 6A; data not shown), and a markedly greater reduction in both the numbers and the size of colonies seen in a soft-agar colony formation assay (Fig. 6B). GSK690693 and SMI-4a blocked the proliferation of DU145 in a similar fashion (Supplementary Fig. S8).

Figure 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Figure 6.

Combined inhibition of AKT and Pim shows synergistic antitumor activity. A, PC3-LN4 cells were treated with increasing doses of GSK690693 and SMI-4a (4a) as indicated in media containing 0.2% serum for 72 hours followed by a MTT assay. Similar results were obtained from 3 independent experiments. One representative experiment is shown. B, PC3-LN4 cells were plated in 10% serum and 0.7% agarose-containing medium with 10 μmol/L of GSK690693 or SMI-4a alone or in combination. Colonies were stained with crystal violet and counted after 21 days and the data are mean ± SD of 3 independent experiments. Bar, 200 micrometer. C, Nu/Nu mice bearing PC3-LN4 tumors were randomized into 4 groups: vehicle, GSK690693 (30 mg/kg i.p. daily), SMI-4a (60 mg/kg oral twice/day), and the combination. Tumor size was measured every 3 days. The results are presented as the mean tumor volume ± SEM (n = 6 mice/group). **, P < 0.02 for the GSK + SMI-4a group versus all other treatment groups. D, immunoblot analyses of tumors in C with the indicated antibodies. Tumors were harvested on day 36, 6 hours after the last dose of therapy. The numbers above each lane represent individual tumors in that treatment group. E, a model for the feedback upregulation of RTK expression mediated by Pim-1 kinase.

To test the activity of these agents in vivo, PC3-LN4 cells were injected into mice and treated with GSK690693 alone, SMI-4a alone, or both drugs in combination on a daily basis for 21 days starting at 15 days after tumor implantation. When used alone, treatment of these drugs caused a modest inhibition of tumor growth, whereas the combined treatment resulted in a markedly greater inhibition of tumor growth (Fig. 6C). As shown in Fig. 6D, immunoblot analysis of lysates of tumors harvested at the termination of the experiment on day 36 had upregulated the levels of MET, EPHA2, and HER3 protein in mice treated with GSK690693 as compared with the tumors from mice treated with vehicle (Fig. 6D). Interestingly, the levels of Pim-1 were increased in the combined therapy, and could suggest an in vivo interaction between these agents cannot be ruled out. This upregulation of the RTKs was significantly reduced in the tumors from mice treated with a combination of GSK690693 and SMI-4a (Fig. 6D).

Discussion

The results of these experiments provide insights into the mechanisms underlying the compensatory interplay between AKT and Pim-1 in the regulation of prostate cancer cell behavior influenced by the expression of RTKs. They suggest a model in which reduction in AKT activity is associated with an increase in the levels of Pim-1 protein kinase that occurs through a transcriptional mechanism. This increase in Pim-1 kinase is associated, in turn, with promotion of the expression of RTKs through a cap-independent mechanism. Downregulation of Pim-1 blocks the feedback elevation in RTKs associated with inhibition of AKT (Fig. 6E). Likewise inhibitors of Pim synergize with small-molecule AKT inhibitors to block the growth of prostate cancer cells.

The control of Pim-1 protein levels is complex and has been shown to involve the ubiquitin proteasome pathway and translational mechanisms (43). In the current study, we show that inhibition of AKT can increase the levels of Pim-1 through a transcriptional mechanism; however, it is possible that additional alternative mechanisms could also play a role in increasing Pim-1 protein levels. The induction of Pim-1 by AKT inhibition coincides with suppression of total protein synthesis (Supplementary Fig. S9) and is not inhibited by further treatment with mTORC inhibitors (Fig. 3A), suggesting that Pim-1 protein levels could also be regulated in a cap-independent manner. The Pim-1 5′-UTR may contain an IRES that could also be regulated by specific cellular growth conditions (44), although the existence of this IRES is controversial (45).

It has been shown previously that inhibition of AKT regulates the transcription of RTKs by modulating the activity of Foxo transcription factors (7); however, in the same study no change was seen in the level of HER2, RET, or MET mRNAs, suggesting that the levels of specific RTKs might be controlled by other mechanisms. Cap-dependent translation plays a role in both PI3K/AKT and Pim-2 enhancement of the synthesis of specific proteins (46). It should be noted, however, that molecules that blocked mTORC1 activity could not inhibit the Pim-2 protein kinase and an agent that blocked eIF4A function, which is known to take part in IRES mediated translation, was required. In addition, small-molecule mTORC inhibitors can decrease the translation of many mRNAs, for example, 5′-terminal oligopyrimidine tracts mRNAs, while increasing the level of translation of RTKs (26), again suggesting that these RTK mRNAs may be translated in a cap-independent fashion. Moreover, further inhibition of cap-dependent translation with the mTORC1/2 inhibitors, PP242 and AZD8055, had no effect on the ability of GSK690693 or Pim-1 to induce RTKs (Fig. 3A), suggesting that in the experimental conditions used in these studies, the mechanism by which this agent controls RTK levels is not cap dependent.

Our results are consistent with the hypothesis put forward by Muranen and colleagues that inhibition of PI3K/mTOR could lead to enhanced cap-independent translation (8). Cloning of the Met 5′-UTR into a dicistronic luciferase vector showed that it can function as an IRES element, although weakly in comparison with viral sequences, and its activity is enhanced by GSK690693 and Pim-1 overexpression (Fig. 4A and B). Further supporting evidence of the ability of GSK690693 and Pim-1 to regulate the activity of the IRES is the observation that the IGF-IR IRES (47) is stimulated by these agents and that Pim-1 knockdown decreases the activity of this element (Fig. 4C). Our data further suggest that Pim-1 may be essential for full IRES activity of additional viral and cellular IRES elements, including HCV, CrPV, HIF1α, and Myc (Fig. 4D), suggesting a general role of Pim in the control of cap-independent translation.

It has been suggested previously that because they are both survival kinases, AKT and Pim protein kinases could be important pharmacologic targets to inhibit tumor growth (12). Our experiments show a high degree of synergism between small-molecule inhibitors of AKT and Pim in their ability to kill prostate cancer cells both in tissue culture and in a xenograft model (Fig. 6). Analysis of the tumors from treated animals showed that AKT inhibitor treatment elevates RTKs in the tumor cells grown in vivo and that simultaneous treatment with a Pim inhibitor downregulates this effect (Fig. 6D). Because both these kinase pathways are highly activated in human prostate cancer, dual inhibitor treatment of these tumors could be a particularly attractive chemotherapeutic strategy.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed by the authors.

Authors' Contributions

Conception and design: B. Cen, A.S. Kraft

Development of methodology: B. Cen, W. Wang

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): B. Cen, W. Wang, A.S. Kraft

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): B. Cen, S. Mahajan, A.S. Kraft

Writing, review, and/or revision of the manuscript: B. Cen, A.S. Kraft

Study supervision: B. Cen, A.S. Kraft

Grant Support

This work is supported by NIH Grant 1K01DK085196 (B. Cen), DOD W81XWH-09-1-0300 (A.S. Kraft), DOD W81XWH-10-1-0249 (A.S. Kraft), NIH/NCRR Grant UL1RR029882, and in part by pilot research funding, Hollings Cancer Center's Cancer Center Support Grant P30 CA138313 at the Medical University of South Carolina.

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

Acknowledgments

The authors thank members of the Kraft laboratory, including Dr. Zanna Beharry for critical reading of the manuscript, Dr. Jin Song for helping to carry out experiments, Dr. Marina Zemskova for providing the luciferase construct containing the human Pim-1 promoter, and GlaxoSmithKline for supplying GSK690693. Dicistronic plasmids were kindly provided by Drs. Scott Blume, University of Alabama at Birmingham (IGF-IR; Birmingham, AL); Gregory Goodall, Institute of Medical and Veterinary Science, (HIF, c-Myc, and VEGF; Adelaide, Australia); Gregg Johannes, Drexel University (EMCV; Philadelphia, PA); and Robert Gemmill, Medical University of South Carolina (CrPV and HCV; Charleston, SC).

Footnotes

  • Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/).

  • Received December 18, 2012.
  • Revision received March 1, 2013.
  • Accepted March 26, 2013.
  • ©2013 American Association for Cancer Research.

References

  1. 1.↵
    1. Vivanco I,
    2. Sawyers CL
    . The phosphatidylinositol 3-Kinase AKT pathway in human cancer. Nat Rev Cancer 2002;2:489–501.
    OpenUrlCrossRefPubMed
  2. 2.↵
    1. Yoshimoto M,
    2. Cunha IW,
    3. Coudry RA,
    4. Fonseca FP,
    5. Torres CH,
    6. Soares FA,
    7. et al.
    FISH analysis of 107 prostate cancers shows that PTEN genomic deletion is associated with poor clinical outcome. Br J Cancer 2007;97:678–85.
    OpenUrlCrossRefPubMed
  3. 3.↵
    1. Suzuki H,
    2. Freije D,
    3. Nusskern DR,
    4. Okami K,
    5. Cairns P,
    6. Sidransky D,
    7. et al.
    Interfocal heterogeneity of PTEN/MMAC1 gene alterations in multiple metastatic prostate cancer tissues. Cancer Res 1998;58:204–9.
    OpenUrlAbstract/FREE Full Text
  4. 4.↵
    1. Bellacosa A,
    2. Kumar CC,
    3. Di Cristofano A,
    4. Testa JR
    . Activation of AKT kinases in cancer: implications for therapeutic targeting. Adv Cancer Res 2005;94:29–86.
    OpenUrlCrossRefPubMed
  5. 5.↵
    1. Hynes NE,
    2. Lane HA
    . ERBB receptors and cancer: the complexity of targeted inhibitors. Nat Rev Cancer 2005;5:341–54.
    OpenUrlCrossRefPubMed
  6. 6.↵
    1. LoPiccolo J,
    2. Blumenthal GM,
    3. Bernstein WB,
    4. Dennis PA
    . Targeting the PI3K/Akt/mTOR pathway: effective combinations and clinical considerations. Drug Resist Updat 2008;11:32–50.
    OpenUrlCrossRefPubMed
  7. 7.↵
    1. Chandarlapaty S,
    2. Sawai A,
    3. Scaltriti M,
    4. Rodrik-Outmezguine V,
    5. Grbovic-Huezo O,
    6. Serra V,
    7. et al.
    AKT inhibition relieves feedback suppression of receptor tyrosine kinase expression and activity. Cancer Cell 2011;19:58–71.
    OpenUrlCrossRefPubMed
  8. 8.↵
    1. Muranen T,
    2. Selfors LM,
    3. Worster DT,
    4. Iwanicki MP,
    5. Song L,
    6. Morales FC,
    7. et al.
    Inhibition of PI3K/mTOR leads to adaptive resistance in matrix-attached cancer cells. Cancer Cell 2012;21:227–39.
    OpenUrlCrossRefPubMed
  9. 9.↵
    1. Chakrabarty A,
    2. Sanchez V,
    3. Kuba MG,
    4. Rinehart C,
    5. Arteaga CL
    . Feedback upregulation of HER3 (ErbB3) expression and activity attenuates antitumor effect of PI3K inhibitors. Proc Natl Acad Sci U S A 2012;109:2718–23.
    OpenUrlAbstract/FREE Full Text
  10. 10.↵
    1. Dhanasekaran SM,
    2. Barrette TR,
    3. Ghosh D,
    4. Shah R,
    5. Varambally S,
    6. Kurachi K,
    7. et al.
    Delineation of prognostic biomarkers in prostate cancer. Nature 2001;412:822–6.
    OpenUrlCrossRefPubMed
  11. 11.↵
    1. Hsi ED,
    2. Jung SH,
    3. Lai R,
    4. Johnson JL,
    5. Cook JR,
    6. Jones D,
    7. et al.
    Ki67 and PIM1 expression predict outcome in mantle cell lymphoma treated with high dose therapy, stem cell transplantation and rituximab: a Cancer and Leukemia Group B 59909 correlative science study. Leuk Lymphoma 2008;49:2081–90.
    OpenUrlCrossRefPubMed
  12. 12.↵
    1. Amaravadi R,
    2. Thompson CB
    . The survival kinases Akt and Pim as potential pharmacological targets. J Clin Invest 2005;115:2618–24.
    OpenUrlCrossRefPubMed
  13. 13.↵
    1. Sussman MA
    . Mitochondrial integrity: preservation through Akt/Pim-1 kinase signaling in the cardiomyocyte. Expert Rev Cardiovasc Ther 2009;7:929–38.
    OpenUrlCrossRefPubMed
  14. 14.↵
    1. Muraski JA,
    2. Rota M,
    3. Misao Y,
    4. Fransioli J,
    5. Cottage C,
    6. Gude N,
    7. et al.
    Pim-1 regulates cardiomyocyte survival downstream of Akt. Nat Med 2007;13:1467–75.
    OpenUrlCrossRefPubMed
  15. 15.↵
    1. Gambarotta G,
    2. Pistoi S,
    3. Giordano S,
    4. Comoglio PM,
    5. Santoro C
    . Structure and inducible regulation of the human MET promoter. J Biol Chem 1994;269:12852–7.
    OpenUrlAbstract/FREE Full Text
  16. 16.↵
    1. Pettaway CA,
    2. Pathak S,
    3. Greene G,
    4. Ramirez E,
    5. Wilson MR,
    6. Killion JJ,
    7. et al.
    Selection of highly metastatic variants of different human prostatic carcinomas using orthotopic implantation in nude mice. Clin Cancer Res 1996;2:1627–36.
    OpenUrlAbstract
  17. 17.↵
    1. Lin YW,
    2. Beharry ZM,
    3. Hill EG,
    4. Song JH,
    5. Wang W,
    6. Xia Z,
    7. et al.
    A small molecule inhibitor of Pim protein kinases blocks the growth of precursor T-cell lymphoblastic leukemia/lymphoma. Blood 2010;115:824–33.
    OpenUrlAbstract/FREE Full Text
  18. 18.↵
    1. Serra V,
    2. Scaltriti M,
    3. Prudkin L,
    4. Eichhorn PJ,
    5. Ibrahim YH,
    6. Chandarlapaty S,
    7. et al.
    PI3K inhibition results in enhanced HER signaling and acquired ERK dependency in HER2-overexpressing breast cancer. Oncogene 2011;30:2547–57.
    OpenUrlCrossRefPubMed
  19. 19.↵
    1. Engelman JA,
    2. Chen L,
    3. Tan X,
    4. Crosby K,
    5. Guimaraes AR,
    6. Upadhyay R,
    7. et al.
    Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat Med 2008;14:1351–6.
    OpenUrlCrossRefPubMed
  20. 20.↵
    1. Okuzumi T,
    2. Fiedler D,
    3. Zhang C,
    4. Gray DC,
    5. Aizenstein B,
    6. Hoffman R,
    7. et al.
    Inhibitor hijacking of Akt activation. Nat Chem Biol 2009;5:484–93.
    OpenUrlCrossRefPubMed
  21. 21.↵
    1. Xia Z,
    2. Knaak C,
    3. Ma J,
    4. Beharry ZM,
    5. McInnes C,
    6. Wang W,
    7. et al.
    Synthesis and evaluation of novel inhibitors of Pim-1 and Pim-2 protein kinases. J Med Chem 2009;52:74–86.
    OpenUrlCrossRefPubMed
  22. 22.↵
    1. Pogacic V,
    2. Bullock AN,
    3. Fedorov O,
    4. Filippakopoulos P,
    5. Gasser C,
    6. Biondi A,
    7. et al.
    Structural analysis identifies imidazo[1,2-b]pyridazines as PIM kinase inhibitors with in vitro antileukemic activity. Cancer Res 2007;67:6916–24.
    OpenUrlAbstract/FREE Full Text
  23. 23.↵
    1. Hsieh AC,
    2. Truitt ML,
    3. Ruggero D
    . Oncogenic AKTivation of translation as a therapeutic target. Br J Cancer 2011;105:329–36.
    OpenUrlCrossRefPubMed
  24. 24.↵
    1. Feldman ME,
    2. Apsel B,
    3. Uotila A,
    4. Loewith R,
    5. Knight ZA,
    6. Ruggero D,
    7. et al.
    Active-site inhibitors of mTOR target rapamycin-resistant outputs of mTORC1 and mTORC2. PLoS Biol 2009;7:e38.
    OpenUrlCrossRefPubMed
  25. 25.↵
    1. Chresta CM,
    2. Davies BR,
    3. Hickson I,
    4. Harding T,
    5. Cosulich S,
    6. Critchlow SE,
    7. et al.
    AZD8055 is a potent, selective, and orally bioavailable ATP-competitive mammalian target of rapamycin kinase inhibitor with in vitro and in vivo antitumor activity. Cancer Res 2010;70:288–98.
    OpenUrlAbstract/FREE Full Text
  26. 26.↵
    1. Thoreen CC,
    2. Chantranupong L,
    3. Keys HR,
    4. Wang T,
    5. Gray NS,
    6. Sabatini DM
    . A unifying model for mTORC1-mediated regulation of mRNA translation. Nature 2012;485:109–13.
    OpenUrlCrossRefPubMed
  27. 27.↵
    1. Martin F,
    2. Barends S,
    3. Jaeger S,
    4. Schaeffer L,
    5. Prongidi-Fix L,
    6. Eriani G
    . Cap-assisted internal initiation of translation of histone H4. Mol Cell 2011;41:197–209.
    OpenUrlCrossRefPubMed
  28. 28.↵
    1. Sherrill KW,
    2. Byrd MP,
    3. Van Eden ME,
    4. Lloyd RE
    . BCL-2 translation is mediated via internal ribosome entry during cell stress. J Biol Chem 2004;279:29066–74.
    OpenUrlAbstract/FREE Full Text
  29. 29.↵
    1. Hsieh AC,
    2. Liu Y,
    3. Edlind MP,
    4. Ingolia NT,
    5. Janes MR,
    6. Sher A,
    7. et al.
    The translational landscape of mTOR signalling steers cancer initiation and metastasis. Nature 2012;485:55–61.
    OpenUrlCrossRefPubMed
  30. 30.↵
    1. Komar AA,
    2. Hatzoglou M
    . Cellular IRES-mediated translation: the war of ITAFs in pathophysiological states. Cell Cycle 2011;10:229–40.
    OpenUrlCrossRefPubMed
  31. 31.↵
    1. Lang KJ,
    2. Kappel A,
    3. Goodall GJ
    . Hypoxia-inducible factor-1alpha mRNA contains an internal ribosome entry site that allows efficient translation during normoxia and hypoxia. Mol Biol Cell 2002;13:1792–801.
    OpenUrlAbstract/FREE Full Text
  32. 32.↵
    1. Landry DM,
    2. Hertz MI,
    3. Thompson SR
    . RPS25 is essential for translation initiation by the Dicistroviridae and hepatitis C viral IRESs. Genes Dev 2009;23:2753–64.
    OpenUrlAbstract/FREE Full Text
  33. 33.↵
    1. Stein I,
    2. Itin A,
    3. Einat P,
    4. Skaliter R,
    5. Grossman Z,
    6. Keshet E
    . Translation of vascular endothelial growth factor mRNA by internal ribosome entry: implications for translation under hypoxia. Mol Cell Biol 1998;18:3112–9.
    OpenUrlAbstract/FREE Full Text
  34. 34.↵
    1. Stoneley M,
    2. Chappell SA,
    3. Jopling CL,
    4. Dickens M,
    5. MacFarlane M,
    6. Willis AE
    . c-Myc protein synthesis is initiated from the internal ribosome entry segment during apoptosis. Mol Cell Biol 2000;20:1162–9.
    OpenUrlAbstract/FREE Full Text
  35. 35.↵
    1. Stoneley M,
    2. Paulin FE,
    3. Le Quesne JP,
    4. Chappell SA,
    5. Willis AE
    . C-Myc 5′ untranslated region contains an internal ribosome entry segment. Oncogene 1998;16:423–8.
    OpenUrlCrossRefPubMed
  36. 36.↵
    1. Giraud S,
    2. Greco A,
    3. Brink M,
    4. Diaz JJ,
    5. Delafontaine P
    . Translation initiation of the insulin-like growth factor I receptor mRNA is mediated by an internal ribosome entry site. J Biol Chem 2001;276:5668–75.
    OpenUrlAbstract/FREE Full Text
  37. 37.↵
    1. Chiocchetti A,
    2. Gibello L,
    3. Carando A,
    4. Aspesi A,
    5. Secco P,
    6. Garelli E,
    7. et al.
    Interactions between RPS19, mutated in Diamond-Blackfan anemia, and the PIM-1 oncoprotein. Haematologica 2005;90:1453–62.
    OpenUrlAbstract/FREE Full Text
  38. 38.↵
    1. Iadevaia V,
    2. Caldarola S,
    3. Biondini L,
    4. Gismondi A,
    5. Karlsson S,
    6. Dianzani I,
    7. et al.
    PIM1 kinase is destabilized by ribosomal stress causing inhibition of cell cycle progression. Oncogene 2010;29:5490–9.
    OpenUrlCrossRefPubMed
  39. 39.↵
    1. Robledo S,
    2. Idol RA,
    3. Crimmins DL,
    4. Ladenson JH,
    5. Mason PJ,
    6. Bessler M
    . The role of human ribosomal proteins in the maturation of rRNA and ribosome production. RNA 2008;14:1918–29.
    OpenUrlAbstract/FREE Full Text
  40. 40.↵
    1. Badhai J,
    2. Frojmark AS,
    3. Razzaghian HR,
    4. Davey E,
    5. Schuster J,
    6. Dahl N
    . Posttranscriptional down-regulation of small ribosomal subunit proteins correlates with reduction of 18S rRNA in RPS19 deficiency. FEBS Lett 2009;583:2049–53.
    OpenUrlCrossRefPubMed
  41. 41.↵
    1. Lohrum MA,
    2. Ludwig RL,
    3. Kubbutat MH,
    4. Hanlon M,
    5. Vousden KH
    . Regulation of HDM2 activity by the ribosomal protein L11. Cancer Cell 2003;3:577–87.
    OpenUrlCrossRefPubMed
  42. 42.↵
    1. Zhang Y,
    2. Wolf GW,
    3. Bhat K,
    4. Jin A,
    5. Allio T,
    6. Burkhart WA,
    7. et al.
    Ribosomal protein L11 negatively regulates oncoprotein MDM2 and mediates a p53-dependent ribosomal-stress checkpoint pathway. Mol Cell Biol 2003;23:8902–12.
    OpenUrlAbstract/FREE Full Text
  43. 43.↵
    1. Nawijn MC,
    2. Alendar A,
    3. Berns A
    . For better or for worse: the role of Pim oncogenes in tumorigenesis. Nat Rev Cancer 2011;11:23–34.
    OpenUrlCrossRefPubMed
  44. 44.↵
    1. Johannes G,
    2. Carter MS,
    3. Eisen MB,
    4. Brown PO,
    5. Sarnow P
    . Identification of eukaryotic mRNAs that are translated at reduced cap binding complex eIF4F concentrations using a cDNA microarray. Proc Natl Acad Sci U S A 1999;96:13118–23.
    OpenUrlAbstract/FREE Full Text
  45. 45.↵
    1. Wang Z,
    2. Weaver M,
    3. Magnuson NS
    . Cryptic promoter activity in the DNA sequence corresponding to the pim-1 5′-UTR. Nucleic Acids Res 2005;33:2248–58.
    OpenUrlAbstract/FREE Full Text
  46. 46.↵
    1. Schatz JH,
    2. Oricchio E,
    3. Wolfe AL,
    4. Jiang M,
    5. Linkov I,
    6. Maragulia J,
    7. et al.
    Targeting cap-dependent translation blocks converging survival signals by AKT and PIM kinases in lymphoma. J Exp Med 2011;208:1799–807.
    OpenUrlAbstract/FREE Full Text
  47. 47.↵
    1. Meng Z,
    2. King PH,
    3. Nabors LB,
    4. Jackson NL,
    5. Chen CY,
    6. Emanuel PD,
    7. et al.
    The ELAV RNA-stability factor HuR binds the 5′-untranslated region of the human IGF-IR transcript and differentially represses cap-dependent and IRES-mediated translation. Nucleic Acids Res 2005;33:2962–79.
    OpenUrlAbstract/FREE Full Text
PreviousNext
Back to top
Cancer Research: 73 (11)
June 2013
Volume 73, Issue 11
  • Table of Contents
  • Table of Contents (PDF)
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Elevation of Receptor Tyrosine Kinases by Small Molecule AKT Inhibitors in Prostate Cancer Is Mediated by Pim-1
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Elevation of Receptor Tyrosine Kinases by Small Molecule AKT Inhibitors in Prostate Cancer Is Mediated by Pim-1
Bo Cen, Sandeep Mahajan, Wenxue Wang and Andrew S. Kraft
Cancer Res June 1 2013 (73) (11) 3402-3411; DOI: 10.1158/0008-5472.CAN-12-4619

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Elevation of Receptor Tyrosine Kinases by Small Molecule AKT Inhibitors in Prostate Cancer Is Mediated by Pim-1
Bo Cen, Sandeep Mahajan, Wenxue Wang and Andrew S. Kraft
Cancer Res June 1 2013 (73) (11) 3402-3411; DOI: 10.1158/0008-5472.CAN-12-4619
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • Introduction
    • Materials and Methods
    • Results
    • Discussion
    • Disclosure of Potential Conflicts of Interest
    • Authors' Contributions
    • Grant Support
    • Acknowledgments
    • Footnotes
    • References
  • Figures & Data
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • H3B-6527 for FGF19-Driven HCC
  • N-myristoyltransferase 1 Promotes Src-Mediated Cancer
  • ATR in Synovial Sarcoma
Show more Therapeutics, Targets, and Chemical Biology
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement