Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Title: Alternative in triple-negative breast tumors allows NRAS and c-JUN to bypass PUMILIO post-transcriptional regulation

Running Title: Alternative polyadenylation in triple-negative breast cancer

Authors: Wayne O. Miles 1, 2, 7, Antonio Lembo 3, 4, Angela Volorio 5, Elena Brachtel 5, Bin Tian 6, Dennis Sgroi 5, Paolo Provero 3, 4 and Nicholas J. Dyson 1, 7

1 Department of Molecular Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, 149 13th Street, Charlestown, MA, 02129, USA.

2 Department of Molecular Genetics, The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210

3 Department of Molecular Biotechnology and Health Sciences, University of Turin, Italy.

4 Center for Translational Genomics and Bioinformatics, San Raffaele Scientific Institute, Milan, Italy.

5 Pathology Department, Massachusetts General Hospital Cancer Center and Harvard Medical School, 149 13th Street, Charlestown, MA, 02129, USA.

6 Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA

7 Corresponding authors: [email protected] (N.D.) and [email protected] (W.O.M.) Conflict of interest

The authors have no conflict of interest.

Financial Support

1

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

D. Sgroi supported by NIH grant R01 CA112021, the Avon Foundation, DOD Breast Cancer Research Program and the Susan G. Komen for the Cure. N.J. Dyson is supported by NIH grants; R01 CA163698 and R01 CA64402. N.J. Dyson is the James and Shirley Curvey MGH Research Scholar.

Abstract

Alternative polyadenylation (APA) is a process that changes the post-

transcriptional regulation and translation potential of mRNAs via addition or

deletion of 3'UTR sequences. To identify post-transcriptional regulatory events

affected by APA in breast tumors, tumor datasets were analyzed for recurrent

APA events. Motif mapping of the changed 3'UTR region found that APA-

mediated removal of Pumilio Regulatory Elements (PRE) was unusually common.

Breast tumor sub-type specific APA profiling identified triple-negative breast tumors as having the highest levels of APA. To determine the frequency of these events, an independent cohort of triple-negative breast tumors and normal breast tissue was analyzed for APA. APA-mediated shortening of NRAS and c-JUN was seen frequently, and this correlated with changes in expression of downstream targets. mRNA stability and Luciferase assays demonstrated APA-dependent

alterations in RNA and levels of affected candidate . Examination of

clinical parameters of these tumors found those with APA of NRAS and c-JUN to

be smaller and less proliferative, but more invasive than non-APA tumors. RT-

PCR profiling identified elevated levels of polyadenylation factor CSTF3 in tumors

with APA. Overexpression of CSTF3 was common in triple-negative breast cancer

cell lines, and elevated CSTF3 levels was sufficient to induce APA of NRAS and c-

JUN. Our results support the hypothesis that PRE-containing mRNAs are

2

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

disproportionately affected by APA, primarly due to high sequence similarity in the motifs utilized by polyadenylation machinery and the PUM complex.

3

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Introduction

Cancer genome sequencing has identified driver mutations associated with disease. In addition, non-genetic changes such as epigenetic reprogramming and aberrant post- transcriptional regulation also impact tumor phenotypes. Post-transcriptional controls that affect mRNA processing and stability, including microRNAs (miRNAs) and RNA- binding (RBPs) [1]. These factors bind to seed sequences within the 3’ untranslated region (UTR) of transcripts [2] and are commonly misregulated in cancers

[3]. Tumor specific fluctuations in the levels of RBPs and miRNAs may be particularly important, as transcripts from oncogenes and tumor-suppressor genes contain a dis- proportionately high number of post-transcriptional regulatory motifs.

In line with these idea, we recently discovered that the expression of the human oncogene E2F3 is post-transcriptionally controlled by the Pumilio complex [4]. The

Pumilio complex is comprised of two RNA-binding proteins in humans, Pumilio (PUM) and Nanos (NOS), which bind to a conserved motif of UGUAXAUA [5-7] and a GU-rich element within the 3’UTR of their substrates [2, 8]. The Pumilio repressor has evolutionarily conserved roles in the regulation of proliferation and differentiation [6-9].

PUM complexes function to inhibit translation [10] or induce the degradation of their substrates [4, 11-14]. The PUM repressor regulates the translation potential of hundreds of transcripts including a large number of oncogenes and tumor-suppressor genes [5].

While analyzing the role of PUM in modulating E2F3 levels in cancer cell lines, we found the 3’UTR of E2F3 transcripts were often shortened. This change in mRNA length, removed a number of regulatory elements, including the PRE motifs [4]. The

4

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

shortened E2F3 mRNA in these cells was polyadenylated, indicating that they were generated by alternative polyadenylation (APA). APA is a poorly understood developmental process that leads to the use of non-canonical polyadenylation sites [15-

17]. APA affects the inclusion/exclusion of regulatory elements and can alter mRNA stability and translation potential [18]. During normal development, APA is utilized to regulate differentiation [15, 16, 19-21]. In cultured human cells, APA has been linked with genes responsible for cell proliferation [22, 23].

APA is also a likely contributor to oncogenesis [24], as APA has been correlated with poor patient prognosis in a variety of malignancies [25-27]. Despite this, little is known about APA in tumor development. Our finding that the E2F3 mRNA is altered by APA in cancer cell lines, prompted us to investigate this phenomenon further. To do this we looked for mRNAs that are altered by APA in human breast tumor samples compared to normal tissue. This analysis found that APA is widespread in breast tumors and that these APA events disproportionately remove putative PUM-regulatory elements.

Breast tumor sub-type specific APA profiles were then generated and unexpectedly, this revealed that triple-negative breast tumors have a distinct APA-profile from other sub- types. Four candidate APA events were then selected and their frequency assayed in an independent cohort of triple-negative breast tumors. These results led us to focus on two recurrent APA-altered mRNAs, c-JUN and NRAS, for detailed analysis. These

genes were selected as they displayed high levels of APA in tumors. Here, we show that these mRNAs are targets of PUM-mediated repression and demonstrate that APA circumvents this regulation. Strikingly, both c-JUN and NRAS were known to be de- regulated in triple negative breast tumors [28, 29], but the underlying mechanism was

5

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

unclear. In addition, APA of NRAS and c-JUN altered the expression of downstream target genes. Intriguingly, unlike previous reports, APA in this tumor cohort correlated with smaller, slower growing but more invasive node-positive tumors. Collectively these findings indicated that APA of PUM-substrates is a common feature of triple-negative breast tumors. These results show that recurrent APA of PRE-containing mRNAs, allows transcripts to circumvent PUM-mediated repression, and provide a mechanistic explanation for the increased activity of NRAS and c-JUN in tumors.

Materials and Methods

Bioinformatics

We used the data of [30] ( Expression Omnibus [31] (GSE3744)) to compare 38 tumor samples to 7 normal breast samples. Analysis of differential APA between tumors and normal tissues was performed as described previously [26]. Briefly, the method exploits the 3' bias and probe-set organization of 3'IVT Affymetrix arrays to evaluate the relative expression of mRNA isoforms using alternative polyadenylation sites. APA sites were obtained from the Poly-A DB database.

Real Time Quantitative PCR (RT-PCR)

RNA was purified using the RNeasy Extraction Kit (Qiagen). Reverse Transcription was performed using Taq-Man Reverse Transcriptase and SYBR Green detection chemistry.

Cell culture, transfections and Luciferase expression constructs

6

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Cell lines were provided and authenticated by The Center of Molecular Therapeutics,

Massachusetts General Hospital (July 2014). c-JUN plasmids were kind gifts from Dr.

Lily Vardimon [32]. NRAS Luciferase plasmids [33] obtained from Addgene. Human cell lines were transfected for 48 hours with X-tremeGENE transfection reagent or

Lipofectamine 2000. CSTF3 overexpression plasmid was a kind gift from Bin Tian [34].

Antibodies

Tubulin (DSH Bank/E7), PUM1 (Bethyl/A300-201A), PUM2 (Bethyl/A300-202A),

PABPN1 (Abcam/AB75855), FOXO1 (Cell signaling/2880), JUN (Cell signaling/9165),

PTEN (Cell signaling/9552) and NRAS (Abcam/ab77392).

Luciferase assays

Cells were transfected and Luciferase levels were measured 48 hours post-transfection

(data are expressed as mean +/- s.d., n=3). Luciferase readings taken using the Dual-

Luciferase Reporter Assay System (Promega).

Lenti-viral shRNA

The DNA preparation, transfections, and virus preparation methods have been published elsewhere [4, 35]. siRNA transfection

Cells were transfected with siRNAs targeting PABPN1 and a scrambled control

(Dharmacon) using Lipofectamine RNAiMAX. Cells were analyzed using RT-PCR and western blotting 2 days post-transfection. Individual siRNAs targeting PUM1, PUM2 and

CSTF3 were from life technologies (Sequences in Supplementary table 2).

7

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

RNA stability assays

Cells were treated with Actinomycin D (5µM) and collected every hour. Transcript levels were assayed using RT-PCR. Cells were depleted using shRNA specific to PUM1,

PUM2 and scrambled controls and placed under puromycin selection for 4 days. Knock- down was validated by western blot.

3’RACE RT-PCR

Cells were lysed after selection for Lenti-viral infection and RNA was extracted using the

Qiagen RNeasy kit. cDNA was generated using an extended oligo-dT primer (P7 oligo-

dT). This cDNA was used in a standard RT-PCR reaction using a gene and poly-A site

specific primer as a reverse P7 primer [36].

Patients and Tumor Samples

All triple negative breast cancer specimens were formalin-fixed paraffin-embedded

(FFPE) biopsy derived from patients diagnosed at the Massachusetts General Hospital.

The diagnostic criteria/tumor grading were described previously [37]. The study was

approved by the Massachusetts General Hospital human research committee in

accordance with NIH human research study guidelines.

Quantitative Real-time PCR from tumor samples

RNA was isolated from tumor samples isolated using the Recoverall Total Nucleic Acid

Isolation Kit. KI-67 expression level was assessed using TaqMan

8

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Assay (4331182), and the expression of the two reference genes was assessed using custom primers/probes.

Primers

For primer sequences see Supplementary table 2.

Results

To identify which post-transcriptional regulatory motifs that were dis-proportionately affected by APA in tumor samples, we analyzed breast tumor datasets for APA events.

Breast tumors were chosen for this study as APA of E2F3 was common in breast cancer cells [4] and APA has previously been detected in this tumor type [26].

Microarray data from 38 breast tumors and 7 normal breast tissue samples were examined for APA events. As described in [26], microarray data obtained from the

Affymetrix 3' IVT series were used to assess APA using the 3' bias of their probes. This revealed mRNAs that showed a significantly different prevalence of 3’UTR isoforms in tumors compared to normal tissue. This analysis identified 921 shortened mRNAs and

189 lengthened mRNAs in human breast tumors at a false discovery rate of 5% [26]. To understand how APA of these transcripts changed the capacity of the post- transcriptional regulatory machinery to target these mRNAs, an unbiased motif mapping examination of the affected 3’UTRs was conducted. This computational search looked for post-transcriptional regulatory motif(s) that were enriched among the 3’UTR regions that underwent shortening or lengthening in cancer. This work found that the PRE

(UGUAXAUA) was the most frequently lost motif (p=0.01) in 3' UTR sequences

9

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

shortened in cancer (Fig. 1A and Supp Table 1A). In addition, the PRE was also the motif most often gained through APA (Fig. 1B and Supp Table 1B). These results suggested that PRE-containing transcripts are frequently altered by APA in tumors including numerous genes that are known to be important for oncogenesis.

Breast tumors can be sub-divided into sub-types based on distinct molecular alterations

[38, 39]. To understand how APA impacts these sub-types, we constructed APA profiles for each breast tumor group. This analysis revealed that although the majority of APA events occur in all breast tumor sub-types (821), basal-like and triple-negative breast tumors display more extensive and exclusive patterns of APA than luminal derived tumors (Figure 1C). analysis of APA specific events in triple-negative breast tumors, showed that APA affected mRNAs involved in the negative regulation of apoptosis, kinase activity and nucleotide binding (Figure 1C).

To determine the frequency of candidate 3’UTR alterations in tumors, an independent cohort of 34 triple-negative breast tumors and 8 samples of control breast tissue was examined. Triple-negative breast tumors were selected for this analysis, as this group had the most extensive APA profiles (Figure 1C) and exhibit the worst patient prognosis

[40]. RT-PCR was used to determine the relative expression levels of each 3’UTR isoform. Five candidate genes were selected for this analysis. Forkhead Box O1

(FOXO1) is the sole candidate with an extended 3’UTR sequence in tumor samples.

Phosphatase and tensin homolog (PTEN), Neuroblastoma RAS viral (v-Ras) oncogene homolog (NRAS) and the Jun proto-oncogene (c-JUN), all showed recurrent 3’UTR shortening in our database analysis. E2F4 was used as a negative control and showed no detectable level of APA. In this cohort of tumors, E2F4 was not altered by APA

10

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

(Supp Figure 1A-C) and 3’UTR extensions of FOXO1 occurred only rarely (twice)

(Figure 2A, Supp Fig 1D/E). In contrast, 3’UTR shortening was much more prevalent.

Using RT-PCR, we identified tumors containing short 3’UTR isoforms of NRAS, c-JUN

and PTEN (Figure 2B-D, Supp Fig 1F-H, Supp Fig 2/3). In particular, shortening of the

NRAS (16/34) and c-JUN 3’UTRs (8/34) were frequent events (Figure 2C-E).

Due to the frequency of these APA events, we focused on understanding how APA

changed NRAS and c-JUN mRNA-regulation. We asked whether changes in 3’UTR

length of NRAS and c-JUN correlated with changes in downstream target gene expression. Tumors expressing the shortened isoform of the NRAS 3’UTR (<50%) displayed elevated levels of Forkhead Box A1 (FOXA1) and SMG1 Phosphatidylinositol

3-Kinase-Related Kinase (SMG1), genes previously shown to respond to NRAS signaling [41] (Figure 2F). Tumors expressing the shortened form of c-JUN had higher expression of the c-JUN activated genes, Cytokeratin 7 (KRT7) and TIMP

Metallopeptidase Inhibitor 1 (TIMP1) [42], and lower levels of the c-JUN repressed gene, Cytokeratin 19 (KRT19) [42] (Figure 2G). These results suggest that APA effects the levels and activity of its targets and leads to changes in the patterns of downstream transcriptional programs.

To examine the possible role for PUM in regulating our candidate genes (FOXO1,

PTEN, NRAS and c-JUN), we assayed the consequence of depleting PUM1 or PUM2 from MDA-MB-231 triple-negative breast cancer (TNBC) cells. As a control, the length of each candidate mRNA was measured using RT-qPCR. This analysis found that the shortened 3’UTR isoform of c-JUN was expressed at high levels in MDA-MB-231 cells

(Figure 3A). siRNA-mediated knock-down of PUM1 or PUM2 in MDA-MB-231 cells

11

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

increased the protein levels of PTEN, NRAS and FOXO1 but not c-JUN (Figure 3B).

The triple-negative breast cancer cell line with the lowest levels of c-JUN APA, CAL51

(Figure 3C), and was then used to test how depleting PUM1 or PUM2 changed c-JUN

protein levels. As shown in Figure 3D, reduced PUM levels modestly increased c-JUN

protein levels.

To determine how PUM affected cell proliferation, the effect of PUM depletion on CAL51 cells was assayed. As shown in Figure 3E, reduced PUM1 or PUM2 levels modestly slowed CAL51 growth rates, suggesting that PUM-regulation may help to maintain

proliferation in TNBC cells. To assay how APA-mediated shortening of c-JUN effected the invasive potential of cells, the capacity of the c-JUN coding sequence and either the short or full length 3’UTR to rescue the invasive capacity of c-JUN-/- mouse embryonic fibroblasts (MEFs) was tested. The ability of HRAS to promote cellular invasion was dependent on c-JUN (compare Wild-type vs. c-JUN-/- MEFs + HRAS) (Figure 3F) and provided an ideal model to evaluate APA. In this system, the over-expression of the c-

JUN CDS + short 3’UTR greatly improved the invasive capacity of cells, compared to the expression of the c-JUN CDS + full length 3’UTR (Figure 3F). These results suggest that PUM and APA regulation of c-JUN may contribute to the proliferation and invasion potential of cells.

To examine how APA modified PUM-mediated control, the short and long 3’UTR isoform of each candidate was cloned downstream of Luciferase reporter genes. These plasmids were then transfected into MDA-MB-231 cells depleted of PUM1, PUM2 or scrambled control sequences (Supp Figure 4A). Lengthening of the FOXO1 3’UTR increased the number of PRE elements to 4 (Figure 4A). Extension of the FOXO1

12

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

3’UTR decreased Luciferase levels (Full length vs. Extended in Scr) (Figure 4A). This

effect is partially dependent on PUM activity (Figure 4A). To examine how APA change

mRNA degradation, mRNA stability assays were conducted. Cells were treated with the

polymerase inhibitor, Actinomycin D, and Luciferase mRNA levels measured over time.

Extension of the FOXO1 3’UTR increased the stability of the Luciferase mRNA (Figure

4B). To determine PUM’s contribution to these effects, these experiments were repeated in MDA-MB-231 cells depleted of PUM. Reduced PUM activity increased the mRNA stability of the full length 3’UTR but not the extended 3’UTR isoform (Supp

Figure 4B). These results show that the PUM protein complex modulates FOXO1 levels

and that APA-mediated lengthening increased PUM-regulation.

This dual approach, Luciferase reporter and mRNA stability assays, was used to test

how shortening of the candidate genes might affect Luciferase production. APA of

PTEN removed the sole PRE sequence from the 3’UTR (Figure 4C) and increased

Luciferase levels (Figure 4C). Knock-down of PUM1 or PUM2 modestly elevated

Luciferase production from the short 3’UTR isoform (Figure 4C). Surprisingly, APA of

PTEN or PUM depletion did not change the overall stability of the mRNA (Figure 4D,

Supp Figure 4C). These results suggested that PUM-mediated regulation of PTEN is

only partially responsible for constraining PTEN levels.

Shortening of the NRAS 3’UTR circumvents PUM repression by removing the sole PRE

(Figure 4E) and significantly increased Luciferase production (Figure 4E). Depletion of

PUM1 or PUM2 only moderately elevated protein levels from the full-length 3’UTR

(Figure 4E, Scr vs. PUM). This suggested that other regulatory mechanisms are also

circumvented upon NRAS 3’UTR shortening. Site-directed mutagenesis was used to

13

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

inactivate the sole PRE within the NRAS 3’UTR (UGUAXAUA-UCCAXAUA). This

Luciferase construct was then transfected into MDA-MB-231 and MCF7 cells depleted

of PUM1 or PUM2 (Supp Figure 4D). As shown in Supp Figure 4E/F, inactivation of the

PRE within the NRAS 3’UTR only partially rescued protein levels. To examine how APA

and PUM modulate the stability of both 3’UTR isoforms of NRAS we conducted RNA

stability assays. These results showed that the half-life of the short 3’UTR of NRAS was

greatly increased, compared to full-length (Figure 4F) and that this increase was due to reduced PUM regulation (Supp Figure 5A). In support of this, depletion of PUM1 or

PUM2 increased the stability of the NRAS full length 3’UTR (Supp Figure 5A). These results indicate that PUM regulates the stability of NRAS mRNA.

Unlike the other shortened candidates, APA of c-JUN only removes 1 of the 4 PRE motifs (Figure 4G). In the Luciferase assay system, shortening the c-JUN 3’UTR increased Luciferase levels. In addition, both 3’UTR isoforms of c-JUN were sensitive to

PUM activity (Figure 4G). RNA stability assays surprisingly showed that the shortened

3’UTR was more unstable than the full-length 3’UTR (Figure 4H) and that this was in part PUM dependent (Supp Figure 5B). These findings suggest that PUM tightly modulates both the stability and the translation of c-JUN mRNA.

To investigate the clinical characteristics of tumors with APA in this cohort, tumors were sub-divided based on the APA status and clinical parameters. Tumors containing APA of c-JUN and/or NRAS were split from those expressing the full length versions. Tumors were then divided into groups based on pathological tumor grade. This produced 3 groups: Grade 2 tumors, all of which had APA (n=6), and Grade 3 tumors, with (n=12) and without APA (n=12). In these tumors, every grade 2 tumor displayed APA, and so

14

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

no grade 2 without APA group was formed. This analysis revealed a number of unexpected results. First, APA was more prevalent in smaller, lower grade tumors

(Figure 5A). Secondly, APA of NRAS and c-JUN was evident in less-proliferative tumors, as measured by Ki67 RT-PCR (Figure 5B). Thirdly, triple-negative breast tumors utilizing APA appear more invasive, as they were more frequently associated with positive lymph node status, compared to non-APA tumors (Figure 5C). These results support the idea that APA of PUM substrates may contribute to tumor phenotypes and demonstrate that APA in triple-negative breast tumors does not directly correlate with proliferation.

To understand why APA disproportionately affected transcripts containing PRE motifs, we asked whether PUM proteins have a role in regulating APA. RT-PCR was used to compare PUM levels between tumors but showed no variation in expression (Figure

5D). To directly evaluate PUM’s role in regulating APA, PUM1, PUM2 or the APA regulator Poly-A Binding Protein Nuclear 1 (PABPN1) [36] were depleted from MDA-

MB-231 cells and APA levels compared. Reduced PUM levels did not modify previously characterized APA substrates, compared to PABPN1 (Figure 5E and Supp Figure 6A-

D). These findings are in agreement with previous work discounting PUM as a direct regulator of APA in cancer cell lines [36].

To investigate the molecular drivers of APA in these tumors, the expression levels of the polyadenylation machinery was analyzed using RT-PCR. These factors represent prime

APA-regulating candidates, as they are directly responsible for poly-A site selection/catalysis. This analysis identified elevated levels of two components of the polyadenylation machinery in tumors with APA; Cleavage Stimulation Factor 3 (CSTF3)

15

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

and Cleavage Polyadenylation Specificity Factor 3 (CPSF3) (Figure 5F). These

observations suggested that APA in triple-negative breast tumors may be regulated by

fluctuations in the levels of the polyadenylation machinery.

As CSTF3 levels are elevated in both tumor grades with APA, we examined its potential

role in modulating APA. To do this RPE cells (which have low levels of APA), were

transfected with plasmids expressing either CSTF3 [34] or an empty plasmid. In RPE

cells, the overexpression of CSTF3 (Supp Figure 6E) was sufficient to induce APA-

mediated 3’UTR shortening of c-JUN and NRAS but not E2F4 (Figure 5G). To ask if the over-expression of CSTF3 was common in triple-negative breast cancer cell lines, we analyzed mRNA levels of CSTF3. As shown in Figure 6A, triple-negative breast cancer cell lines (TNBC) express higher levels of CSTF3 than other breast cancer cell lines

(non-TNBC). To determine the contribution of CSTF3 to proliferation in breast cancer cells, CSTF3 was depleted from SK-BR-3 (non-TNBC), BT-20 (TNBC) and BT-549

(TNBC) cells using single siRNAs. Each of these cell lines expressed different levels of

CSTF3 (Figure 6B) however siRNA-treatment resulted in strong CSTF3 depletion

(Figure 6C). Proliferation assays from each cell line indicated that CSTF3 silencing reduced the proliferation TNBC but not non-TNBC cells (Figure 6D). These results suggest that fluctuations in the expression of CSTF3, can modify the 3’UTR length of

NRAS and c-JUN and contribute to proliferation.

Discussion

These results show that divergent patterns of APA occur in different subtypes of breast cancers and highlight the significance of APA as a modulator of NRAS and c-JUN gene

16

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

expression in triple-negative breast tumors/cells. We propose that APA is an adaptive

mechanism that alters gene expression without requiring genetic re-arrangements. In

agreement with this hypothesis, previous studies have identified aberrant c-JUN and

NRAS activity in triple-negative breast tumors without detecting a mechanism of activation [28]. In addition, the over-expression of the c-JUN and NRAS target genes,

KRT7 and FOXA1, and the silencing of KRT19, which is directly repressed by c-JUN, have been previously correlated to increased local recurrence in triple-negative breast cancer patients [43, 44]. These findings suggest that non-genetic changes in c-JUN and

NRAS may alter their respective transcriptional profiles and contribute to localized tumor relapse.

The prevalence of APA in cancer cells has been linked to multiple parameters including chromatin structure [45, 46], silencing of APA inhibiting proteins [36, 47, 48] and the aberrant expression of the polyadenylation machinery [25, 27]. One emerging theme from studies investigating APA, is that imbalances in the levels of the polyadenylation machinery, such as the down-regulation of CFIm25 [25] or the increased expression of

CSTF2 [27], changes the APA spectrum. In our study, APA levels in tumors correlated with the elevated expression of polyadenylation factor, CSTF3. These results show that increased CSTF3 expression is sufficient to trigger APA shortening of both NRAS and c-JUN. Our results are consistent with previous work and suggest that changes in the expression of the polyadenylation machinery promote APA.

These findings pose the question: Why are mRNAs that contain binding sites for PUM, a cytoplasmic RNA binding protein, disproportionately affected by the nuclear process of poly-A site selection? Insight into this relationship was provided when we compared

17

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

the motifs utilized by the PUM complex to those used by the polyadenylation machinery.

This comparison revealed that the two central polyadenylation complexes, the Cleavage

Factor I (CFI) complex and the Cleavage Stimulation Factor (CSTF) complex, interact

with GU-rich sequences highly similar to the PRE and the sequence bound by the PUM-

interacting protein, NANOS. The CFI complex, which functions upstream of the Poly-A

site, to mediate mRNA cleavage, interacts with the same RNA sequence as the PRE,

CFI: UGUA [25, 49] and PRE: UGUAXAUA [5] (Figure 7A). In addition, the CSTF

complex, which functions downstream of the Poly-A site binds to a GU rich region [49],

homologous to the GUUGU repeats favored by the PUM interacting protein, NANOS [2,

8] (Figure 7A). These findings support a simple model: that changes in the expression

of the polyadenylation machinery promotes APA through the preferential use of

sequences that are also recognized by PUM and the PUM-interacting protein, NANOS

(Figure 7B).

Our research reveals a surprisingly dis-proportionate level of APA on PRE-containing

mRNAs in triple-negative breast tumors. It is possible that this is a unique feature of

these tumors, but given the general similarity between the motifs used by the

polyadenylation machinery and by PUM complexes, we anticipate that this association

is likely to hold true in other contexts. Indeed, we note that PUM2 motifs were present

in putative APA substrates identified in glioblastoma cells [50] and that several of the

APA targets identified in developmental studies are, in fact, PRE-containing substrates

[16, 20]. Thus, due to the similarity of the sequences used by the PUM complex and by the polyadenylation machinery, it seems likely that PRE-containing substrates are often dis-proportionately affected by APA.

18

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

In summary, our results suggest that in triple-negative breast tumors, APA is re- activated by the aberrant over-expression of the polyadenylation machinery. APA of

PUM targets, NRAS and c-JUN, leads to the removal of functional PRE motifs and the aberrant activation of these substrates, which contributes to changes in the expression of their downstream targets. Based on our findings, we propose that APA is a form of gene regulation that is exploited by triple-negative breast tumors to promote changes in the transcriptome and proteome that are conducive to tumor growth.

Acknowledgements

We would like to thank Shyamala Maheswaran, Leif Ellisen and Lee Zou for their valuable insights.

19

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Bibliography

1. Kechavarzi, B. and S.C. Janga, Dissecting the expression landscape of RNA-

binding proteins in human cancers. Genome Biol, 2014. 15(1): p. R14.

2. Miles, W.O., M. Korenjak, L.M. Griffiths, M.A. Dyer, P. Provero, and N.J. Dyson,

Post-transcriptional gene expression control by NANOS is up-regulated and

functionally important in pRb-deficient cells. EMBO J, 2014.

3. Ciafre, S.A. and S. Galardi, microRNAs and RNA-binding proteins: a complex

network of interactions and reciprocal regulations in cancer. RNA Biol, 2013.

10(6): p. 935-42.

4. Miles, W.O., K. Tschop, A. Herr, J.Y. Ji, and N.J. Dyson, Pumilio facilitates

miRNA regulation of the E2F3 oncogene. Genes Dev, 2012. 26(4): p. 356-68.

5. Galgano, A., M. Forrer, L. Jaskiewicz, A. Kanitz, M. Zavolan, and A.P. Gerber,

Comparative analysis of mRNA targets for human PUF-family proteins suggests

extensive interaction with the miRNA regulatory system. PLoS One, 2008. 3(9):

p. e3164.

6. Gerber, A.P., D. Herschlag, and P.O. Brown, Extensive association of

functionally and cytotopically related mRNAs with Puf family RNA-binding

proteins in yeast. PLoS Biol, 2004. 2(3): p. E79.

7. Gerber, A.P., S. Luschnig, M.A. Krasnow, P.O. Brown, and D. Herschlag,

Genome-wide identification of mRNAs associated with the translational regulator

PUMILIO in melanogaster. Proc Natl Acad Sci U S A, 2006. 103(12):

p. 4487-92.

20

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

8. Sonoda, J. and R.P. Wharton, Recruitment of Nanos to hunchback mRNA by

Pumilio. Genes Dev, 1999. 13(20): p. 2704-12.

9. Jaruzelska, J., M. Kotecki, K. Kusz, A. Spik, M. Firpo, and R.A. Reijo Pera,

Conservation of a Pumilio-Nanos complex from Drosophila germ plasm to human

germ cells. Dev Genes Evol, 2003. 213(3): p. 120-6.

10. Friend, K., Z.T. Campbell, A. Cooke, P. Kroll-Conner, M.P. Wickens, and J.

Kimble, A conserved PUF-Ago-eEF1A complex attenuates translation elongation.

Nat Struct Mol Biol, 2012. 19(2): p. 176-83.

11. Cao, Q., K. Padmanabhan, and J.D. Richter, Pumilio 2 controls translation by

competing with eIF4E for 7-methyl guanosine cap recognition. RNA, 2010. 16(1):

p. 221-7.

12. Van Etten, J., T.L. Schagat, J. Hrit, C.A. Weidmann, J. Brumbaugh, J.J. Coon, et

al., Human Pumilio proteins recruit multiple deadenylases to efficiently repress

messenger RNAs. J Biol Chem, 2012. 287(43): p. 36370-83.

13. Kedde, M., M. van Kouwenhove, W. Zwart, J.A. Oude Vrielink, R. Elkon, and R.

Agami, A Pumilio-induced RNA structure switch in p27-3' UTR controls miR-221

and miR-222 accessibility. Nat Cell Biol, 2010. 12(10): p. 1014-20.

14. Nolde, M.J., N. Saka, K.L. Reinert, and F.J. Slack, The

pumilio homolog, puf-9, is required for the 3'UTR-mediated repression of the let-7

microRNA target gene, hbl-1. Dev Biol, 2007. 305(2): p. 551-63.

15. Hilgers, V., M.W. Perry, D. Hendrix, A. Stark, M. Levine, and B. Haley, Neural-

specific elongation of 3' UTRs during Drosophila development. Proc Natl Acad

Sci U S A, 2011. 108(38): p. 15864-9.

21

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

16. Ji, Z., J.Y. Lee, Z. Pan, B. Jiang, and B. Tian, Progressive lengthening of 3'

untranslated regions of mRNAs by alternative polyadenylation during mouse

embryonic development. Proc Natl Acad Sci U S A, 2009. 106(17): p. 7028-33.

17. Li, Y., Y. Sun, Y. Fu, M. Li, G. Huang, C. Zhang, et al., Dynamic landscape of

tandem 3' UTRs during zebrafish development. Genome Res, 2012. 22(10): p.

1899-906.

18. Gupta, I., S. Clauder-Munster, B. Klaus, A.I. Jarvelin, R.S. Aiyar, V. Benes, et al.,

Alternative polyadenylation diversifies post-transcriptional regulation by selective

RNA-protein interactions. Mol Syst Biol, 2014. 10: p. 719.

19. Tian, B., J. Hu, H. Zhang, and C.S. Lutz, A large-scale analysis of mRNA

polyadenylation of human and mouse genes. Nucleic Acids Res, 2005. 33(1): p.

201-12.

20. Smibert, P., P. Miura, J.O. Westholm, S. Shenker, G. May, M.O. Duff, et al.,

Global patterns of tissue-specific alternative polyadenylation in Drosophila. Cell

Rep, 2012. 1(3): p. 277-89.

21. Yoon, O.K., T.Y. Hsu, J.H. Im, and R.B. Brem, Genetics and regulatory impact of

alternative polyadenylation in human B-lymphoblastoid cells. PLoS Genet, 2012.

8(8): p. e1002882.

22. Sandberg, R., J.R. Neilson, A. Sarma, P.A. Sharp, and C.B. Burge, Proliferating

cells express mRNAs with shortened 3' untranslated regions and fewer

microRNA target sites. Science, 2008. 320(5883): p. 1643-7.

22

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

23. Fu, Y., Y. Sun, Y. Li, J. Li, X. Rao, C. Chen, et al., Differential genome-wide

profiling of tandem 3' UTRs among human breast cancer and normal cells by

high-throughput sequencing. Genome Res, 2011. 21(5): p. 741-7.

24. Mayr, C. and D.P. Bartel, Widespread shortening of 3'UTRs by alternative

cleavage and polyadenylation activates oncogenes in cancer cells. Cell, 2009.

138(4): p. 673-84.

25. Masamha, C.P., Z. Xia, J. Yang, T.R. Albrecht, M. Li, A.B. Shyu, et al., CFIm25

links alternative polyadenylation to glioblastoma tumour suppression. Nature,

2014.

26. Lembo, A., F. Di Cunto, and P. Provero, Shortening of 3'UTRs correlates with

poor prognosis in breast and lung cancer. PLoS One, 2012. 7(2): p. e31129.

27. Aragaki, M., K. Takahashi, H. Akiyama, E. Tsuchiya, S. Kondo, Y. Nakamura, et

al., Characterization of a cleavage stimulation factor, 3' pre-RNA, subunit 2, 64

kDa (CSTF2) as a therapeutic target for lung cancer. Clin Cancer Res, 2011.

17(18): p. 5889-900.

28. Lehmann, B.D., J.A. Bauer, X. Chen, M.E. Sanders, A.B. Chakravarthy, Y. Shyr,

et al., Identification of human triple-negative breast cancer subtypes and

preclinical models for selection of targeted therapies. J Clin Invest, 2011. 121(7):

p. 2750-67.

29. Parikh, R.R., D. Housman, Q. Yang, D. Toppmeyer, L.D. Wilson, and B.G. Haffty,

Prognostic value of triple-negative phenotype at the time of locally recurrent,

conservatively treated breast cancer. Int J Radiat Oncol Biol Phys, 2008. 72(4):

p. 1056-63.

23

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

30. Richardson, A.L., Z.C. Wang, A. De Nicolo, X. Lu, M. Brown, A. Miron, et al., X

chromosomal abnormalities in basal-like human breast cancer. Cancer Cell,

2006. 9(2): p. 121-32.

31. Barrett, T., S.E. Wilhite, P. Ledoux, C. Evangelista, I.F. Kim, M. Tomashevsky, et

al., NCBI GEO: archive for functional genomics data sets--update. Nucleic Acids

Res, 2013. 41(Database issue): p. D991-5.

32. Polak, P., A. Oren, I. Ben-Dror, D. Steinberg, S. Sapoznik, A. Arditi-Duvdevany,

et al., The cytoskeletal network controls c-Jun translation in a UTR-dependent

manner. Oncogene, 2006. 25(5): p. 665-76.

33. Johnson, S.M., H. Grosshans, J. Shingara, M. Byrom, R. Jarvis, A. Cheng, et al.,

RAS is regulated by the let-7 microRNA family. Cell, 2005. 120(5): p. 635-47.

34. Luo, W., Z. Ji, Z. Pan, B. You, M. Hoque, W. Li, et al., The conserved intronic

cleavage and polyadenylation site of CstF-77 gene imparts control of 3' end

processing activity through feedback autoregulation and by U1 snRNP. PLoS

Genet, 2013. 9(7): p. e1003613.

35. Miles, W.O., M. Korenjak, L.M. Griffiths, M.A. Dyer, P. Provero, and N.J. Dyson,

Post-transcriptional gene expression control by NANOS is up-regulated and

functionally important in pRb-deficient cells. EMBO J, 2014. 33(19): p. 2201-15.

36. Jenal, M., R. Elkon, F. Loayza-Puch, G. van Haaften, U. Kuhn, F.M. Menzies, et

al., The poly(A)-binding protein nuclear 1 suppresses alternative cleavage and

polyadenylation sites. Cell, 2012. 149(3): p. 538-53.

24

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

37. Sgroi, D.C., E. Carney, E. Zarrella, L. Steffel, S.N. Binns, D.M. Finkelstein, et al.,

Prediction of late disease recurrence and extended adjuvant letrozole benefit by

the HOXB13/IL17BR biomarker. J Natl Cancer Inst, 2013. 105(14): p. 1036-42.

38. Bertucci, F., P. Finetti, J. Rougemont, E. Charafe-Jauffret, N. Cervera, C. Tarpin,

et al., Gene expression profiling identifies molecular subtypes of inflammatory

breast cancer. Cancer Res, 2005. 65(6): p. 2170-8.

39. Jacquemier, J., C. Ginestier, J. Rougemont, V.J. Bardou, E. Charafe-Jauffret, J.

Geneix, et al., Protein expression profiling identifies subclasses of breast cancer

and predicts prognosis. Cancer Res, 2005. 65(3): p. 767-79.

40. Shah, S.P., A. Roth, R. Goya, A. Oloumi, G. Ha, Y. Zhao, et al., The clonal and

mutational evolution spectrum of primary triple-negative breast cancers. Nature,

2012. 486(7403): p. 395-9.

41. Neben, K., S. Schnittger, B. Brors, B. Tews, F. Kokocinski, T. Haferlach, et al.,

Distinct gene expression patterns associated with FLT3- and NRAS-activating

mutations in acute myeloid leukemia with normal karyotype. Oncogene, 2005.

24(9): p. 1580-8.

42. Rinehart-Kim, J., M. Johnston, M. Birrer, and T. Bos, Alterations in the gene

expression profile of MCF-7 breast tumor cells in response to c-Jun. Int J Cancer,

2000. 88(2): p. 180-90.

43. Parikh, R.R., Q. Yang, S.A. Higgins, and B.G. Haffty, Outcomes in young women

with breast cancer of triple-negative phenotype: the prognostic significance of

CK19 expression. Int J Radiat Oncol Biol Phys, 2008. 70(1): p. 35-42.

25

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

44. Fujisue, M., R. Nishimura, Y. Okumura, R. Tashima, Y. Nishiyama, T. Osako, et

al., Clinical Significance of CK19 Negative Breast Cancer. Cancers (Basel),

2012. 5(1): p. 1-11.

45. Lee, C.Y. and L. Chen, Alternative polyadenylation sites reveal distinct chromatin

accessibility and histone modification in human cell lines. Bioinformatics, 2013.

29(14): p. 1713-7.

46. Huang, H., J. Chen, H. Liu, and X. Sun, The nucleosome regulates the usage of

polyadenylation sites in the . BMC Genomics, 2013. 14: p. 912.

47. Berg, M.G., L.N. Singh, I. Younis, Q. Liu, A.M. Pinto, D. Kaida, et al., U1 snRNP

determines mRNA length and regulates isoform expression. Cell, 2012. 150(1):

p. 53-64.

48. Kaida, D., M.G. Berg, I. Younis, M. Kasim, L.N. Singh, L. Wan, et al., U1 snRNP

protects pre-mRNAs from premature cleavage and polyadenylation. Nature,

2010. 468(7324): p. 664-8.

49. Martin, G., A.R. Gruber, W. Keller, and M. Zavolan, Genome-wide analysis of

pre-mRNA 3' end processing reveals a decisive role of human cleavage factor I

in the regulation of 3' UTR length. Cell Rep, 2012. 1(6): p. 753-63.

50. Shao, J., J. Zhang, Z. Zhang, H. Jiang, X. Lou, B. Huang, et al., Alternative

polyadenylation in glioblastoma multiforme and changes in predicted RNA

binding protein profiles. OMICS, 2013. 17(3): p. 136-49.

26

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure Legends

Figure 1: PRE-containing mRNAs are dis-proportionately targeted by alternative polyadenylation

(A) Percentage of the 921 mRNAs shortened by alternative polyadenylation in breast tumors that lose PRE (PUM) or miRNA seed sequences (**p<0.01). (B) Percentage of the 189 mRNAs lengthened by alternative polyadenylation in breast tumors that gain

PRE (PUM) or miRNA seed sequences. (C) Breast tumor sub-type shared and unique

APA events. Table: Gene ontology of PRE containing mRNAs shortened in triple- negative breast tumors.

Figure 2: Candidate gene alternative polyadenylation status in an independent cohort of triple-negative breast tumors

(A) RT-PCR of FOXO1 from tumor and control samples. Tumors were sub-divided based on FOXO1 length at UTR (>50% or <50%), short and long. Black bracket indicates lengthened 3’UTRs in tumors and the number represents the number of tumors displaying this APA event. (B) RT-PCR of PTEN from tumor and control samples. Black bracket indicates number of shortened 3’UTRs. (C) RT-PCR of NRAS from tumor and control samples. Black bracket indicates number of shortened 3’UTRs.

(D) RT-PCR of the c-JUN from tumors and control samples. Black bracket indicates number of shortened 3’UTRs. (E) Percentage of tumors displaying APA of each transcript. E2F4 was used as a negative control. (F) NRAS target gene expression

(FOXA1 and SMG1) in tumors expressing either the long or short 3’UTR isoform of

NRAS (**p<0.01, *p<0.05). (G) c-JUN target gene expression (KRT7 and TIMP1:

27

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

activated, KRT19: repressed) in tumors expressing either the long or short 3’UTR

isoform of c-JUN (**p<0.01, *p<0.05).

Figure 3: PUM-regulation of candidate gene protein levels

(A) RT-PCR from triple-negative breast cancer cells MDA-MB-231 for PTEN, NRAS, c-

JUN and FOXO1. (B) Western blots from siRNA treated MDA-MB-231 cells. (C)

3’RACE RT-PCR from CAL51 cells for c-JUN. (D) Western blots of CAL51 cells

transfected with individual siRNAs targeting GFP, PUM1 and PUM2. (E) Cell number

assay from CAL51 cells transfected with individual siRNAs. (F) Number of HRAS- mediated invasive cells from wild-type (WT) or c-JUN null MEFs (JUN-/-), infected with the c-JUN coding sequences and either 3’UTR isoform (*p<0.05).

Figure 4: The effect of APA on mRNA stability and protein production on

candidate genes

(A) Table: Change in PRE number (Δ in PRE) and total PRE after APA of FOXO1.

Graph: Luciferase assays comparing FOXO1 Full length and extended 3’UTR isoforms

in MDA-MB-231 cells depleted of PUM1, PUM2 or control Scrambled sequence (Scr)

(**p<0.01). Full length 3’UTR isoform set to 1. (B) mRNA stability assays of the FOXO1

Full length or extended 3’UTR isoforms from MDA-MB-231 cells treated with the

Polymerase inhibitor, Actinomycin D (**p<0.01, *p<0.05). (C) Table: Transcript information for the shortened 3’UTR of PTEN. Graph: Luciferase assays comparing

PTEN full-length and short 3’UTR isoforms in MDA-MB-231 cells depleted of PUM1,

PUM2 or Scrambled (**p<0.01). (D) mRNA stability assays of the PTEN short or long

3’UTR isoforms from MDA-MB-231 cells treated with Actinomycin D (**p<0.01). (E)

28

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Table: Transcript information for the shortened 3’UTR of NRAS. Graph: Luciferase assays comparing NRAS full-length and short 3’UTR isoforms in MDA-MB-231 cells depleted of PUM1, PUM2 or Scrambled (*p<0.05). (F) mRNA stability assays of the

NRAS short or long 3’UTR isoforms from MDA-MB-231 cells treated with Actinomycin

D. (G) Table: Transcript information for the shortened 3’UTR of c-JUN. Graph:

Luciferase assays comparing c-JUN full-length and short 3’UTR isoforms in MDA-MB-

231 cells depleted of PUM1, PUM2 or Scrambled (**p<0.01, *p<0.05). (H) mRNA stability assays of the c-JUN short or long 3’UTR isoforms from MDA-MB-231 cells treated with Actinomycin D (**p<0.01).

Figure 5: APA in triple-negative breast tumors correlates with slower growth, smaller yet more invasive tumors.

(A) Tumor size of triple-negative breast tumors, divided by grade and APA status

(*p<0.05). Grade 2 tumors (n=6), Grade 3 APA tumors (n=12) and Grade 3 non-APA tumors (n=12). (B) Ki67 levels of triple-negative breast tumors, divided by grade and

APA status (*p<0.05). (C) Lymph node status (positive or negative) from patients with triple-negative breast tumors, divided by grade and APA status. (D) RT-PCR analysis of

PUM1 and PUM2 from triple-negative breast tumors, divided by grade and APA status.

(E) 3’RACE experiments from MDA-MB-231 cells depleted of PUM1, PUM2, PABPN1 or Scrambled control sequence by shRNA. Change in 3’UTR length calculated by dividing distal values with proximal readings. (F) RT-PCR analysis of the polyadenylation machinery from triple-negative breast tumors, divided by grade and

APA status (**p<0.01). (G) RT-PCR measuring mRNA length from cells transfected with either the pCDNA-empty vector or pCDNA-CSTF3 (**p<0.01, *p<0.05).

29

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 6: Triple-negative breast cancer cell lines are more dependent on CSTF3

(A) Expression of CSTF3 from triple-negative breast cancer cell lines (TNBC) vs. breast

cancer cells from other breast cancer sub-types. (B) CSTF3 expression levels from SK-

BR-3 (non-TNBC), BT-20 (TNBC) and BT-549 (TNBC) cells. (C) Relative CSTF3

depletion from the three cell lines using independent siRNAs. (D) Cell numbers from

SK-BR-3, BT-20 and BT-549 depleted of GFP or CSTF3 by siRNA (**p<0.01, *p<0.05).

Figure 7: The polyadenylation machinery utilizes GU-rich sequences of the PUM

complex to catalyze APA.

(A) Schematic of the positioning and sequence composition of motifs recognized by the

CFI, CSTF and PUM complexes. (B) Model describing how the CFI and CSTF complex

can use motifs of the PUM complex for APA.

30

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844

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

A i S h o r te n e d tr a n s c r ip ts B

n

i L e n g th e n e d tr a n s c r ip ts

s 4 0

f

s e

i 3 0

f

t

e

i u

** t

o

u

s

o

s

s

m

i

s

m

t

i

d 3 0

t

l

d

e

l

a

e g

a 2 0

m

g

n

r

m

n

a

r

o

a h

2 0 o

n

h

c

n

.

c

f

.

s

f

o

s

v

o

v

e 1 0

s e

r 1 0

g

s

r

g

a

o

t

a

o

t

m

n

m

n

e

u

t

e

u

c t

r 0 c

r 0

e e p M p 4 p 5 2 3 p 0 3 U - 1 - M 3 0 0 - 0 U - P 4 9 -5 iR P iR -3 -5 R m m i iR iR m m m M o tifs M o tifs C Gene ontology of shortened mRNAs containing putative Sub-type specific APA events PREs in Triple-negative breast tumors Molecular Process Enrichment P-value Basal-like Luminal A 37 Negative regulation of apoptosis 1.43 3.6e-2 2 Ion homeostasis 1.26 4.3e-2 Kinase activity 1.12 4.1e-2 821 Nucleotide binding 1.05 3.5e-2 3 Luminal B Regulation of phosphorylation 1.05 8.3e-2 Triple-negative 58 T cell activation 1.01 6.5e-1 ATP Binding 0.93 5.3e-2 Cell cycle 0.69 1.4e-1

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Miles Figure 1 Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

A B C F O X O 1 m R N A le n g th P T E N m R N A le n g th N R A S m R N A le n g th

1 .2 1 .2 1 .2

n n

n 1 .0 1 .0 1 .0

o

o

i

o

i

i

s

s

s

s

s s

e 0 .8 0 .8 e

e 0 .8

r

r

r

p

p

p

x

x

x

e

e e

0 .6 0 .6

0 .6

e

e

e

n

n

n

e

e

e

g g

0 .4 0 .4 g 0 .4

T u m o r 3 'U T R E xte n d e d 2 T u m o r S h o rt

e e

4 e T u m o r S h o rt

v

v

v

i i

T u m o r L o n g i

t t

0 .2 T u m o r F u ll L e n g th 0 .2 t 0 .2 16 a

a T u m o r L o n g

a

l l

N o rm a l B re a s t l

e e

N o rm a l B re a s t e N o rm a l B re a s t

R R 0 .0 0 .0 R 0 .0

1 2 1 .1 2 1 2 1 2 1 2 1 2 2 1 S S R R S S S S . . R . D D T 1 T R R 1 1 2 R D D T T D D R R T R C C U T U C C U U C C T T U T U U U U P T E N P R IM E R S E T F O X O 1 P R IM E R S E T N R A S P R IM E R S E T S

N R A S ta r g e t g e n e s c -J U N ta r g e t g e n e s D c -J U N m R N A le n g th E T rip le -n e g a tiv e b re a s t F G tu m o rs F O X A 1 K R T 7 T IM P 1

1 .2 A S M G 1 K R T 1 9

P 5 0

n 4 2 .0

n

o

A

i

o n

1 .0 i

s ** **

g **

o

s

i

s n

4 0 s

s

i

e

s

e r

y 3 1 .5 r

e 0 .8

p

a

r

p

l

x

p

x

p e

x 3 0

e

s

e

i *

0 .6

e

e e

d 2 1 .0

n

n

n

e

s e

e 2 0 r

g * g

g 0 .4

o

e e

e T u m o r S h o rt 8

v

v

m v

i 0 .5 i

1 i

t

u t

0 .2 T u m o r L o n g 1 0 t

t

a

a

a

l

l

l f

e N o rm a l B re a s t

e

e

o

R

R 0 .0 R

0 0 0 .0 % 2 1 2 .1 1 N S G T G T S R R 4 N R R D T T 2 O E U A N N R F O O C U U T 2 X T J R O O - L H L H U E O P c N S F S P re d o m in a n t c -J U N c -J U N P R IM E R S E T S P re d o m in a n t N R A S P u ta tiv e A P A c h a n g e s 3 'U T R is o fo rm 3 'U T R is o fo rm Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Miles Figure 2 Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

A M D A -M B -2 3 1 B C D CAL51 cells F u ll L e n g th S h o rt siRNA siRNA 1 .0 1 .0

PUM1

h

s t

PUM2 l 0 .8 g

e c-JUN

n

v

e

e

l

l

NRAS 0 .6

N

R U T PUM1

PTEN J U

0 .5 -

'

c 3

0 .4

e

e FOXO1

v

i

v t

i PUM2

t

a l

a c-JUN 0 .2

l

e

e R

R TUB TUB 0 .0 0 .0 t h r N S N 1 MDA-MB-231 cells t o CAL51 cells E A U O g h n S T R -J X e P N c O L F ll u m R N A F 3 'U T R Is o fo rm

E CAL51 cells F

2 .5 1 5 0 0 *

s

l

l e

) G F P 1 s iR N A

c 6

2 .0 ^

G F P 2 s iR N A e 0

v W T i

1 1 0 0 0

( ** s

P U M 1 -1 s iR N A a

r 1 .5 W T + c -J U N S h o rt

v e

P U M 1 -2 s iR N A n b

i W T + c -J U N F L

f m

1 .0 P U M 1 -3 s iR N A o u

J U N -/-

r 5 0 0

n

e

l P U M 2 -1 s iR N A

l J U N -/- + c -J U N S h o rt b

e 0 .5

P U M 2 -2 s iR N A m

C J U N -/- + c -J U N F L u

P U M 2 -3 s iR N A N 0 .0 0 l o S A A A A A A A A r A N N N N N N N N t n R iR iR iR iR iR iR iR iR o H s s s s s s s s c 1 2 1 2 3 1 2 3 P P ------F F 1 1 1 2 2 2 p B A B E e x p r e s s io n c o n s tru c t G G M M M M M M U U U U U U P P P P P P s iR N A tre a tm e n t Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Miles Figure 3 Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

A C E G Lengthened in cancer Shortened in cancer Shortened in cancer Shortened in cancer Gene ΔPRE PRE Gene ΔPRE PRE Gene ΔPRE PRE Gene ΔPRE PRE FOXO1 +1 4 PTEN -1 0 NRAS -1 0 c-JUN -1 3

F O X O 1 P T E N N R A S c -J U N 2 .5 5 F u ll le n g th L o n g S h o rt 5 L o n g S h o rt 1 6 L o n g S h o rt

E xte n d e d

e

e e

2 .0 4 e ** *

s s

s 4

s

a a

** a

a

r r

1 2 r

r

e

e

e

e

f

f

f

i

f

i i

1 .5 3 i

c c ** c 3

c **

u

u

u

u

L L

** * * L **

L 8

**

e e e **

1 .0 2 e 2

v v

** v

i

v

i

i

i

t t

** t

t

a

a

a

l

a

l

l l

e 4

e e

0 .5 1 e 1

R

R

R R

0 .0 0 0 0 r r r 1 2 c 1 2 c 1 2 r 1 2 c M M c M M S S M M S M M S U U U U U U U U P P P P P P P P s h R N A tr e a tm e n t s h R N A tr e a tm e n t s h R N A tr e a tm e n t s h R N A tr e a tm e n t B D F H

F O X O 1 m R N A s ta b ility P T E N m R N A s ta b ility N R A S m R N A s ta b ility c -J U N m R N A s ta b ility

A

A

A

A

N

N N N 1 .0 1 .2 1 .0

R 1 .2

R

R R

m **

m m

* m

**

e

e e e **

s **

s

s

s

a

a

a

a

r

r r

0 .8 r 0 .8

e

e e

e **

f

f

f

f

i

i

i

i

c

c c

c 0 .5 0 .5

u

u

u

u

L

L

L

L

e

e e

0 .4 e 0 .4

v

v

v

v

i

i

i i

t S h o rt

t t

E xte n d e d t S h o rt S h o rt

a

a

a

a

l

l l

l L o n g L o n g

e

e e

e F u ll le n g th L o n g

R R R R 0 .0 0 .0 0 .0 0 .0 r r r r r r r r r r r r h h h h h h h h h h h h 0 2 6 0 2 6 0 2 6 0 2 6 DownloadedT im e (h rfrom) cancerres.aacrjournals.orgT im one September (h r) 25, 2021. © 2016 AmericanT i Associationm e (h r) for Cancer T im e (h r) Research. Miles Figure 4 Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

A B K i6 7 le v e ls C L y m p h n o d e p o s itiv e A P A a n d tu m o r s iz e 5 2 .0 * 4 2

* s

r o ) *

4 1 .5 m

m *

u

c

s

t l

( 2 8

f e

e 3

v

o

z

e

i l

1 .0 e

s

g

7

r

a

6 i

2 t

o n

K 1 4

m e

u 0 .5

c r

T 1

e p Relative Expression Expression Relative

0 0 .0 0 A A A A A A A A A P P P P P P P P P A A -A A A -A A A -A + + n + + + + n n 2 3 o 2 3 o 2 3 o n n n e e e e e e d d + d + d d + a a a d r r 3 r a 3 ra ra e r d G G e G G e G G a d d r a a r r G G G Miles Figure 5 T u m o r g r a d e T u m o r g r a d e a n d A P A s ta tu s T u m o r g r a d e

E F T rip le -n e g a tiv e b r e a s t tu m o rs G M D A -M B -2 3 1 G ra d e 3 + n o n -A P A E2F4 NRAS c-JUN

l G ra d e 3 + A P A

a 1 .5

2 0 3 .5 G ra d e 2 l

e

m

n

i

v

o

x

i

e

l

o s 3 .0 **

r 0

s

n

P

e

o

/

i r

l 2 .5 1 .0

s

a

p

t s

-2 0 x

s

e e i **

2 .0 r

D *

e p

**

n

x

n

i e

-4 0 1 .5 e

g 0 .5

e ** e

N R A S

g

v e

1 .0 i

n v

JU N t i

a -6 0

a

t

l h

F A M 1 1 0 a C D S 1 U T R 1 e

l 0 .5

C e

T M E M 1 1 9 R C D S 2 U T R 2

R 0 .0 0 .0 3 3 3 R 1 2 1 A A A N H 5 9 8 1 1 1 1 2 3 L 4 L 6 7 1 2 T 3 N F N F N F C M M D 2 5 6 P N C F F F 3 F 4 F F F F 2 F T T T S U U P I P P S S S F S F S S T T F T D D D B P Im Im Im F S S S P P A B B P P P S P S P P S S S S C C C C C C A F F F C C C P C P C C C C T C p p p G C C C A A P P P C C C

s h RDownloadedN A tr e a tm frome n cancerres.aacrjournals.orgt P o ly -a d e n ony l aSeptembertio n m a 25,c h i2021.n e ry © g 2016e n e American e x p re s Associations io n for CancerE x p r e s s io n c o n s tr u c t Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

A B C D G F P 1 s iR N A C S T F 3 -1 s iR N A

) G F P 2 s iR N A C S T F 3 -2 s iR N A 2 1 .2 1 1 g 1 0 C S T F 3 -3 s iR N A

o 3

s

s

l

l

l

(

e e

1 )

s

v

v

6

l

e

e

e

^

l

l

1 0

v

0

n

3 e

0 .8 1

l 9

(

o F

2

i *

r

T

n

s

e

S

o

s

i b

e 9 C

s **

r

m

s

p

e

e

u x

v 0 .4

r i

8 n

e 1

t

p

l

a

l

3 x

8 l

e

e

F

e

T

C

3

R

S

F C T 0 .0

7 7 0 S

C 1 2 1 2 3 N O N -T N B C T N B C 3 0 9 - - - -3 0 9 - 2 4 P P 3 3 3 2 4 R - F F F F F R - 5 -5 T - B T G G T T T B C a n c e r c e ll lin e s - B T - B T S S S K B K B C C C S S B re a s t C a n c e r C e ll lin e s iR N A tre a tm e n t B re a s t C a n c e r C e ll lin e

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Miles Figure 6 Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

A Miles Figure 7 CFI complex CSTF complex PRE MOTIF: UGUAXAUA NANOS 70bp 70bp

Poly-A site UGUA GU Rich GUUGU

70 60 50 40 30 20 10 0 -10 -20 -30 -40 30 20 10 0 -10 -20 -30 -40 -50 -60 -70 CFI CSTF PUM NANOS

B Limited levels of the polyadenylation machinery in normal cells restrict polyadenylation is distal poly-A sites

CFI CSTF Poly-A SITE Full length 3’UTR CFI CSTF CFI GENE GENE AAAA PRE NRE UGUA GU PRE NRE UGUA

Elevated levels of the polyadenylation machinery in tumors cells promote alternative polyadenylation by utilizing PRE and NRE sites

CFI CSTF CSTF CSTF Full length and truncated 3’UTRs

CFI GENE AAAA CFI CSTF CFI CSTF PRE NRE UGUA GENE PRE NRE UGUA GU CFI GENE PRE AAAA UGUA

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 10, 2016; DOI: 10.1158/0008-5472.CAN-16-0844 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Alternative polyadenylation in triple-negative breast tumors allows NRAS and c-JUN to bypass PUMILIO post-transcriptional regulation

Wayne O Miles, Antonio Lembo, Angela Volorio, et al.

Cancer Res Published OnlineFirst October 10, 2016.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-16-0844

Supplementary Access the most recent supplemental material at: Material http://cancerres.aacrjournals.org/content/suppl/2016/10/08/0008-5472.CAN-16-0844.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/2016/10/08/0008-5472.CAN-16-0844. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 25, 2021. © 2016 American Association for Cancer Research.