Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2010 Mar 15.
Published in final edited form as: Cancer Res. 2009 Mar 3;69(6):2210–2218. doi: 10.1158/0008-5472.CAN-08-2308

Heterogeneous Nuclear Ribonucleoprotein K is a Novel Regulator of Androgen Receptor Translation

Nishit K Mukhopadhyay 1, Jayoung Kim 1, Bekir Cinar 1, Aruna Ramachandran 1, Martin H Hager 1, Dolores Di Vizio 1, Rosalyn M Adam 1, Mark A Rubin 2, Pradip Raychaudhuri 3, Arrigo De Benedetti 4, Michael R Freeman 1,5
PMCID: PMC2659763  NIHMSID: NIHMS89692  PMID: 19258514

Abstract

Regulation of androgen receptor (AR) expression in prostate cancer (PCa) is still poorly understood. Activation of the epidermal growth factor receptor (EGFR) in PCa cells was previously shown to lower AR expression by a rapamycin-sensitive, post-transcriptional mechanism involving the AR mRNA 5′-untranslated region (5′-UTR). In a search for an intermediate within the EGFR/PI3-kinase/Akt/mTOR pathway that regulates AR at this site, we identified the nucleic acid binding protein, heterogeneous nuclear ribonucleoprotein K (hnRNP-K), by mass spectrometric analysis of Akt immune complexes from lipid raft-enriched subcellular fractions. We show here that hnRNP-K is a novel inhibitor of AR mRNA translation that regulates androgen-responsive gene expression and PCa cell proliferation. A functional hnRNP-K binding site involved in down-regulating AR protein levels was identified in the AR mRNA 5′-UTR. Further analysis revealed that hnRNP-K is also able to inhibit AR translation in the absence of the 5′-UTR, consistent with the presence of additional predicted hnRNP-K binding sites within the AR open reading frame and in the 3′-UTR. Immunohistochemical analysis of a human PCa tissue microarray revealed an inverse correlation between hnRNP-K expression and AR protein levels in organ-confined PCa tumors and a substantial decline in cytoplasmic hnRNP-K in metastases, despite an overall increase in hnRNP-K levels in metastatic tumors. These data suggest that translational inhibition of AR by hnRNP-K may occur in organ-confined tumors but possibly at a reduced level in metastases. HnRNP-K is the first protein identified that directly interacts with and regulates the AR translational apparatus.

Introduction

Prostate cancer (PCa) is the most commonly diagnosed malignancy among men in the United States (1). Androgens, the male sex steroids, mediate growth and development of reproductive tissues and are also prominently involved in PCa as well as certain other pathological conditions (25). Androgens bind and activate the androgen receptor (AR), a 110 kDa nuclear receptor transcription factor (5). PCa is initially androgen-dependent early in disease course. However, progression to a state of castration resistance typically occurs in the case of non-localized disease after androgen deprivation therapy (611). AR is expressed in primary PCa and also in castrate resistant tumors, where AR expression may actually increase with progression to metastases (12). Recent work from a number of laboratories confirms the continuing involvement of the AR throughout disease progression (1315).

Our group recently showed that activation of the epidermal growth factor receptor (EGFR) by the prostate stromal growth factor, HB-EGF, regulates AR expression by a rapamycin-sensitive mechanism in LNCaP human PCa cells (16). EGFR activation by HB-EGF suppressed AR protein levels, while rapamycin, an inhibitor of the serine-threonine kinase mTOR, antagonized this effect. Rapamycin alone also potently increased AR protein levels. Suppression of AR expression by EGFR activation was also seen in LNCaP xenografts in vivo in a separate study (17). The sensitivity to the mTOR inhibitor suggests that a repressive signal to AR passes through the phosphoinositide 3-kinase (PI3K)/Akt/mTOR pathway. In a search for intermediates within this pathway, we immunoprecipitated the serine-threonine kinase Akt from lipid raft-enriched subcellular fractions isolated from LNCaP cells and identified co-precipitating proteins by mass spectrometry. Using this approach, we recently found that the serine/threonine kinase, Mst1/STK-4, a pro-apoptotic protein, is a novel inhibitor of Akt that functions by a mechanism involving direct interaction between the two kinases (18). Here we describe the application of a similar strategy that resulted in the finding that the nucleic acid binding protein, heterogeneous ribonucleoprotein K (hnRNP-K), regulates AR expression by a post-transcriptional mechanism.

HnRNP-K was originally identified as a poly(C) binding protein (PCBP) that exhibits a high affinity, sequence-specific interaction with poly(C) RNA (19). PCBPs comprise two subsets in mammalian cells: (1) hnRNP-K and hnRNP-J and (2) the α-complex proteins (α-CPs), (20)). HnRNP-K contains three conserved KH (K homology) nucleic acid binding domains and a KI (kinase interacting) region that mediates interaction with a variety of protein targets (21). HnRNP-K resides in both the nucleus and the cytoplasm and has been linked to a variety of cellular processes, including mRNA translation, transcription, RNA processing, RNA shuttling and stabilization, chromatin remodeling, and cell survival (2226). These diverse functions arise from the capability of hnRNP-K to bind both RNA and DNA. Here we present the first evidence that hnRNP-K is an endogenous inhibitor of AR protein translation, along with additional data consistent with a role for this protein in PCa.

Materials and Methods

Materials

R1881 was purchased from Sigma Chemical Co. (St. Louis, MO). AR and hnRNP-K antibodies are from Santa Cruz Biotechnology (Santa Cruz, CA). EGFR, lamin, α-tubulin, and Akt antibodies are from Cell Signaling Technology (Beverly, MA). KDR antibody is a generous gift from Dr. Soumitra Pal (Children’s Hospital Boston). ECL detection kit was from New England Nuclear, Inc. (Boston, MA). Smart pool siRNA for hnRNP-K is from Dharmacon, Inc. (Lafayette, CO). Casodex is a gift from Zeneca Pharmaceuticals (Cheshire, SK10 2NA, UK). The TNT Quick Coupled Transcription/Translation Systems is from Promega (Madison, WI). LightShift Chemiluminescent EMSA Kit, and Biotin Labeling Kit are from Pierce (Rockford, IL).

Cell culture

LNCaP cells were cultured in RPMI 1640 and COS or HeLa cells in Dulbecco’s modified Eagle (DMEM) medium supplemented with 10% FBS, 2% glutamine, and 1% antibiotics. Cell proliferation was determined by viability assay using crystal violet.

Plasmid constructions and mutagenesis

HnRNP-K constructs (27) and PSA-Luc reporter (p61-Luc) and AR 5′UTR-Luc (16) were described. HnRNP-K binding site mutants 414 and 478 were constructed by site-directed mutagenesis according to the QuikChange II Mutagenesis Kit (Stratagene, La Jolla, CA). Primers used to exchange nucleotides were 5′-ctcaccacccttctcaaaacccgcccacccgccc – 3′/5′-gggcgggtgggcgggttttgagaagggtggtgag for 414 and 5′-tgcagagaggtaactcaatttggctgcgagcggg- 3′/5′-cccgctcgcagccaaattgagttacctctctgca - 3′ for 478. Combination of 414/478 (double mutants) was achieved using the QuikChange Site-Directed Mutagenesis Kit (Stratagene) according to the manufacturer’s protocol.

Nuclear extract preparation

Cells were suspended in 1 ml of cold buffer [20 mM HEPES (pH 7.4), 10 mM KCl, 2 mM MgCl2, 0.5% IGEPAL, 1mM DTT, 100 μM vanadate, and a cocktail of protease inhibitors] and placed on ice for 10 min. The 1500 × g supernatant was centrifuged again at 13000 × g for 15 min to collect the cytoplasmic fraction. The pellet, after washing, suspended in the above buffer with 0.5 M NaCl. Lysates were centrifuged for 15 min at 13,000 × g at 4°C, and supernatants (nuclear extract) collected.

Electroporation

Nucleofector (Amaxa Inc, Gaithersburg, MD) was used to electroporate 2 μg of human hnRNP-K plasmids or 100 nM of each siRNA into LNCaP cells according to the manufacturer’s protocols in presence and absence of R1881.

Transient transfections and luciferase assays

Transient transfections were performed with Lipofectamine 2000 (Invitrogen, Carlsbad, CA) as described (29). Plasmid DNA (0.5–1 μg) was used and either R1881 (1 nM), rapamycin (100 nM) or respective vehicles (ethanol/BSA) was added for 20 h in serum-free medium.

Luciferase activity was measured using a dual luciferase assay kit as described (29). Either Renilla or total protein was used to normalize the relative luciferase activity. Background luciferase activity (pGL3 basic for p61PSA-Luc) was also measured for comparison. All experiments were carried out in triplicate and repeated three times using different preparations of plasmids.

RNA silencing

ON-TARGETplus SMART pool small interfering RNA (siRNA) oligonucleotides (Dharmacon) were designed to target different splice variants of hnRNP-K. Controls were scrambled sequences.

Immunoprecipitation and Western blot analysis

Triton X100 soluble (TS) and insoluble (TI) lipid raft proteins were prepared essentially as described (28). Protein (400 μg) was immunoprecipitated with either polyclonal hnRNP-K or monoclonal Akt antibody with protein A Sepharose beads. Bead-bound complexes were immunodetected as described (29). Dilutions for primary antibodies were: 1:1000 for AR, HA, and Akt, 1: 2000 for EGFR and KDR, and 1:10,000 for β-actin). Horseradish peroxidase conjugated secondary antibodies were used at a dilution of 1:5000. Immunoreactive bands were visualized by chemiluminescence.

GST pull-down

Bacterially expressed GST-hnRNP-K fusion proteins were adsorbed to glutathione-Sepharose beads (GE-Healthcare BioSciences AB), incubated with purified recombinant Akt1 (Upstate Biotechnology, Lake Placid, NY) in binding buffer (20 mM Tris pH 8.0, 137 mM NaCl, 10% glycerol, 1% NP-40) for 60 min at 4°C, and analyzed by western blotting using Akt-specific antibody (Cell Signaling).

RNA gel mobility-shift (REMSA)

Nonradioactive REMSA was performed with biotinylated AR 5′-UTR RNA probe and purified GST-hnRNP-K fusion protein. The biotinylated probe was prepared using Biotin-16-UTP and Xba I digested 5′-UTR AR construct using the AmpliScribe T7 and T3-Flash transcription kits. The binding reaction was performed using the LightShift Chemiluminescent EMSA kit. The RNase T1 step (10 min at 22°C) and heparin (5mg/ml) addition step (10 min at 22°C) was performed prior to incubation with labeled riboprobe. For competition assays, unlabelled RNA (25 and 100 ng) was added for 10 min prior to the incubation with labeled probe. The binding reaction, gel electrophoresis, and the electrophoretic transfer to nylon membrane were as described in the Pierce protocol. Samples were fixed on the membrane by UV cross-linking for 3 min (UV Stratalinker 2400, Stratagene) and detected by chemiluminescence.

In vitro translation

T7-hAR constructs (16) were transcribed and translated in a coupled rabbit reticulocyte lysate system (Promega) in the absence and presence of recombinant GST-hnRNP-K fusion protein. Reactions were performed at 30°C for 90 min either in presence of [35S]methionine for radioactive exposure or of unlabeled methionine. Translation of luciferase was used as a control.

Immunoprecipitation/reverse transcription-PCR

Cells were lysed in a buffer containing 10 mM HEPES pH 7.6, 3 mM MgCl2, 40 mM KCl, 5% glycerol, 0.2% NP-40, 1mM DTT, standard protease inhibitors and 50 units of RNaseOUT, and were incubated with either IgG or hnRNP-K antibodies for 45 min at 4°C. A mixture of protein-A and protein-G beads was added and incubated for 30 min. RNA was extracted and the One Step RT-PCR procedure (Invitrogen) was followed using the primers 5′-GCATGCCAGAGTGCCCTATC-3′ and 5′-TCCCAGAGTCATCCCTGCTTCAT-3′ for AR.

Polysome analysis

Polyribosome preparation was performed as described (30). In brief, LNCaP cells were treated with cyclohexamide (50 μg/ml) and in some cases with 200 μM puromycin for 60 min before collection. The cell pellet was resuspended in 300 μl low salt buffer (10 mM Tris, pH 7.4, 15 mM NaCl, 12.5 mM MgCl2, 500 μg/ml heparin, 50 μg/ml cyclohexamide) and 250 μl of lysis buffer (25 mM Tris, pH 7.4, 15 mM NaCl, 12.5 mM MgCl2, 1.2% NP-40, 500 μg/ml heparin, 5% (wt/vol) sucrose) was added. The 1000 × g extract was loaded onto a linear sucrose gradient [15 to 45% (wt/wt) sucrose] in 10 mM Tris buffer, pH 7.4, 200 mM NaCl, 10 mM MgCl2, 200 μg heparin with a SW 28 rotor and centrifuged at 126,000 × g at 4°C for 80 min. Twelve gradient fractions were collected (0.5 ml each) and polysome profile (OD260) was determined. RNA was extracted by Trizol reagent (Invitrogen) and RT-PCR was performed using the primers (5′-CACCCGTCTTCAGGGCTTCTTGGTTT-3′ and 5′-CATTTCACCATCTGGTTGGCTGGCTC-3′.

Tissue microarray immunohistochemistry

The human prostate tissue microarray (TMA) consists of benign and prostate tumor tissues from the Brigham and Women’s Hospital (Boston, MA), University of Michigan (Ann Arbor, MI), and the University of Ulm (Ulm, Germany) and is composed of benign prostate (n=18), localized PCa (n=36), and metastases (n=36). The metastatic cohort consists of 18 lymph-node hormone naïve and 18 hormone- (castrate-) resistant distant metastases. One pathologist (DDV) excluded cases if adequate paraffin-embedded tumor tissue was not available; consequently a total of 14 benign, 24 organ-confined tumor and 24 metastases were included. Immunohistochemistry for hnRNP-K was performed on 5μm sections using the avidin-biotin procedure, and analyzed semi-quantitatively by the ACIS System (ChromaVision Medical System, Inc., San Juan Capistrano, CA) as described (18,31). The obtained values were analyzed alone and in combination with serial sections stained with anti-AR antibody (Michael P. Lisanti, unpublished observations). The resulting intensity expression values (range from 0 to 255), for each TMA core, were transformed, mean centered and standard deviation set to 1, to standardize the variables to the same scale. Quadruplicate data points of each case were averaged, after assessment of intra-case expression homogeneity with respect to intra-diagnostic group variability (32). Expression level differentiation among diagnostic groups was assessed by t-test for unpaired data. To assess potential correlations, intensity mean values were dichotomized and Fisher exact test was applied on contingency tables. All p-values were considered 2-tailed and 0.05 was used as upper threshold for statistical significance. Origin 8 software (OriginLab Corporation, Northampton, MA) was used for statistical analysis. Specimens positive for hnRNP-K were classified as expressing 1) nuclear (N) or 2) nuclear + cytosolic (N+C) signal and the number of samples with cytoplasmic localization were counted and statistically analyzed across the diagnostic groups using a t-test for unpaired data.

Results

HnRNP-K down-regulates AR protein levels

LNCaP cells stably engineered to express myristoylated Akt1 (Myr-Akt1) (28), and HEK293 cells transiently transfected with Myr-Akt1, were used to generate Akt1 immune complexes for analysis by tandem mass spectrometry (LC-MS/MS). Lipid raft-enriched fractions were used for this purpose (28). HnRNP-K was identified by LC-MS/MS in these immunoprecipitates in multiple independent trials (not shown). We verified by co-immunoprecipitation that endogenous hnRNP-K forms a complex with Myr-Akt1 preferentially in lipid raft fractions (Fig. S1A). GST-hnRNP-K also formed a complex with purified Akt in vitro (Fig. S1B), verifying the mass spectrometry data.

HnRNP-K has been shown to be regulate translation of a number of mRNAs (3336) and the protein lies downstream from the EGFR in some contexts (37). Consequently, hnRNP-K is a logical intermediate in the previously demonstrated EGFR→mTOR pathway in which AR protein levels are down-regulated at the level of mRNA translation (16).

To test the potential involvement of hnRNP-K in this mechanism, we first examined the effect of hnRNP-K on AR expression in LNCaP cells. Enforced expression of hnRNP-K lowered AR protein levels in both the presence and absence of the synthetic androgen, R1881 (1nM), without any effect on the levels of the signaling receptors EGFR and KDR, or on β-actin (Fig. 1A). To determine whether hnRNP-K alters translocation of AR to the nucleus, we evaluated the extent of accumulation of AR in cytoplasmic versus nuclear fractions after enforced expression of hnRNP-K. We found a similar lowering of AR in nuclei and cytosol (Fig. 1A, right panel), indicating that hnRNP-K does not influence AR transit from cytoplasm to the nucleus, while it does alter steady-state levels of AR. Conversely, knockdown of endogenous hnRNP-K by RNA interference (Fig. 1B) increased AR levels, suggesting that endogenous hnRNP-K is conferring the same function as the overexpressed protein. HnRNP-K siRNA also attenuated the suppressive effect of HB-EGF on AR mRNA levels (Fig. 1C, Fig. S2), suggesting that hnRNP-K controls AR levels downstream from the EGFR.

Figure 1.

Figure 1

HnRNP-K downregulates AR expression in the presence and absence of androgen. (A, left panel) LNCaP cells were transiently transfected with either hnRNP-K or vector constructs using Nucleofection (Amaxa). Protein was transferred to nitrocellulose and blotted either with AR, HA (for hnRNP-K), EGFR, KDR, or β-actin antibody. (A, right panel) Fractionation of LNCaP cells before and after hnRNP-K transfection using Lipofectamine 2000 (Cy, cytoplasmic fraction and NE, nuclear fraction); α-tubulin and lamin were used as cytoplasmic and nuclear markers, respectively. (B, left panel) Knockdown of hnRNP-K increased AR expression in the presence and absence of 1 nM R1881 (using Nucleofection as transfection method). (B, right panel) Nuclear (NE) and cytoplasmic (Cy) fractionation of LNCaP cells after knockdown of endogenous hnRNP-K. (C) Knockdown of endogenous hnRNP-K reverts HB-EGF- (EGFR-) mediated down-regulation of AR. (D) Steady-state AR protein level in the presence and absence of actinomycin D (5 μM for 24 h) and R1881 (1 nM) after hnRNP-K transfection. In each case, data were quantified using NIH Image 1.63 software and represented as the ratio of AR and β-actin levels.

In order to assess whether the effect of hnRNP-K on AR is confined to LNCaP cells, (AR-negative) HeLa cells were transiently co-transfected with hnRNP-K along with human AR expression constructs and the effect of enforced expression of hnRNP-K on AR levels was monitored. Our results showed a significant decrease of AR expression in the presence of hnRNP-K in the presence and absence of androgen (Fig. S3), consistent with the findings in the LNCaP cell background.

Post-transcriptional regulation of AR by hnRNP-K

The experiments with HeLa cells suggested that the effect on AR was unlikely to be a result of changes in transcription of AR mRNA. Consistent with this hypothesis, we observed no changes in AR mRNA following enforced expression of hnRNP-K in LNCaP cells (Fig. S4A). HnRNP-K over-expression also did not affect AR mRNA levels when cells were treated with the transcription inhibitor, actinomycin D, for up to 20 h (Fig. S4B). Actinomycin D similarly did not alter the observed hnRNP-K effects on AR protein levels (Fig. 1D).

The selective mTOR inhibitor, rapamycin, increased AR expression in LNCaP cells, consistent with published results (16) (Fig. 2A). HnRNP-K siRNA evoked a comparable, but not additive, effect on AR protein levels, suggesting that hnRNP-K may act upstream of mTOR (Fig. 2A, right panel). Rapamycin exerts regulatory effects on cap-dependent mRNAs (38). EGFR activation was previously shown to attenuate AR protein levels in a rapamycin-sensitive manner, at least partly through the AR mRNA 5′-UTR (16). Thus, we analyzed the effect of enforced expression of hnRNP-K on a 570 nt fragment of the AR 5′-UTR, which contains several cis-acting elements and cap- and stem-loop secondary structures and was shown previously to be rapamycin-sensitive (16). To do this, we used a luciferase plasmid system in which the 5′-UTR fragment regulates the CMV promoter and that reports effects on cap-dependent mRNA translation (16). Consistent with published findings (16), rapamycin induced luciferase expression from this plasmid construct (Fig. 2B, left panel). In the presence and absence of rapamycin, transient expression of hnRNP-K in LNCaP cells inhibited luciferase expression. These data strongly suggest that hnRNP-K exerts its attenuating effects on AR expression at the level of mRNA translation. Consistent with this interpretation, we verified the direct binding of hnRNP-K with AR 5′-UTR RNA corresponding to this same UTR region by RNA gel mobility shift in concert with purified GST-hnRNP-K (Fig. S5), and also with endogenous AR mRNA in whole cells using immunoprecipitation/reverse transcription-PCR (Fig. 2B, right panel). The inhibitory effect of hnRNP-K on expression driven by the AR 5′-UTR (Fig. 2B) does not require the AR, since similar findings were obtained in AR-negative COS cells (Fig. 2C, left panel).

Figure 2.

Figure 2

HnRNP-K regulates AR expression at the AR 5′-untranslated region (5′-UTR). (A, left panel) LNCaP cells were transfected with hnRNP-K expression plasmid and treated with either 100 nM rapamycin and/or 1 nM R1881. (A, right panel) Determination of AR level in presence and absence of hnRNP-K siRNA and 100 nM rapamycin alone or in combination. All data were analysed by NIH Image 1.63 and represented as the ratio of AR and β-actin. (B, left panel) Effect of hnRNP-K on AR 5′-UTR luciferase activity in presence and absence of rapamycin. AR 5′-UTR luciferase plasmid (500 ng) or the control plasmid (500 ng) were cotransfected with 500 ng of hnRNP-K in LNCaP cells. Lysates were assayed for luciferase activity and protein content. (B, right panel) To verify that hnRNP-K binds to AR mRNA, LNCaP cells were immunoprecipitated with either IgG or hnRNP-K antibody and RNA was extracted and subjected to RT-PCR analysis using AR specific primers. Negative controls included: no added reverse transcriptase (-RT), beads (protein A/G) alone, and IgG immunoprecipitate. Specific PCR band in extracts of LNCaP cells was used as a positive control. (C) COS and LNCaP cells were transfected with control plasmid (pIR-Luc) plasmid or plasmid containing the AR 5′-UTR in the presence of varying amounts of hnRNP-K. (D, left panel) Schematic representation of AR 5′-UTR-Luc constructs highlighting the conserved hnRNP-K binding motifs (UCCCCA and UCCCU) in human AR mRNA. These two motifs were altered to UCAAAA (414 mutant) and UCAAU (478 mutant). (D, right panel) Either AR 5′UTR-Luc wild type, or single (M414 and M478) and double mutants (DM 414/478) were tested for activity in the presence of varying doses of hnRNP-K.

Inspection of the 570 nt rapamycin-sensitive region in the AR 5′-UTR using the consensus sequence U(C)nA/U (40) revealed two potential hnRNP-K binding sites at nt 412–417 (UCCCCA) and 476–480 (UCCCU). To evaluate whether either of these UC-rich regions mediates some or all of the inhibitory effects of hnRNP-K on the translation reporter, we generated single and double mutants within the rapamycin-sensitive fragment (Fig. 2D, left panel). Alteration of the consensus sequence (by mutating UCCCCA to UCAAAA) at the 412–417 site, but not alteration of the 476–480 site, abolished the inhibitory effects of hnRNP-K on reporter expression (Fig. 2D, right panel), confirming the involvement of the 412–417 site within the AR 5′-UTR in the regulatory effect we observed with hnRNP-K.

HnRNP-K inhibits translation of AR mRNA

HnRNP-K regulates translation of the L2 capsid protein of human papillomavirus type 16 by binding within the open reading frame of the coding region (35). Inspection of the entire AR mRNA indicated that the message may harbor as many as 9 other potential hnRNP-K binding sites in the coding region and in the 3′-UTR (Table 1) suggesting the possibility that hnRNP-K may also regulate AR expression through one or more of these additional sites. To address this question, we examined the effect of hnRNP-K on translation of AR mRNA using a construct containing only the AR coding region, which contains 6 potential hnRNP-K binding sites (Table 1). AR was synthesized with a coupled transcription/translation system, using either [35S]methionine for radioactive detection or unlabeled methionine for Western blot identification. Addition of purified GST-hnRNP-K dose-dependently decreased the synthesis of the 100 and 110 kDa translated products (Fig. 3). In contrast, no translation inhibition was observed on the internal control luciferase mRNA (lower panels). Results with cold methionine (Fig. 3B) confirmed the identity of the translation products as AR by immunoblotting. In a complementary experiment, enforced expression of hnRNP-K in HeLa cells reduced the expression of AR expressed from the same hAR construct used in the coupled transcription-translation experiments (Fig. S3). These findings indicate that hnRNP-K employs one or more binding sites within the coding region in a manner that is functionally compatible with the single binding site we identified in the rapamycin-sensitive region of the 5′-UTR.

Table 1.

Consensus hnRNP-K[U(C)nA/U] binding sites in human AR mRNA

Position Sequence
5′UTR 412–17 UCCCCA

476–80 UCCCU

664–68 UCCCA

ORF 1403–08 UCCCCA

1510–14 UCCCA

1608–12 UCCCU

2255–66 UCCCCA-UCCCCA

2656–65 UCCCA-UCCCA

3′UTR 3887–901 UCCCCA

4090–94 UCCCA

4115–19 UCCCA

Figure 3.

Figure 3

HnRNP-K inhibits the translation of AR mRNA. (A) Effect of purified GST-hnRNP-K on translation of AR mRNA as determined by coupled in vitro transcription/translation using [35S]methionine. (B) Western blot analysis with monoclonal AR antibody after in vitro translation with unlabeled methionine. Luciferase (lower panel) was used as a translation control in A and B. (C) LNCaP cells were treated with R1881 in the presence and absence of hnRNP-K for 24 h. A cytoplasmic extract was prepared and centrifuged through a linear sucrose gradient to isolate polysomes. Total RNA was extracted from each of twelve sucrose gradient fractions. RT-PCR was performed using AR and β-tubulin primers. PCR products were separated on ethidium bromide stained 1% agarose gels. The polysome profile was determined by measurement of the OD260 for each fraction. (D) Polysome profile (OD260) and polysome analyses were performed in the presence of 200 μM puromycin. LNCaP cells were treated with indicated amount of puromycin for 30 min before sucrose gradient fractionation.

In an independent test of the ability of hnRNP-K to act as a regulator of AR translation, we probed the distribution of AR mRNA in polysomal fractions isolated from LNCaP cells under conditions where hnRNP-K expression was enforced. If hnRNP-K acts as an inhibitor of AR mRNA translation at the level of initiation, then its forced expression would result in the dissociation of AR mRNA from actively translating ribosomes and a repartitioning of the mRNA toward the lighter fractions of the gradient. As indicated in Figure 3C (left and right panels), hnRNP-K had no such effect on the distribution of AR mRNA in gradient fractions, suggesting that the protein does not inhibit translation initiation of AR mRNA. HnRNP-K had no effect on the distribution of tubulin mRNA (Fig. 3C, bottom panels), as expected. Next, we tested the possibility that the AR mRNA under conditions of enforced hnRNP-K expression was associated with translationally-inactive ribosomes, an event generally associated with a defect in peptide elongation. To test this, we used puromycin, which causes premature termination and release of all actively elongating mRNAs from polysomes. If an mRNA species is not being actively translated, puromycin will not be incorporated into the peptide chain and mRNA release from polysomes will not occur. In the absence of elevated hnRNP-K, puromycin caused a release of tubulin and AR mRNAs from the polysomal fractions (compare Fig. 3C left panel, with 3D left panel, particularly lanes 8–11 which contain AR and tubulin mRNAs associated with large polysomes). However, when hnRNP-K expression was enforced, AR mRNA was retained in heavy polysomal fractions, even after puromycin treatment (compare Fig. 3C and D), indicating that hnRNP-K evokes a reduction in rate of AR mRNA chain elongation.

HnRNP-K inhibits PSA promoter activity and LNCaP cell proliferation

In order to determine whether AR down-regulation by hnRNP-K has functional consequences in PCa cells, we assessed the effect of enforced hnRNP-K expression on the androgen-responsive human prostate-specific antigen (PSA) promoter, and on LNCaP cell growth. Androgen-induced activation of the reporter gene was strongly and dose-dependently inhibited by hnRNP-K (Fig. 4A), indicating that hnRNP-K antagonizes androgenic signaling, consistent with a role as an AR antagonist. Similarly, hnRNP-K overexpression suppressed LNCaP cell proliferation (Fig. 4B), a result consistent with the PSA promoter-reporter data.

Figure 4.

Figure 4

HnRNP-K inhibits AR-dependent gene expression and LNCaP cell growth. (A) Enforced expression of hnRNP-K dose dependently inhibits a luciferase reporter driven by the PSA promoter. (B) Enforced expression of hnRNP-K inhibits proliferation of androgen-dependent LNCaP cells. The AR inhibitor Casodex (bicalutamide) was used at a concentration of 10 μM.

HnRNP-K expression in human prostate cancer

Multiple data sets in the Oncomine cancer profiling database (www.oncomine.org) indicate that hnRNP-K is expressed in human PCa. We analyzed a human PCa TMA containing benign prostate tissues, organ-confined cancers, and metastases [hormone naive (HN) and hormone (castration) resistant (HR)] using a monospecific anti hnRNP-K antibody. HnRNP-K was detected in the nucleus of the majority of benign and carcinoma samples at high levels, and expression was significantly increased in metastatic tumors in comparison with benign tissues and organ-confined tumors (p = 0.0001 and 0.0004, respectively) (Fig. 5A, left panel), a trend that was maintained when HR metastases were compared with HN metastases (Fig. 5A, right panel). Notably, hnRNP-K and AR levels were significantly inversely correlated in localized PCa specimens (p < 0.001) (Fig. 5B). Finally, hnRNP-K was cytosolic and nuclear localized in organ-confined tumors, but cytosolic expression was significantly decreased in the metastases (Fig. 5C, D).

Figure 5.

Figure 5

Quantitative analysis of hnRNP-K and AR levels in a human prostate TMA. (A) Relative levels of hnRNP-K and AR as assessed by ChromaVision analysis of TMA containing benign, local PCa, and metastatic tumors (left panel). The right panel shows averaged values of hnRNP-K and AR in the metastatic subset (HN: hormone naïve; HR: hormone resistant). (B) Regression analysis showing a significant inverse correlation between hnRNP-K and AR in organ-confined tumors. (C) Representative micrographs showing hnRNP-K expression and subcellular localization. Note the loss of cytoplasmic staining in the metastatic tissue (lower right panel). (D) The graph shows the number of cases among the different diagnostic groups with predominant nuclear (N) or nuclear + cytosolic (N+C) staining. PCa, organ confined prostate cancer; Mets, metastases.

Discussion

In this study we demonstrate that the nucleic acid binding protein hnRNP-K is a physiologically relevant mediator of AR expression, which functions by the novel mechanism of repression of AR mRNA translation. We verified a previous observation that a rapamycin-sensitive region within the 5′-UTR of the AR message harbors regulatory information that modifies rates of AR translation (16), however we now go on to show that hnRNP-K also uses information within the AR coding region independently of the 5′-UTR to regulate AR translation. We found that hnRNP-K acts to repress AR expression, androgen-regulated promoter activity, and androgen-induced PCa cell growth. Although AR expression is regulated by transcriptional and protein stability mechanisms (4042), the potential for translational regulation of the AR mRNA is poorly studied and has not been fully established prior to this report.

Mizokami and Chang (43) implicated a ~0.6 kb region of the AR 5′-UTR as a component of the AR expression regulatory mechanism. In that study, this segment of the untranslated region was shown to regulate expression of a reporter gene in a transcription-independent manner, suggesting the possibility of the existence within this region of one or more cis elements that control the translational apparatus in vivo. A more recent study (16) identified this same region in the AR 5′-UTR as sensitive to the mTOR inhibitor, rapamycin. This result is consistent with an earlier report demonstrating that continuous in vivo exposure of LNCaP xenografts to the EGFR activating ligand, HB-EGF, caused a stable reduction of AR levels in the tumors (17). Our identification of a functional hnRNP-K regulatory element at nt positions 412–417 within the rapamycin-sensitive element of the 5′-UTR is in agreement with these previous observations. The results we now describe indicate that this site is required to mediate attenuation of luciferase expression from a cap-dependent AR 5′-UTR translation reporter when hnRNP-K expression is enforced (Fig. 2). Surprisingly, however, we found that this site is not required for the down-regulating effect of hnRNP-K on AR levels, pointing to redundancy in the mechanism controlling AR expression.

We have shown that enforced expression of hnRNP-K in LNCaP cells dramatically lowered AR protein levels, while siRNA knockdown of endogenous hnRNP-K elevated AR expression. HnRNP-K siRNA inhibited the ability of EGFR activation to reduce steady-state AR levels and also mimicked the AR-inducing effects of rapamycin seen in LNCaP cells. Similarly, hnRNP-K overexpression antagonized the ability of rapamycin to enhance AR expression. We confirmed that hnRNP-K binds to AR mRNA and demonstrated that hnRNP-K binds and inhibits the activity of an AR 5′-UTR translation reporter. HnRNP-K inhibited AR translation in both in vivo and in vitro experimental formats. Finally, we showed that enforced expression of hnRNP-K in LNCaP cells redistributed AR mRNA to translationally-inactive ribosomes in concert with the attenuation of androgen-dependent biological responses. Collectively, these data provide strong evidence that hnRNP-K is an endogenous regulator of AR translation in human cells.

Analysis of a human PCa tissue microarray demonstrated that hnRNP-K and AR levels were inversely correlated in local PCa, and substantial loss of cytosolic staining was seen with with progression to metastases. Thus, the presence of the AR inhibitor, hnRNP-K, correlates with reduced AR levels in a tumor subset. Furthermore, a reduction in cytosolic hnRNP-K, which presumably would be available to inhibit translation, is seen with progression to the lethal phenotype, where the AR is believed to remain active in the absence of normal levels of circulating androgen. If hnRNP-K serves to inhibit expression of AR in vivo, these data may reflect the regulatory process we identify here in the human disease.

Yeap et al. reported that several RNA binding proteins, including HuR, CP1 and CP2 bind AR mRNA at specific sites within the 3′-UTR (44), although the function of these interactions was not explored in that study. It is possible that dysregulation of multiple mechanisms affecting AR mRNA translation and/or mRNA stability may be operative in PCa. Our data are compatible with this possibility. We observed here that metastatic tumors retain hnRNP-K in tumor cell nuclei, suggesting that the functional role of the protein may change with disease progression. Consistent with this idea, hnRNP-K was recently identified as a component of a transcriptional complex capable of regulating the AR gene in LNCaP cells (45). The possibility that hnRNP-K serves multiple functions in prostate and other tumor cells in a manner that can affect disease progression deserves further study.

A role for hnRNP-K in translation of other mRNAs has been identified (3336,46,47), with the protein described as a translational regulator of c-myc (33), renin (34), human papillomavirus type 16 L2 capsid protein (35), and reticulocyte-15-lipoxygenase (LOX) (36) mRNAs. The premature expression of LOX in erythroid precursor cells is restricted by a translational silencing mechanism mediated by hnRNP-K binding to the differentiation control element (DICE) in the LOX mRNA (36). In contrast to translational silencing of LOX, hnRNP-K promotes internal ribosome entry on c-myc mRNA (33), indicating that the protein can play stimulatory as well as inhibitory functions in translational regulation. Interestingly, Atsushi et al. recently reported that cytoplasmic accumulation of hnRNP-K mediates metastasis in human fibrosarcoma HT1080 cells (48). Because the AR is an important mediator of masculinization and other physiologic mechanisms in multiple cell types, including in non-reproductive tissues such as skeletal muscle and brain, it is possible that hnRNP-K regulates AR translation in other contexts. Furthermore, it cannot be assumed that the protein will always act to suppress AR. In fact, the literature suggests that a more complicated role for hnRNP-K in regulating AR expression in heterologous contexts is more likely. Because AR expression in breast cancer may be protective rather than tumor-promoting (49), it would be interesting to compare the role of hnRNP-K in regulating the AR in this alternative context.

In summary, we have provided some of the first mechanistic evidence for a prominent role for mRNA translation in regulating AR protein expression. Because our data suggest that hnRNP-K is downstream from the EGFR, and most likely Akt, the protein may be important in the network of mechanisms that control AR expression and function in a variety of normal and pathologic conditions in which these signaling pathways have been implicated. In addition, given the prominent role of the AR in PCa progression, including in castrate resistant disease, these findings provide a rationale to consider chemotherapeutic agents directed against the protein synthesis machinery in PCa treatment (50).

Acknowledgments

The authors thank Delia Lopez and Paul Guthrie for technical assistance, Christopher Lafargue, Sven Pern, and Francesca Demichelis for support with the Chromavision system, and Matteo Morello for assistance with evaluation of the TMA. This study was supported by NIH R37 DK47556 and R01 CA112303 (to M.R.F.) and US Department of Defense (DoD) grant W81XWH-08-1-0150 (to M.R.F.). D.D.V. is a recipient of K99 CA131472. M.H.H. is an AUA Foundation Research Scholar. M.H.H. and B.C. were supported by US DoD Postdoctoral Fellowships. J.K. is an Edwin Beer Scholar of the New York Academy of Medicine.

References

  • 1.Denmeade SR, Isaacs JT. Development of prostate cancer treatment: the good news. Prostate. 2004;58:211–24. doi: 10.1002/pros.10360. [DOI] [PubMed] [Google Scholar]
  • 2.Roy AK, Lavrovsky Y, Song CS, et al. Regulation of androgen action. Vit Horm. 1999;55:309–52. doi: 10.1016/s0083-6729(08)60938-3. [DOI] [PubMed] [Google Scholar]
  • 3.Schatzl G, Madersbacher S, Gsur A, et al. Association of polymorphisms within androgen receptor, 5a-reductase, and PSA genes with prostate volume, clinical parameters, and endocrine status in elderly men. Prostate. 2002;52:130–38. doi: 10.1002/pros.10101. [DOI] [PubMed] [Google Scholar]
  • 4.Lee D. High androgen receptor levels are predictive of decreased survival in prostate cancer. Clin Prostate cancer. 2003;1:13–14. doi: 10.1016/s1540-0352(11)70012-9. [DOI] [PubMed] [Google Scholar]
  • 5.Heinlein CA, Chang C. Androgen receptor in prostate cancer. Endocrine Reviews. 2008;25:276–308. doi: 10.1210/er.2002-0032. [DOI] [PubMed] [Google Scholar]
  • 6.Grossmann ME, Huang H, Tindall DJ. Androgen receptor signaling in androgen-refractory prostate cancer. J Natl Cancer Inst. 2001;93:1687–97. doi: 10.1093/jnci/93.22.1687. [DOI] [PubMed] [Google Scholar]
  • 7.Hara T, Nakamura K, Araki H, et al. Enhanced androgen receptor signaling correlates with the androgen refractory growth in a newly established MDA Pca 2b-hr human prostate cancer cell subline. Cancer Res. 2003;63:5622–8. [PubMed] [Google Scholar]
  • 8.Chen CD, Welsbie DS, Tran C, et al. Molecular determinants of resistance to antiandrogen therapy. Nat Med. 2004;10:33–9. doi: 10.1038/nm972. [DOI] [PubMed] [Google Scholar]
  • 9.Taplin ME, Balk SP. Androgen receptor: a key molecule in the progression of prostate cancer to hormone independence. J Cell Biochem. 2004;91:483–90. doi: 10.1002/jcb.10653. [DOI] [PubMed] [Google Scholar]
  • 10.Debes JD, Tindall DJ. Mechanisms of androgen refractory prostate cancer. N Engl J Med. 2004;351:1488–90. doi: 10.1056/NEJMp048178. [DOI] [PubMed] [Google Scholar]
  • 11.Scher HI, Sawyers CL. Biology of progressive, castration-resistant prostate cancer: directed therapies targeting the androgen-receptor signaling axis. J Clin Oncol. 2005;23:8253–61. doi: 10.1200/JCO.2005.03.4777. [DOI] [PubMed] [Google Scholar]
  • 12.Hara T, Miyazaki H, Lee A, et al. Androgen receptor and invasion in prostate cancer. Cancer Res. 2008;68:1128–35. doi: 10.1158/0008-5472.CAN-07-1929. [DOI] [PubMed] [Google Scholar]
  • 13.Henshall SM, Quinn DL, Lee CS, et al. Altered expression of androgen receptor in the malignant epithelium and adjacent stroma is associated with early relapse in prostate cancer. Cancer Res. 2001;61:423–7. [PubMed] [Google Scholar]
  • 14.Hobisch A, Culig Z, Radmayr C, et al. Distant metastases from prostatic carcinoma express androgen receptor protein. Cancer Res. 1995;55:3068–72. [PubMed] [Google Scholar]
  • 15.Linja M, Savinainen K, Saramaki O, et al. Amplification and overexpression of androgen receptor gene in hormone-refractory prostate cancer. Cancer Res. 2001;61:3550–5. [PubMed] [Google Scholar]
  • 16.Cinar B, De Benedetti A, Freeman MR. Post-translational regulation of the androgen receptor by Mammalian target of rapamycin. Cancer Res. 2005;65:2547–53. doi: 10.1158/0008-5472.CAN-04-3411. [DOI] [PubMed] [Google Scholar]
  • 17.Adam RM, Kim J, Lin J, et al. Heparin-binding epidermal growth factor-like growth factor stimulates androgen-independent prostate tumor growth and antagonizes androgen receptor function. Endocrinology. 2002;143:4599–608. doi: 10.1210/en.2002-220561. [DOI] [PubMed] [Google Scholar]
  • 18.Cinar B, Fang PK, Lutchman M, et al. The proapoptotic kinase Mst1 and its caspase cleavage products are direct inhibitors of Akt. EMBO J. 2007;26:4523–34. doi: 10.1038/sj.emboj.7601872. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Swanson MS, Dreyfuss G. Classification and purification of proteins of heterogeneous nuclear ribonucleoprotein particles by RNA-binding specificities. Mol Cell Biol. 1988;8:2237–41. doi: 10.1128/mcb.8.5.2237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Matunis MJ, Michael WM, Dreyfuss G. Characterization and primary structure of the poly (C)-binding heterogeneous nuclear ribonucleoprotein complex K protein. Mol Cell Biol. 1992;12:164–71. doi: 10.1128/mcb.12.1.164. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Bomsztyk K, Denisenko O, Ostrowski J. hnRNP K: One protein multiple processes. BioEssays. 2004;26:629–38. doi: 10.1002/bies.20048. [DOI] [PubMed] [Google Scholar]
  • 22.Ostrowski J, Bomsztcyk K. Nuclear shift of hnRNP-K protein in neoplasms and other states of enhanced cell proliferation. Br J Cancer. 2003;89:1493–150. doi: 10.1038/sj.bjc.6601250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Michelotti EF, Michelotti GA, Aronsohn AI, Levens D. Heterogenous nuclear ribonucleoprotein K is a transcription factor. Mol Cell Biol. 1996;16:2350–60. doi: 10.1128/mcb.16.5.2350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Holcik M, Liebhaber SA. Four highly stable eukaryotic mRNAs assemble 3′ untranslated region RNA-protein complexes sharing cis and trans components. Proc Natl Acad Sci USA. 1997;94:2410–14. doi: 10.1073/pnas.94.6.2410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Bomsztyk K, Van Seuningen I, Suzuki H, et al. Diverse molecular interactions of the hnRNP K protein. FEBS Lett. 1997;403:113–115. doi: 10.1016/s0014-5793(97)00041-0. [DOI] [PubMed] [Google Scholar]
  • 26.Mikula M, Dzwonek A, Karczmarski J, et al. Landscape of the hnRNP K protein-protein interactome. Proteomics. 2006;6:1–12. doi: 10.1002/pmic.200500632. [DOI] [PubMed] [Google Scholar]
  • 27.Lee MH, Mori S, Raychaudhuri P. Trans-activation by the hnRNP K protein involves an increase in RNA synthesis from the reporter genes. J Biol Chem. 1996;271:3420–27. doi: 10.1074/jbc.271.7.3420. [DOI] [PubMed] [Google Scholar]
  • 28.Adam RM, Mukhopadhyay NK, Kim J, et al. Cholesterol sensitivity of endogenous and myristoylated Akt. Cancer Res. 2007;67:6238–46. doi: 10.1158/0008-5472.CAN-07-0288. [DOI] [PubMed] [Google Scholar]
  • 29.Mukhopadhyay NK, Cinar B, Mukhopadhyay L, et al. The zinc finger protein ras-responsive element binding protein-1 is a coregulator of the androgen receptor: Implications for the role of the ras pathway in enhancing androgenic signaling in prostate cancer. Mol Endocrinology. 2007;21:2056–70. doi: 10.1210/me.2006-0503. [DOI] [PubMed] [Google Scholar]
  • 30.Ewen ME, Oliver CJ, Sluss HK, et al. p53-dependent repression of CDK4 translation in TGF-β-induced G1 cell-cycle arrest. Genes & Dev. 1994;9:204–17. doi: 10.1101/gad.9.2.204. [DOI] [PubMed] [Google Scholar]
  • 31.Di Vizio D, Demichelis F, Simonetti S, et al. Skp2 expression is associated with high risk and elevated Ki67 expression in gastrointestinal stromal tumours. BMC Cancer. 2008;8(1):134. doi: 10.1186/1471-2407-8-134. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Di Vizio D, Adam RM, Kim J, et al. Caveolin-1 interacts with a lipid raft-associated population of fatty acid synthase. Cell Cycle. 2008;7(14):2257–67. doi: 10.4161/cc.7.14.6475. [DOI] [PubMed] [Google Scholar]
  • 33.Evans JR, Mitchell SA, Spriggs KA, et al. Members of the poly (rC) binding protein family stimulate the activity of the c-myc internal ribosome entry segment in vitro and in vivo. Oncogene. 2003;22:8012–20. doi: 10.1038/sj.onc.1206645. [DOI] [PubMed] [Google Scholar]
  • 34.Skalweit A, Doller A, Huth A, et al. Posttranscriptional control of renin synthesis: identification of proteins interacting with renin mRNA 3′-untranslated region. Circ Res. 2003;92:419–27. doi: 10.1161/01.RES.0000059300.67152.4E. [DOI] [PubMed] [Google Scholar]
  • 35.Collier B, Goobar-Larsson L, Sokolowski M, Schwartz S. Translational inhibition in vitro of human papillomavirus type 16 L2 mRNA mediated through interaction with heterogenous ribonucleoprotein K and poly(rC)-binding proteins 1 and 2. J Biol Chem. 1998;273:22648–56. doi: 10.1074/jbc.273.35.22648. [DOI] [PubMed] [Google Scholar]
  • 36.Ostareck DH, Ostareck-lederer A, Shatsky IN, Hentze MW. Lipoxygenase mRNA silencing in erythroid differentiation: The 3′UTR regulatory complex controls 60S ribosomal subunit joining. Cell. 2001;104:281–90. doi: 10.1016/s0092-8674(01)00212-4. [DOI] [PubMed] [Google Scholar]
  • 37.Mandal M, Vadlamudi R, Nguyen D, et al. Growth factors regulate heterogeneous nuclear ribonucleoprotein K expression and function. J Biol Chem. 2001;276:9699–704. doi: 10.1074/jbc.M008514200. [DOI] [PubMed] [Google Scholar]
  • 38.Harris TE, Lawrence JC., Jr Tor signaling. Sci STKE. 2003;2003(212):re15. doi: 10.1126/stke.2122003re15. [DOI] [PubMed] [Google Scholar]
  • 39.Thiele BJ, Doller A, Kahne T, et al. RNA-binding proteins heterogeneous nuclear ribonucleoproteinA1, E1, and K are involved in post-transcriptional control of collagen I III synthesis. Circ Res. 2004;95:1058–66. doi: 10.1161/01.RES.0000149166.33833.08. [DOI] [PubMed] [Google Scholar]
  • 40.Gregory C, Johnson RJ, Mohler J, et al. Androgen receptor stabilization in recurrent prostate cancer is associated with hypersensitivity to low androgen. Cancer Res. 2001;61:2892–8. [PubMed] [Google Scholar]
  • 41.Wang LG, Ossowski I, Ferrari A. Overexpressed androgen receptor linked to p21/WAF1 silencing may be responsible for androgen independence and resistance to apoptosis of a prostate cancer cell line. Cancer Res. 2001;61:7544–51. [PubMed] [Google Scholar]
  • 42.Burnstein KL. Regulation of androgen receptor levels: Implications for prostate cancer progression and therapy. J Cell Biochem. 2005;95:657–69. doi: 10.1002/jcb.20460. [DOI] [PubMed] [Google Scholar]
  • 43.Mizokami A, Chang C. Induction of translation by the 5′-untranslated region of human androgen receptor mRNA. J Biol Chem. 1994;269:25655–59. [PubMed] [Google Scholar]
  • 44.Yeap BB, Voon DC, Vivian JP, et al. Novel binding of HuR and polyC- binding protein to a conserved UC-rich motif within the 3′-untranslated region of the androgen receptor messenger RNA. J Biol Chem. 2002;277:27183–192. doi: 10.1074/jbc.M202883200. [DOI] [PubMed] [Google Scholar]
  • 45.Wang LG, Johnson EM, Kinoshita Y, et al. Androgen receptor overexpression in prostate cancer linked to Purα loss from a novel repressor complex. Cancer Res. 2008;68:2678–88. doi: 10.1158/0008-5472.CAN-07-6017. [DOI] [PubMed] [Google Scholar]
  • 46.Ostareck-Lederer A, Ostareck DH, Neubauer G, et al. Src mediated phosphorylation of hnRNP-K drives translational activation of specifically silenced mRNAs. Mol Cell Biol. 2002;22:4535–43. doi: 10.1128/MCB.22.13.4535-4543.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ostareck-Lederer A, Ostareck DH, Standart N, Thiele BJ. Translation of 15-lipoxygenase mRNA is inhibited by a protein that binds to a repeated sequence in the 3′ untranslated region. EMBO J. 1994;13:1476–81. doi: 10.1002/j.1460-2075.1994.tb06402.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Inoue A, Sawata SY, Taira K, et al. Loss-of-function screening by randomized intracellular antibodies: Identification of hnRNP-K as a potential target for metastasis. Proc Natl Acad Sci USA. 2007;104:8983–8. doi: 10.1073/pnas.0607595104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Buchanan G, Birrell SN, Peters AA, et al. Decreased androgen receptor levels and receptor function in breast cancer contribute to the failure of response to medroxyprogesterone acetate. Cancer Res. 2005;65:8487–96. doi: 10.1158/0008-5472.CAN-04-3077. [DOI] [PubMed] [Google Scholar]
  • 50.Graff JR, Konicek BW, Vincent TM, et al. Therapeutic suppression of translation initiation factor eIF4E expression reduces tumor growth without toxicity. J Clin Invest. 2007;117:2638–48. doi: 10.1172/JCI32044. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES