Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

LEO1 is regulated by PRL-3 and mediates its oncogenic properties in Acute Myeloid Leukemia

Phyllis SY Chong1, Jianbiao Zhou1, Lip-Lee Cheong2, Shaw-Cheng Liu1, Jingru Qian3, Tiannan Guo3, Siu Kwan Sze3, Qi Zeng4, Wee Joo Chng1,2,5

1Cancer Science Institute of Singapore, National University of Singapore, Singapore 2Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 3Department of Biological Sciences, Nanyang Technological University, Singapore, Singapore 4Institute of Molecular and Cell Biology, A*STAR, Singapore 5Department of Haematology-Oncology, National University Cancer Institute of Singapore, National University Health System, Singapore

P. S-Y. Chong and J. Zhou contributed equally to this work.

Running title: LEO1 mediate PRL-3 oncogenic function in AML Keywords: PRL-3 phosphatase; Leo1; PAF complex; SILAC-based mass spectrometry Precis: This study advances understanding of an important oncogenic phosphatase in AML by identifying a critical downstream target in mediating its effects, with possible implications for prognosis and therapy.

Correspondence Associate Professor Wee-Joo Chng Cancer Science Institute of Singapore 14, Medical Drive, Singapore 117599 Telephone: (65) 6772 4613, Fax: (65) 6777 5545; Email: [email protected]

The authors disclose no potential conflicts of interest.

Word count: 5136 Figures: 7

1

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Abstract

PRL-3, an oncogenic dual-specificity phosphatase, is over-expressed in 50% of AML and associated with poor survival. We found that stable expression of PRL-3 confers cytokine independence and growth advantage of AML cells. However, how PRL-3 mediates these functions in AML is not known. To comprehensively screen for PRL3-regulated proteins in

AML, we performed SILAC-based quantitative proteomics analysis and discovered 398 significantly perturbed proteins after PRL-3 overexpression. We show that Leo1, a component of RNA polymerase II-associated factor (PAF) complex, is a novel and important mediator of PRL-3 oncogenic activities in AML. We described a novel mechanism where elevated PRL-3 protein increases JMJD2C histone demethylase occupancy on Leo1 promoter, thereby reducing the H3K9me3 repressive signals and promotes Leo1 expression.

Furthermore, PRL-3 and Leo1 levels were positively associated in AML patient samples

(N=24; p-value <0.01). On the other hand, inhibition of Leo1 reverses PRL-3 oncogenic phenotypes in AML. Loss of Leo1 leads to destabilization of the PAF complex and downregulation of SOX2 and SOX4, potent oncogenes in myeloid transformation. In conclusion, we identify an important and novel mechanism by which PRL-3 mediates its oncogenic function in AML.

2

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Introduction

PRL-3, encoded by the PTP4A3 gene, is a small dual-specificity phosphatase characterized by the conserved C(X5)R catalytic domain, and a unique C-terminal prenylation domain essential for its proper subcellular localization (1, 2). PRL-3 has been shown to promote cellular processes such as cell motility, invasion, cell growth and survival through various mechanisms (2-7). PRL-3 was first linked to cancer when it was consistently found at elevated levels in colorectal cancer (CRC) metastases, but at much lower levels in matched early-staged tumor and normal colorectal epithelium (8). Since then, elevated expression of

PRL-3 has been implicated in the progression and metastasis of an array of cancer types, including gastric, ovarian, cervical, lung, liver and breast, and PRL-3 protein was overexpressed in an average of 22.3% of 1008 human carcinoma samples examined using immunohistochemistry (9, 10). Together with the fact that it has a highly restricted basal pattern of expression in adult tissues (11), PRL-3 is deemed as an attractive therapeutic target which spares normal tissues. The potential of this target has been demonstrated using PRL-3 specific antibodies in an in vivo model (12).

In recent years, accumulating evidence suggests that PRL-3 is also a novel therapeutic target and biomarker in leukemia (13, 14). We were the first to report that elevated PRL-3 protein expression occurs in about 47% of human AML cases while absent from normal myeloid cells in bone marrow (13). In addition, a large-scale gene expression profiling study of 454 primary AML samples demonstrates that high PRL-3 levels is an independent negative prognostic factor in AML, both for overall survival and event-free survival (14). These reports collectively suggest that PRL-3 may be of biological and clinical relevance in AML and warrants further investigation.

3

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

In the present study, we created a TF-1 AML cell line stably over-expressing PRL-3 (TF1- hPRL3) as a model to study the biological relevance of PRL-3 in AML, and we use a quantitative proteomic strategy to profile on a global scale changes in protein expression induced by PRL-3 in AML. We found that PRL-3 has pro-oncogenic properties in AML, and

Leo1, a component of the human RNA polymerase II-associated factor (PAF) complex, is one of the most differentially expressed proteins induced by PRL-3. Further, we showed that

PRL-3 upregulates Leo1 through a novel epigenetic mechanism. On the other hand, knockdown of Leo1 significantly diminish the oncogenic effects of PRL-3. Our current work implicates a pro-oncogenic role of PRL-3 in AML, and reveals Leo1 as a novel downstream molecule required for PRL-3 oncogenic function in leukemia.

Materials and Methods

Cell culture HEK293T cells were cultured in Dulbecco's modified Eagle's medium (DMEM) with 10% fetal calf serum (FCS). TF1-derived cell lines were cultured in RPMI 1640 medium containing 10% FCS (R10) supplemented with 5 ng/mL human interleukin-3 (Miltenyi

Biotec). Molm-14, HEL and HL-60 were cultured in R10. Human CD34+ cells were grown in StemSpan SFEM II medium supplemented with StemSpan CC100 cytokine cocktail

(StemCell Technologies). Primary AML cells were grown in same conditions, with the addition of GM-CSF(20ng/ml). Cell lines were obtained from American type culture

Collection (ATCC) and authenticated. Plasmid details are available in Supplementary methods.

SILAC-based Mass Spectrometry

TF-1 was cultured in "light" SILAC medium containing normal lysine and arginine amino acids, while the TF1-hPRL3 was grown in "heavy" SILAC medium with stable isotope-

4

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

13 13 15 labeled C6 lysine (+6-Da shift) and C6 N4 arginine (+10-Da shift) (Thermo Scientific).

The cellular lysates were combined and proteolytically digested by trypsin followed by tandem MS identification. Peptides were subjected to target-decoy database search strategy with a false discovery rate (FDR) of less than 1% (<1%). Differential protein expression was quantified from the relative intensity ratios in the MS spectra between the "heavy" and "light" states.

Xenograft Mode Models

Six-week-old female NOD/SCID mice were provided by Dr Chan Shing-Leng (CSI

Singapore, NUS). Exponentially growing TF1-pEGFP and TF1-hPRL3 cells (5×106) were subcutaneously injected into loose skin between the shoulder blades and the left and right front leg of NOD/SCID recipient mice (3 mice total), respectively. The length (L) and width

(W) of the tumor were measured with callipers, and tumor volume (TV) was calculated as TV

= (L×W2)/2. The protocol is reviewed and approved by Institutional Animal Care and Use

Committee in compliance to the guidelines on the care and use of animals for scientific purpose.

Transfection and lentiviral-mediated shRNA delivery High-titre lentiviruses were produced in HEK293T cells by transfection using JetPrime

(Polyplus) with packaging vectors psPAX2 and pMD2.G together with the respective shRNAs constructs. Virus was harvested, pooled and concentrated by centrifugation

(Amicon). Infection of leukemia cells was done by the spin-infection method.

RNA Extraction and qRT-PCR

Detailed protocol is available in Supplementary Materials and Methods.

5

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Western Blotting

Cells were counted and lysed in RIPA buffer. Anti-PRL3 (clone 318) was a kind gift from Dr.

Zeng Qi; anti-Leo1, anti-Paf1 and anti-Ctr9 antibodies were from Bethyl Labs; anti-actin, anti-hnRNPE1, anti-HSP90, anti-JMJD2C, anti-GFP and anti-c-Myc (9E10) antibodies were from Santa Cruz; anti-Stathmin, anti-HDAC2 and anti-H3, anti-H3K9me2, anti-H3K27me2, anti-H3K27me3, anti-H3K4me1, anti-H3K4me2, anti-H3K4me3 and anti-H3K79me2 antibodies were from Cell Signaling; anti-H3K9me3 antibody was from Active Motif.

Chromatin immunoprecipitation (ChIP)

ChIP assays were performed using the respective antibodies according to manufacturer’s instructions (Thermo Scientific). qPCR was performed using the eluted DNA (sample) and

1% input, with primers spanning Leo1 promoter region with respect to Leo1 transcriptional start site. The % input was calculated as described previously (15). Primer sequences used for

ChIP-qPCR are available in Supplementary Table S3.

Luciferase assay Detailed protocol is available in Supplementary Materials and Methods.

Cell proliferation, Apoptosis and CFA Detailed protocol is available in Supplementary Materials and Methods.

Results

PRL-3 promotes cytokine-independent growth, colony forming capacity of AML cells in vitro and tumorigenecity in vivo

To assess the roles of PRL-3 in pathogenesis of AML, we developed a pair of stable, isogenic cell lines, TF1-pEGFP and TF1-hPRL3 by transfecting pEGFP (vector control) and pEGFP-

6

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

hPRL-3 vectors into TF-1 cells, respectively, followed by G418 selection and FACS sorting

(Figure 1A). TF-1 is a cytokine-dependent AML cell line. qRT-PCR and Western blot validated the overexpression of PRL-3 on both mRNA and protein levels in the TF1-hPRL3 cells relative to TF1-pEGFP cells (Figure 1B). In the absence of cytokine (human IL-3), the majority of TF1-pEGFP cells became apoptotic after 72 h. In contrast, most of TF1-hPRL3 cells were viable (Figure 1C). TF1-hPRL3 cells not only resisted cytokine deprivation- induced apoptosis, but also proliferated well without additional cytokines (Figure 1C).

Furthermore, methylcellulose assay showed only TF1-hPRL3 cells, but not TF1-pEGFP cells, formed colonies (Figure 1D). To determine the tumorigenic role of PRL-3 in vivo, we used the pair of TF1 cells in a subcutaneous mouse xenograft model. Strikingly, only TF1-hPRL3 cells, but not TF1-pEGFP cells, formed tumor mass in immunodeficient mice (Figure 1E).

The average TF1-hPRL3 tumor volume was 813 mm3 at 4 weeks after cell inoculation

(Figure 1E). Overall, these data suggest that PRL-3 confers important oncogenic function in

AML cells in vitro and in vivo.

SILAC quantitative proteomic profiling of PRL-3 overexpressed cell

To understand downstream pathways induced by overexpressing PRL-3, we performed a quantitative proteomic study by combining metabolic labeling using stable isotope labeling by amino acids in cell culture (SILAC) and high-resolution mass spectrometry (MS) approach (Figure S1A). We identified 398 proteins whose expression was significantly perturbed by PRL-3 overexpression, with 331 proteins upregulated and 67 proteins downregulated (>1.5-fold) (Figure S1B).

To gain a functional understanding of the dataset, we subjected the differentially expressed proteins (up- or down-regulated) to (GO) analysis. Upregulated proteins are

7

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

enriched for processes such as nucleic acid metabolism (19.7%), protein metabolism (15.1%) and pre-mRNA processing (5.7%), which are novel biological processes associated with

PRL-3, as well as known involvement in cell-cycle regulation (7.1%) and cell motility (6%) as depicted in Figure S1C. Amongst the differentially regulated proteins, 45.5% of them are nuclear proteins while 54.5% are cytoplasmic proteins (Figure S1D).

Validation of MS results

Expression of several representative proteins, including Leo1, stathmin, HSP-90, hnRNPE1 and HDAC2 were compared between TF1-pEGFP and TF1-hPRL3 cells (Figure 2A), and concordance was observed between the Western blot and MS results. Unexpectedly, we did not detect PRL-3 in our LC-MS/MS result. This is likely due to the trypsin cleavage sites distribution in the PRL-3 that generates peptides either too long or too short for detection by

LC-MS/MS. Nonetheless, PRL-3 overexpression was confirmed at both the mRNA (data not shown) and protein levels by Western blot (Figure 2A).

We further performed lentiviral-based delivery of PRL-3 shRNA into two additional human

AML cells, Molm-14 and HEL, which expresses high endogenous level of PRL-3. The knockdown of PRL-3 reduced the levels of Leo1, stathmin, HSP90 and hnRNPE1 (Figure

2B). An exception was HDAC2, where although MS identifies it as one of the upregulated proteins by PRL-3, western blot was not sensitive enough to detect the differences (Figure

2B). These experiments confirmed the general validity of our proteomic analysis .

Leo1 is upregulated by PRL-3

Leo1, a component of the human polymerase II-associating factor 1 (PAF) complex, was the top candidate from the SILAC LC-MS/MS analysis with the most significant increase in

8

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

protein abundance. We therefore decided to focus on this protein in our subsequent experiments. To confirm that Leo1 is a downstream target of PRL-3, we introduced a Tet-On myc-tagged PRL-3 construct into 293T cells. 48 hours after transfection, 293T cells were exposed to increasing concentrations of the tetracycline (Tc) derivative, doxycycline (Dox) to induce PRL-3 expression. PRL-3 protein accumulated in a dose-dependent manner, and a corresponding increase in Leo1 protein levels was observed (Figure 2C) and quantified

(Figure S2). To complement the overexpression studies, we also performed knockdown by transfecting PRL-3 siRNA into 293T cells. The degree of PRL-3 inhibition correlated to the dose of siRNA added, and Leo1 was downregulated (Figure 2D). Collectively, these observations corroborated the MS results showing that Leo1 expression is positively correlated with PRL-3 levels.

Abrogation of Leo1 reduces PRL3-mediated proliferation and cytokine independence of

AML

To determine whether Leo1 contributes to PRL3-induced leukemic growth, we used lentiviral-based delivery of shRNAs to deplete Leo1 expression and looked at growth properties. qRT-PCR and immunoblotting analysis demonstrated a 5-fold reduction in Leo1 mRNA levels and a marked reduction in the Leo1 protein levels upon treatment with Leo1 shRNA (Figure 3A). Notably, reduction in Leo1 levels significantly impaired growth of TF1- hPRL3 cells to growth rates comparable with TF-1 parental cells (Figure 3B).

To determine whether loss of Leo1 affects the survival of TF1-hPRL3, we plated the cells in the absence of human IL-3 for 72 hours, followed by flow cytometric analysis for apoptotic markers annexin V-PE and 7-AAD. Although ~90% of TF1-hPRL3 population was able to survive without supplementation of cytokine, Leo1 knockdown notably increased the

9

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

apoptosis of TF1-hPRL3 induced by cytokine withdrawal (Figure 3C), indicating that Leo1 abrogation removes the protective effect of PRL-3 towards cytokine deprivation. Leo1 knockdown in PRL-3 cells resulted in significantly fewer colonies, indicating a loss of clonogenic survival of these cells (Figure 3D). These findings collectively indicate that Leo1 is an important mediator of the cell growth and survival of the TF1-hPRL3 cells, and loss of

Leo1 is largely sufficient to reverse these properties conferred by PRL-3 overexpression.

Leo1 and PRL-3 levels are significantly associated in AML patient samples

Since we observed a strong association between Leo1 and PRL-3 levels in cell lines, we determined whether Leo1 is similarly upregulated in human primary AML cells. Western blot analysis of PRL-3 and Leo1 protein expression in an independent cohort of 24 primary AML samples found that 50% of the patient samples (Figure 4A) were positive for PRL-3 protein.

Of these, only one did not express Leo1. Therefore a strong association exists between PRL-3 and Leo1 expression (Chi-square p-value <0.01). Leo1 was also expressed in 5 samples with no PRL-3 expression, suggesting that other mechanisms may be responsible in these cases.

Our findings demonstrated that a significant proportion (50%) of AML cases appeared to co- express PRL-3 and Leo1, suggesting that Leo1 is a likely downstream target of PRL-3.

We also examined the basal expression levels of human PRL-3 and Leo1 in normal hematopoietic cell fractions, namely CD34+, T cell, B cell and myeloid cell. Similarly, we found a relative correlation between PRL-3 and Leo1 levels. Furthermore, we observed that

PRL-3 and Leo1 is expressed at higher levels in CD34+ and myeloid cells (Figure S3).

To validate the functional relevance of PRL-3 in human AML, we downregulate PRL-3 in primary AML cells(Figure 4B). PRL-3 siRNA inhibited AML cell growth in a proliferation

10

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

assay as compared to transfection with control siRNA (Figure 4C). This result suggest that

PRL-3 expression is also of biological relevance in human AML and suggest that specific targeting of PRL-3 may have possible therapeutic applications in AML.

PRL-3 affects Leo1 mRNA levels through histone H3 lysine 9 (H3K9me3) demethylation

To examine whether PRL-3 regulates Leo1 at the transcriptional level, we perturbed PRL-3 with different shRNA constructs, which was accompanied by a concomitant reduction of

Leo1 transcripts levels (Figure 5A). To confirm that PRL-3 regulates Leo1 at the transcriptional level, we established a Leo1 reporter construct containing a 703bp Leo1 promoter fragment cloned upstream of the firefly luciferase gene (pGL3-Leo1). Transfection of the reporter construct, together with vector expressing wild-type PRL-3 increased the luciferase activity in a dose-dependent manner (Figure 5B).

Given that PRL-3 is not a DNA-binding protein, the transcriptional involvement of PRL-3 is likely to be indirect. A recent report implicated an epigenetic role of PRL-3 in colorectal cancer through regulating histone modifying enzymes involved in histone demethylation (16).

To explore the possibility that PRL-3 activates Leo1 through histone methylation status in leukemic cells, we performed an unbiased screen of eight different permissive and repressive histone modifications in the TF-1 and TF1-hPRL3 cells. We found that PRL-3 overexpression specifically leads to a global decrease in di- and tri-methylation at histone H3 lysine 9

(H3K9me2 and 3) residues (Figure 5C). We further confirmed this observation in another

AML cell line, Molm-14 (Figure S4).

To determine whether increased Leo1 expression is associated with local reductions in repressive histone methylation, we compared the basal levels of H3K9me2, H3K9me3 and

11

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

H3K27me3 across Leo1 promoter region in the TF-1 and TF1-hPRL3 cells. In TF1-hPRL3 cells, a marked reduction in H3K9me3, and to a lesser extend H3K9me2, was detected throughout the entire promoter region (P1-P4) as compared to the TF-1 cells (Figure 5D). In contrast, H3K27me3 levels were very low on the promoter (Figure S5). Thus, Leo1 promoter seems to be predominantly modulated by the methylation levels of H3K9.

PRL-3 mediates H3K9me3 demethylation on Leo1 promoter through JMJD2C histone demethylase

Next, we sought to identify which histone demethylase is involved in mediating this process.

The members of the JMJD2 family, including the JMJD2A, JMJD2B, JMJD2C and JMJD2D, are known to exhibit specific lysine-trimethyl demethylation activities (17). We systematically knockdown these candidates and examine the impact on H3K9me3 levels and

Leo1 expression. The JMJD2 shRNAs exhibited high specificity (Figure 6A). We observed that JMJD2C depletion in TF1-hPRL3 cells was able to increase the global levels of

H3K9me3, accompanied by a reduction in Leo1 (Figure 6B), implying the specific involvement of JMJD2C in regulating H3K9me3 related to levels of Leo1.

To demonstrate that JMJD2C directly regulates H3K9me3 on the Leo1 promoter, we examined the levels of H3K9me3 on region P2 on Leo1 promoter. Accordingly, knockdown of JMJD2C reduced the JMJD2C signal on the Leo1 promoter (Figure 6C) and led to a 40% augmentation of the H3K9me3 signal (Figure 6D). However, knockdown of JMJD2A,

JMJD2B and JMJD2D did not modulate H3K9me3 levels on Leo1 promoter (Figure S6A).

Next, we examined how PRL-3 phosphatase regulates JMJD2C histone demethylase.

JMJD2C protein expression did not increase in TF1-hPRL3 cells (Figure S6B). We also examined the gene expression levels of JMJD2A, JMJD2B. JMJD2C and JMJD2D in TF1

12

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

and TF1-PRL3 cells, and found no significant changes between the isogenic cell lines (Figure

S6C). Furthermore, PRL-3 and JMJD2C did not physically interact (Figure S6D). However, an increase in JMJD2C occupancy occurred on the promoter of Leo1 in the TF1-hPRL3 as compared to TF-1 cells (Figure 6E). However, JMJD2C association to the P2 region was significantly abrogated in the PRL-3 knockdown cells, supporting the notion that JMJD2C binding to the Leo1 promoter region is dependent on the presence of the PRL-3 protein

(Figure S6E).

Taken together, these results demonstrate that PRL-3 relieves the repressive H3K9me3 mark on Leo1 promoter through JMJD2C, resulting in a more transcriptionally permissive chromatin state leading to the efficient promotion of Leo1 expression.

Leo1 suppression depletes the levels of PAF complex components and affects the expression of target

The PAF complex subunits have been shown to co-immunoprecipitate with each other.

Therefore, we checked whether knockdown of Leo1 affects the interaction network of the

PAF complex. Interestingly, we found that loss of Leo1 destabilized the PAF complex, and the mRNA levels of Paf1, Ctr9 and Ski8 were collectively downregulated (Figure 7A). In addition, the protein levels of Paf1 and Ctr9 were also decreased with Leo1 depletion (Figure

7B). To determine the functional relevance of Leo1-mediated destabilization of the PAF complex, we further examined the effects on several reported PAF complex target genes. A subset of these genes, namely Sox2, Sox4 and Tbx3 were significantly downregulated when

Leo1 was silenced in Molm-14 cells (Figure 7C).

To determine whether Leo1 plays an independent role in mediating the oncogenic properties

13

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

in AML or through PAF complex as a whole, we compared the effects of Leo1, Paf1 or Ctr9 knockdown in TF1-PRL3 cells. We confirmed the knockdown efficiency using Western Blot

(Figure S7A). Interestingly, we found that knockdown of Leo1 impedes cell proliferation to a greater extent than Paf1 or Ctr9 knockdown (Figure S7B). In addition, Sox2 and Sox4 expression levels were similarly affected in Leo1, Paf1 of Ctr9 knockdown (Figure S7C).

These observations suggest that the entire PAF complex is partly involved in mediating oncogenesis though SOX genes, but other mechanisms involving Leo1 alone is present.

Furthermore, when we analyzed the expression levels of Leo1, Paf1, Cdc73, Ski8 and Ctr9 in

TF1-PRL3 cells, only Leo1 showed an upregulation (Figure S7D).

Since we showed that knockdown of PRL-3 downregulated levels of Leo1, we further examined whether knockdown of PRL-3 similarly destabilizes the PAF complex.

Interestingly, silencing PRL-3 downregulated all components of the PAF complex (Figure

7D), and subsequent restoration of Leo1 levels rescued the levels of Paf1, Ski8 and Ctr9

(Figure 7E). These results further confirm that Leo1 is functionally relevant and is an important downstream mediator of PRL-3 oncogenic functions.

Discussion

Our present work represents the first large-scale quantitative survey of proteins regulated by

PRL-3 in leukemia. We identified several known proteins downstream of PRL-3 including

Stathmin, Nucleolin and Translationally Controlled Tumor Protein (TCTP) (6, 18, 19), which endorsed the validity of our approach. We also discovered additional upregulated candidate oncogenes that were linked to carcinogenesis, such as hnRNPH1, TPI1, HSP90B1, Prohibitin, as well as candidate tumor suppressors that were downregulated, such as POTE, nucleophosmin and BTF3, which were previously not known to be modulated by PRL-3

14

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

(Table S1). On the other hand, some reported substrates of PRL-3 in solid tumor cell lines, such as CDH22, Ezrin, Keratin8, Integrin-1 and EF-2, were not identified in our screen suggesting that PRL-3 might activate both common and distinct networks in solid tumor as compared to leukemia.

From our comprehensive proteomic dataset, we gained several novel insights into the functions of PRL-3. First, we found that PRL-3 unexpectedly alters the expression levels of a significant number of nuclear proteins (50%), although it was reported to localize predominantly in the plasma membrane and cytosol due to the CAAX membrane-targeting signal (20). Despite this, PRL-3 was also found to be able to shuttle between the nucleus and cytoplasm in myeloma and AML cells (21), by an unclear mechanism. Second, gene ontology annotation by biological process reveals a specific involvement of PRL-3 in pre-mRNA splicing and nucleic acid metabolism, as well as cell-cycle regulation and cell motility.

Involvement in cell motility is consistent with published reports that over-expression of PRL-

3 in carcinomas have been linked to processes such as enhanced cell migration, invasion and metastasis (20, 22). Notably, this is the first study to suggest that PRL-3 plays a role in the regulation of RNA-related processes. Overall, our unbiased approach enables us to implicate novel candidate proteins in the oncogenic function of PRL-3, which would fundamentally advance our understanding of how this phosphatase contributes to cancer progression.

In the recent years, several pivotal articles have proposed functions of the PAF complex in development and cancer, and the PAF complex was found to be substrates of another oncogenic phosphatase, SHP2 (23-26). In addition, the PAF complex is essential for leukemogenesis by MLL fusion proteins through inducing the expression of Hox genes, thus maintaining a haematopoietic stem cell (HSC)-like signature and resistance to differentiation

15

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

(15). These led us to focus on the involvement of Leo1 and the PAF complex as the downstream mediators of PRL-3 activities. Although characterization of the individual subunits of the PAF complex revealed opposing roles in cancer (24), our current work suggests that Leo1 is a proto-oncogene in leukemia, and the levels of Ski8, Ctr9 and Paf1 subunits depended on Leo1 levels (Figure 7A and 7B), supporting the notion of an interdependency within the PAF complex.

One important hallmark of AML is the acquisition of self-renewal capacity in the leukemic blast cells (27) through the disruption of self-renewal pathways such as Hox and Wnt/- catenin. Interestingly, the PAF complex has been found to augment the expression of HOXA9

(15), and in another independent report, Leo1 has been found to associate directly with - catenin (28). In addition, the PAF complex has also been reported to regulate directly key pluripotency genes in ESCs. Indeed, when we knockdown Leo1, several pluripotency genes such as Sox2, Sox4 and Tbx3 were downregulated (Figure 7C). The deregulation of Sox genes, especially Sox4, has been shown to be an important collaborating driver of myeloid leukemogenesis in several transgenic murine models of leukemia (29). Thus, our study suggests that PRL-3 could exert oncogenic properties in AML through Leo1 and PAF complex-mediated deregulation of critical pluripotency genes previously implicated in leukemogenesis.

There were few reports on how PRL-3 regulates its targets, in part due to the challenges in identifying the genuine targets of PRL-3. In our study, we revealed a novel mechanism of the transcriptional involvement of PRL-3 in regulating Leo1. Trimethylation on H3K9 is a strong repressive epigenetic mark generally associated with heterochromatin maintenance and transcriptional repression, and deregulation of H3K9me3 levels has been shown to cause

16

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

aberrant gene expression, contributing to the development of cancer (30, 31). We found global demethylation of H3K9me3 in TF1-hPRL3 cells, suggesting that alterations in a histone-modifying enzyme lead to these widespread epigenetic changes (32). JMJD2C, originally identified as GASC1, is a gene located on 9p, and is frequently amplified in cancers (30). Our results identify JMJD2C as the main enzyme involved in the demethylation of H3K9me3 on the Leo1 promoter in TF1-hPRL3 cells, thereby enhancing the expression of Leo1. Interestingly, we noticed an increase in JMJD2C occupancy on the promoter region of Leo1, and this was dependent on PRL-3 expression levels (Figure 6E).

How PRL-3 affects the binding of JMJD2C or possibly other histone demethylase on the promoters of target genes remains to be determined. Importantly, our results demonstrated a possible interface between the activities of a phosphatase and a histone demethylase in gene regulation.

In conclusion, our study unravels downstream signaling pathways of PRL-3, identifying Leo1 as an important downstream target. Taken together, this study provides novel insights into the mechanisms by which PRL-3 promotes the malignant phenotype in AML.

Acknowledgments

WJC is supported by NMRC Clinician Scientist Investigator award. This work is partly supported by Singapore Cancer Syndicate Grant. This research is supported by the National

Research Foundation Singapore and the Singapore Ministry of Education under the Research

Centers of Excellence initiative. J.Z. Is also supported by a NMRC Clinician-Scientist IRG

Grant CNIG11nov38.

We thank Prof Daniel G. Tenen, Prof H. Phillip Koeffler and A/Prof Motomi Osato for helpful

17

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

suggestions.

18

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

References

1. Zeng Q, Si X, Horstmann H, Xu Y, Hong W, Pallen CJ. Prenylation-dependent association of protein-tyrosine phosphatases PRL-1, -2, and -3 with the plasma membrane and the early endosome. J Biol Chem. 2000;275(28):21444-21452.

2. Al-Aidaroos AQ, Zeng Q. PRL-3 phosphatase and cancer metastasis. J Cell Biochem.

2010;111(5):1087-1098.

3. Kato H, Semba S, Miskad UA, Seo Y, Kasuga M, Yokozaki H. High expression of

PRL-3 promotes cancer cell motility and liver metastasis in human colorectal cancer: a predictive molecular marker of metachronous liver and lung metastases. Clin Cancer Res.

2004;10(21):7318-7328.

4. Polato F, Codegoni A, Fruscio R, Perego P, Mangioni C, Saha S, et al. PRL-3 phosphatase is implicated in ovarian cancer growth. Clin Cancer Res. 2005;11(19 Pt 1):6835-

6839.

5. Radke I, Gotte M, Kersting C, Mattsson B, Kiesel L, Wulfing P. Expression and prognostic impact of the protein tyrosine phosphatases PRL-1, PRL-2, and PRL-3 in breast cancer. Br J Cancer. 2006;95(3):347-354.

6. Chu ZH, Liu L, Zheng CX, Lai W, Li SF, Wu H, et al. Proteomic analysis identifies translationally controlled tumor protein as a mediator of phosphatase of regenerating liver-3- promoted proliferation, migration and invasion in human colon cancer cells. Chin Med J

(Engl). 2011;124(22):3778-3785.

7. Jiang Y, Liu XQ, Rajput A, Geng L, Ongchin M, Zeng Q, et al. Phosphatase PRL-3 is a direct regulatory target of TGFbeta in colon cancer metastasis. Cancer Res. 2011;71(1):234-

244.

8. Saha S, Bardelli A, Buckhaults P, Velculescu VE, Rago C, Croix BS, et al. A phosphatase associated with metastasis of colorectal cancer. Science. 2001;294(5545):1343-

19

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1346.

9. Bardelli A, Saha S, Sager JA, Romans KE, Xin B, Markowitz SD, et al. PRL-3 expression in metastatic cancers. Clin Cancer Res. 2003;9(15):5607-5615.

10. Wang H, Vardy LA, Tan CP, Loo JM, Guo K, Li J, et al. PCBP1 suppresses the translation of metastasis-associated PRL-3 phosphatase. Cancer Cell. 2010;18(1):52-62.

11. Bessette DC, Qiu D, Pallen CJ. PRL PTPs: mediators and markers of cancer progression. Cancer Metastasis Rev. 2008;27(2):231-252.

12. Guo K, Li J, Tang JP, Tan CBP, Hong CW, Al-Aidaroos AQ, et al. Targeting intracellular oncoproteins with antibody therapy or vaccination. Sci Transl Med.

2011;3(99):99ra85.

13. Zhou J, Bi C, Chng WJ, Cheong LL, Liu SC, Mahara S, et al. PRL-3, a metastasis associated tyrosine phosphatase, is involved in FLT3-ITD signaling and implicated in anti-

AML therapy. PLoS One. 2011;6(5):e19798.

14. Beekman R, Valkhof M, Erkeland SJ, Taskesen E, Rockova V, Peeters JK, et al.

Retroviral integration mutagenesis in mice and comparative analysis in human AML identify reduced PTP4A3 expression as a prognostic indicator. PLoS One. 2011;6(10):e26537.

15. Muntean AG, Tan J, Sitwala K, Huang Y, Bronstein J, Connelly JA, et al. The PAF complex synergizes with MLL fusion proteins at HOX loci to promote leukemogenesis.

Cancer Cell. 2010;17(6):609-621.

16. Liu Y, Zheng P, Liu Y, Ji T, Liu X, Yao S, et al. An epigenetic role for PRL-3 as a regulator of H3K9 methylation in colorectal cancer. Gut. 2013;62(4):571-581.

17. Shin S, Janknecht R. Diversity within the JMJD2 histone demethylase family.

Biochem Biophys Res Commun. 2007;353(4):973-977.

18. Semba S, Mizuuchi E, Yokozaki H. Requirement of phosphatase of regenerating liver-

3 for the nucleolar localization of nucleolin during the progression of colorectal carcinoma.

20

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Cancer Sci. 2010;101(10):2254-2261.

19. Zheng P, Liu YX, Chen L, Liu XH, Xiao ZQ, Zhao L, et al. Stathmin, a new target of

PRL-3 identified by proteomic methods, plays a key role in progression and metastasis of colorectal cancer. J Proteome Res. 2010;9(10):4897-4905.

20. Zeng Q, Dong JM, Guo K, Li J, Tan HX, Koh V, et al. PRL-3 and PRL-1 promote cell migration, invasion, and metastasis. Cancer Res. 2003;63(11):2716-2722.

21. Fagerli UM, Holt RU, Holien T, Vaatsveen TK, Zhan F, Egeberg KW, et al.

Overexpression and involvement in migration by the metastasis-associated phosphatase PRL-

3 in human myeloma cells. Blood. 2008;111(2):806-815.

22. Fiordalisi JJ, Keller PJ, Cox AD. PRL tyrosine phosphatases regulate rho family

GTPases to promote invasion and motility. Cancer Res. 2006;66(6):3153-3161.

23. Squazzo SL, Costa PJ, Lindstrom DL, Kumer KE, Simic R, Jennings JL, et al. The

Paf1 complex physically and functionally associates with transcription elongation factors in vivo. EMBO J. 2002;21(7):1764-1774.

24. Chaudhary K, Deb S, Moniaux N, Ponnusamy MP, Batra SK. Human RNA polymerase II-associated factor complex: dysregulation in cancer. Oncogene.

2007;26(54):7499-7507.

25. Wang P, Bowl MR, Bender S, Peng J, Farber L, Chen J, et al. Parafibromin, a component of the human PAF complex, regulates growth factors and is required for embryonic development and survival in adult mice. Mol Cell Biol. 2008;28(9):2930-2940.

26. Takahashi A, Tsutsumi R, Kikuchi I, Obuse C, Saito Y, Seidi A, et al. SHP2 tyrosine phosphatase converts parafibromin/Cdc73 from a tumor suppressor to an oncogenic driver.

Mol Cell. 2011;43(1):45-56.

27. Tenen DG. Disruption of differentiation in human cancer: AML shows the way. Nat

Rev Cancer. 2003;3(2):89-101.

21

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

28. Mosimann C, Hausmann G, Basler K. Parafibromin/Hyrax activates Wnt/Wg target gene transcription by direct association with beta-catenin/Armadillo. Cell. 2006;125(2):327-

341.

29. Sandoval S, Kraus C, Cho EC, Cho M, Bies J, Manara E, et al. Sox4 cooperates with

CREB in myeloid transformation. Blood. 2012;120(1):155-165.

30. Cloos PA, Christensen J, Agger K, Maiolica A, Rappsilber J, Antal T, et al. The putative oncogene GASC1 demethylates tri- and dimethylated lysine 9 on histone H3. Nature.

2006;442(7100):307-311.

31. Bannister AJ, Kouzarides T. Regulation of chromatin by histone modifications. Cell

Res. 2011;21(3):381-395.

32. Fullgrabe J, Kavanagh E, Joseph B. Histone onco-modifications. Oncogene.

2011;30(31):3391-3403.

22

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure Legends Figure 1. Establishment of stable PRL-3 cell line. (A) Images of TF1-pEGFP and TF1- hPRL3 cells in bright field and GFP channel under an invert microscopy. (B) qRT-PCR and

Western blot analysis of PRL-3 mRNA and protein expression in TF1-pEGFP and TF1- hPRL3 cells. -actin was used as a loading control in the protein analysis. (C) After IL-3 starvation, TF1-pEGFP and TF1-hPRL3 cells were stained and assessed with Trypan Blue

Exclusion methods on a hemacytometer. The percentages of viable cells at 72 hours were calculated relative to before IL-3 withdrawal (0 hour). To construct the cell growth curve, 20 x 105 TF1-hPRL3 cell were initially seeded in standard medium without additional human cytokines. Cells were counted and reseeded in fresh medium every two days for up to 6 days.

In both figures, three independent replicates were conducted (mean ± SD). (D) Colony forming assay (CFA) of TF1-pEGFP and TF1-hPRL3. The experiments were duplicated and representative pictures are presented. (E) Mouse xenograft models of TF1-pEGFP and TF1- hPRL3. Five million TF1-pEGFP and TF1-hPRL3 cells were subcutaneously injected into loose skin between the shoulder blades and the left and right front leg of NOD/SCID recipient mice, respectively. At 4 weeks after injection, tumors were harvested and measurement of tumor volume was taken. The tumor volume is shown as the mean ± SD

(mm3). Arrows indicated the TF1-hPRL3 tumor mass at right and no tumor at left (TF1- pEGFP cell injected site) in one mouse. **p<0.005.

Figure 2. Western blot validation of candidate proteins identified by MS. (A) Expression of several representative up-regulated proteins is compared between TF-1 and TF1-hPRL3.

(B) Knockdown of endogenous PRL-3 in AML cell lines Molm-14 and HEL. The same panel of proteins is examined by western blot. (C) 293T cells were stably transfected with an inducible Tet-on myc-tagged PRL-3 construct, and increasing concentration of doxycyclin was used to induce PRL-3 expression. (D) 293T cells were transfected with specified doses

23

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

of PRL-3 siRNA for 48 h before analysis with Western blot.

Figure 3. Effects of stable Leo1 knockdown in TF1-hPRL3 cells. (A) Knockdown of Leo1 in TF1-hPRL3 cells were analysed with qRT-PCR and Western blot. -actin was used as loading control. (B) Growth curve was determined by CellTitre-Glo luminescent assay. Cells were plated in 96-well format and grown for up to 4 days. Experiments were performed in triplicates. (C) TF-1, PRL3-scrambled and PRL3-Leo1 shRNA cells were seeded in medium without human IL-3 for 48 h. The graph is plotted with the percentages of apoptotic cells defined by Annexin V and 7-AAD positivity. (D) TF-1, PRL3-scrambled and PRL3-Leo1 shRNA cells were evenly suspended in HSC-CFU medium (StemMACS) and incubated for

10 days for colonies to develop. **p<0.005.

Figure 4. Leo1 and PRL-3 expression in human primary AML cells. (A) Western blot was performed to show the expression levels of Leo1 and PRL-3 in a cohort of 24 primary AML samples. -actin was used as loading control. Chi-square test indicates highly significant association (P<0.01). (B) Analysis of PRL-3 mRNA expression in primary AML cells transfected with either control siRNA or PRL-3 siRNA for 24 h. (C) Cell proliferation assay of primary AML cells transfected with either control siRNA or PRL-3 siRNA for 3 days.

**p<0.01

Figure 5. PRL-3 regulates Leo1 at the transcriptional level. (A) Molm-14 cells were transfected with PRL-3 shRNA for 24 h, and Leo1 and PRL-3 mRNA levels were measured.

Data represent the mean ± SEM of triplicates. (B) 293T cells were transfected with the respective luciferase reporter and expression vectors, and relative luciferase activity

(Firefly/Renilla) was determined. Data represent the mean ± SEM of triplicates. (C) Nuclear

24

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

extracts were obtained from TF1 or TF1-hPRL3 cells, and immunoblot for respective histone marks. Histone H3 was used as the loading control. Intensity of bands was measured using

ImageJ and provided as a fold-increase using TF-1 as control. (D) Diagram of Leo1 promoter showing the position of four different primer pairs (P1-P4) used for qPCR. ChIP was performed in TF1 and TF1-hPRL3 cells with respective antibodies. Solid lines indicate ChIPs with the indicated antibodies, while dotted lines indicate control rabbit IgG; TF1 cells are in black and TF1-hPRL3 cells in grey. Data are plotted as % Input. Data represent the mean ±

SEM of triplicates. *p<0.05, **p<0.005.

Figure 6. JMJD2C regulates H3K9me3 on the Leo1 promoter. (A) shRNA specificity was tested in TF-1 cells using JMJD2A, 2B, 2C and 2D primers. Graph is plotted relative to scrambled (Mock). Data represent the mean ± SEM of triplicates. (B) TF1-hPRL3 transfected with Mock or JMJD2 shRNA and analyzed by Western Blot using the indicated antibodies.

(C-D) ChIP was performed using antibody against JMJD2C (C) or H3K9me3 (D) on Leo1 promoter P2. (E) ChIP was performed in TF-1 and TF1-hPRL3 cells using JMJD2C antibody on Leo1 promoter. Data shown as % Input. Data represent the mean ± SEM of triplicates.

*p<0.05, **p<0.005.

Figure 7. Leo1 knockdown destabilizes the PAF complex. (A-B) Molm-14 cells were transfected with either Leo1 shRNA or control shRNA (Emp), and qRT-PCR (A) or Western blot (B) was done for the expression of the PAF complex components. Data represent the mean ± SEM of triplicates. (C) qRT-PCR analyses of Sox2, Sox4 and Tbx3 after Leo1 knockdown in Molm-14 cells. Data represent the mean ± SEM of triplicates. (D) Molm-14 was transfected with PRL-3 shRNA and analyzed for PAF components. Data represent the mean ± SEM of triplicates. (E) Molm-14 was co-transfected with PRL-3 shRNA and Leo1

25

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

expression construct and analyzed for PAF components. Data represent the mean ± SEM of

triplicates. *p<0.05, **p<0.005.

26

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 31, 2014; DOI: 10.1158/0008-5472.CAN-13-2321 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

LEO1 is regulated by PRL-3 and mediates its oncogenic properties in Acute Myeloid Leukemia

Phyllis SY Chong, Jianbiao Zhou, Lip Lee Cheong, et al.

Cancer Res Published OnlineFirst March 31, 2014.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-13-2321

Supplementary Access the most recent supplemental material at: Material http://cancerres.aacrjournals.org/content/suppl/2014/04/01/0008-5472.CAN-13-2321.DC1

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet been Manuscript edited.

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/early/2014/03/29/0008-5472.CAN-13-2321. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on October 1, 2021. © 2014 American Association for Cancer Research.