16
Tumor and Stem Cell Biology Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of Mammary Epithelial Cells PengXu Qian 1 , Arindam Banerjee 4 , Zheng-Sheng Wu 1,2,3 , Xiao Zhang 1 , Hong Wang 1 , Vijay Pandey 5 , Wei-Jie Zhang 1 , Xue-Fei Lv 1 , Sheng Tan 1 , Peter E. Lobie 5 , and Tao Zhu 1 Abstract A discontinuous pattern of LOH at chromosome 3p has been reported in 87% of primary breast cancers. Despite the identication of several tumor suppressor genes in this region, there has yet to be a detailed analysis of noncoding RNAs including miRNAs in this region. In this study, we identied 16 aberrant miRNAs in this region and determined several that are frequently lost or amplied in breast cancer. miR-128-2 was the most commonly deleted miRNA. Embedded in the intron of the ARPP21 gene at chromosome 3p22.3, miR- 128-2 was frequently downregulated along with ARPP21 in breast cancer, where it was negatively associated with clinicopathologic characteristics and survival outcome. Forced expression of miR-128 impeded several oncogenic traits of mammary carcinoma cells, whereas depleting miR-128-2 expression was sufcient for oncogenic transformation and stem cell-like behaviors in immortalized nontumorigenic mammary epithelial cells, both in vitro and in vivo. miR-128-2 silencing enabled transforming capacity partly by derepressing a cohort of direct targets (BMI1, CSF1, KLF4, LIN28A, NANOG, and SNAIL), which together acted to stimulate the PI3K/AKT and STAT3 signaling pathways. We also found that miR-128-2 was directly downregulated by SNAIL and repressed by TGF-b signaling, adding 2 additional negative feedback loops to this network. In summary, we have identied a novel TGF-b/SNAIL/miR-128 axis that provides a new avenue to understand the basis for oncogenic transformation of mammary epithelial cells. Cancer Res; 72(22); 603650. Ó2012 AACR. Introduction Mammary epithelial oncogenic transformation and progres- sion is a multistep process that purportedly involves varied genomic alterations and epigenetic modications (1). These aberrations activate oncogenes, inactivate tumor suppressor genes (TSG), and fundamentally convert a normal cell into a symptomatic cancer cell clone, which is rendered with growth advantage and expands in an unregulated manner, leading to local invasion and distant metastasis (2). In the past 4 decades, signicant progress has been made in the discovery of chro- mosomal aberrations in human breast cancer. In familial breast cancer, BRCA1 at 17q21 and BRCA2 at 13q1213 have been isolated as breast cancer susceptibility genes, and their mutations account for the majority of hereditary breast cancer (3). LOH has been discovered on chromosomes 3p, 6q, 7p, 11q, 16q, and 17p in breast cancer, indicating TSGs may harbor in these regions (4). Of special interest, 87% of primary breast cancers exhibit a discontinuous pattern of LOH at chromo- somes 3p and several genes in this region have been identied as TSGs, such as DUTT1 and FHIT (5). However, although large- scale genomic and molecular genetic technologies allow us to efciently identify numerous breast cancer-related protein- coding genes in the abnormal chromosomal regions, detailed analyses of noncoding RNAs locating in these regions are largely not determined. miRNAs are small, endogenous noncoding RNAs consisting of 21 to 24 nucleotides, which repress gene expression post- transcriptionally via recognizing complementary target sites in the 3 0 UTRs of cognate mRNAs (6). Accumulating evidence has revealed that miRNAs are involved in the regulation of varied biologic processes and their dysregulation is associated with various human diseases, including cancer (7). It has been estimated that more than half of known human miRNAs reside in, or close to, fragile chromosomal sites that are susceptible to Authors' Afliations: 1 Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Tech- nology of China, Hefei, Anhui; 2 Department of Pathology, Anhui Medical University, Hefei, Anhui; 3 Department of Pathology, Shanghai Medical College, Fudan University, Shanghai, P.R China; 4 Liggins Institute, Uni- versity of Auckland, Auckland, New Zealand; and 5 Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, Singapore Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Authors: Tao Zhu, Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui 230027, P.R China. Phone: 86-551- 3602461; Fax: 86-551-3601505; E-mail: [email protected]; and Peter E. Lobie, Cancer Science Institute of Singapore, National University of Singapore, Centre for Life Sciences, #03-06C, 28 Medical Drive, Singapore 117456. E-mail: [email protected] doi: 10.1158/0008-5472.CAN-12-1507 Ó2012 American Association for Cancer Research. Cancer Research Cancer Res; 72(22) November 15, 2012 6036 on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

Tumor and Stem Cell Biology

Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3Targets Multiple Stem Cell Factors to PromoteTransformation of Mammary Epithelial Cells

PengXu Qian1, Arindam Banerjee4, Zheng-Sheng Wu1,2,3, Xiao Zhang1, Hong Wang1, Vijay Pandey5,Wei-Jie Zhang1, Xue-Fei Lv1, Sheng Tan1, Peter E. Lobie5, and Tao Zhu1

AbstractA discontinuous pattern of LOH at chromosome 3p has been reported in 87% of primary breast cancers.

Despite the identification of several tumor suppressor genes in this region, there has yet to be a detailedanalysis of noncoding RNAs including miRNAs in this region. In this study, we identified 16 aberrant miRNAsin this region and determined several that are frequently lost or amplified in breast cancer. miR-128-2 was themost commonly deleted miRNA. Embedded in the intron of the ARPP21 gene at chromosome 3p22.3, miR-128-2 was frequently downregulated along with ARPP21 in breast cancer, where it was negatively associatedwith clinicopathologic characteristics and survival outcome. Forced expression of miR-128 impeded severaloncogenic traits of mammary carcinoma cells, whereas depleting miR-128-2 expression was sufficient foroncogenic transformation and stem cell-like behaviors in immortalized nontumorigenic mammary epithelialcells, both in vitro and in vivo. miR-128-2 silencing enabled transforming capacity partly by derepressing acohort of direct targets (BMI1, CSF1, KLF4, LIN28A, NANOG, and SNAIL), which together acted to stimulatethe PI3K/AKT and STAT3 signaling pathways. We also found that miR-128-2 was directly downregulatedby SNAIL and repressed by TGF-b signaling, adding 2 additional negative feedback loops to this network.In summary, we have identified a novel TGF-b/SNAIL/miR-128 axis that provides a new avenue tounderstand the basis for oncogenic transformation of mammary epithelial cells. Cancer Res; 72(22);6036–50. �2012 AACR.

IntroductionMammary epithelial oncogenic transformation and progres-

sion is a multistep process that purportedly involves variedgenomic alterations and epigenetic modifications (1). Theseaberrations activate oncogenes, inactivate tumor suppressorgenes (TSG), and fundamentally convert a normal cell into asymptomatic cancer cell clone, which is rendered with growthadvantage and expands in an unregulated manner, leading to

local invasion and distant metastasis (2). In the past 4 decades,significant progress has been made in the discovery of chro-mosomal aberrations in human breast cancer. In familialbreast cancer, BRCA1 at 17q21 and BRCA2 at 13q12–13 havebeen isolated as breast cancer susceptibility genes, and theirmutations account for the majority of hereditary breast cancer(3). LOH has been discovered on chromosomes 3p, 6q, 7p, 11q,16q, and 17p in breast cancer, indicating TSGs may harbor inthese regions (4). Of special interest, 87% of primary breastcancers exhibit a discontinuous pattern of LOH at chromo-somes 3p and several genes in this region have been identifiedas TSGs, such asDUTT1 and FHIT (5). However, although large-scale genomic and molecular genetic technologies allow us toefficiently identify numerous breast cancer-related protein-coding genes in the abnormal chromosomal regions, detailedanalyses of noncoding RNAs locating in these regions arelargely not determined.

miRNAs are small, endogenous noncoding RNAs consistingof 21 to 24 nucleotides, which repress gene expression post-transcriptionally via recognizing complementary target sites inthe 30UTRs of cognate mRNAs (6). Accumulating evidence hasrevealed that miRNAs are involved in the regulation of variedbiologic processes and their dysregulation is associated withvarious human diseases, including cancer (7). It has beenestimated thatmore than half of known humanmiRNAs residein, or close to, fragile chromosomal sites that are susceptible to

Authors' Affiliations: 1Hefei National Laboratory for Physical Sciences atMicroscale and School of Life Sciences, University of Science and Tech-nology of China, Hefei, Anhui; 2Department of Pathology, Anhui MedicalUniversity, Hefei, Anhui; 3Department of Pathology, Shanghai MedicalCollege, Fudan University, Shanghai, P.R China; 4Liggins Institute, Uni-versity of Auckland, Auckland, NewZealand; and 5Cancer Science Instituteof Singapore and Department of Pharmacology, National University ofSingapore, Singapore

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

Corresponding Authors: Tao Zhu, Hefei National Laboratory for PhysicalSciences at Microscale and School of Life Sciences, University of Scienceand Technology of China, Hefei, Anhui 230027, P.R China. Phone: 86-551-3602461; Fax: 86-551-3601505; E-mail: [email protected]; and PeterE. Lobie, Cancer Science Institute of Singapore, National University ofSingapore, Centre for Life Sciences, #03-06C, 28Medical Drive, Singapore117456. E-mail: [email protected]

doi: 10.1158/0008-5472.CAN-12-1507

�2012 American Association for Cancer Research.

CancerResearch

Cancer Res; 72(22) November 15, 20126036

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 2: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

deletion, amplification, or translocation in the establishmentand progression of tumor (8). Moreover, systematic miRNAexpression profilings have observed that characteristic miRNAsignatures are associated with prognosis and progression ofcertain human cancers (7, 9).Our recent study (10) observed significantly diminished

expression of let-7g, embedded in the WDR82 gene on chro-mosome 3p21.1, in breast cancer comparedwith normal tissue.Previous studies have consistently reported that LOH isfrequently discovered in primary breast cancers at the chro-mosome 3p allele, especially at 3p21.3, 3p22–24, 3p21.2–3p21.3,3p25, 3p14.2, 3p14.3, and 3p12 (5, 11, 12). For these reasons,we undertook to uncover specific miRNAs in these aberrantregions, with the hope that these breakpoint-associatedmiRNAs may provide insights into the causal mechanisms ofbreast cancer development.

Materials and MethodsCell culturesAll human breast cancer cell lines and nontumorigenic

human breast epithelial cell lines used in this study wereobtained from the American Type Culture Collection (ATCC)and cultured in conditions as ATCC recommended.

Cell function assaysAll functional assays such as total cell number, soft agar

colony formation assay, growth on 2-dimensional or in 3-dimensionalmatrigel, in vitro cellmotility (migration, invasion,and wound healing) assays were carried out as described (13).For monolayer cell proliferation, 3,000 cells were seeded into100 mL media (2% FBS) and viability was quantified every 24hours. Colony scattering assays were carried out as previouslyreported (14). All the images were taken under a microscope(Olympus).

Protein extraction and Western blot analysis, RNAextraction and quantitative reverse transcription-PCRThese procedures were carried out as described previously

(10). The sequences of primers used in the real-time PCRexperiments and the characteristics of the antibodies are listedin Supplementary Tables S7 and S8, respectively.

ResultsAberrant miRNAs on chromosomal 3p in humanmammary carcinomaTo screen for miRNAs within the aberrant regions at the

chromosome 3p allele, especially at 3p21.3, 3p22–24, 3p21.2–3p21.3, 3p25, 3p14.2, 3p14.3, and 3p12, we initially searched themiRbase 18.0 database (15) on the basis of their precise loca-tions on the human chromosome and identified 34 presentlyknownmiRNAs (Supplementary Table S1).We then carried outgenomic real-time PCR to determine changes in DNA copynumbers in 20 archived breast cancer specimens comparedwith 4 breast tissue specimens frompatientswithbenignbreastdiseases (normal; Supplementary Fig. S1A–F). As summarizedin Fig. 1A, we uncovered 11 novel miRNAs with genome copylosses (beyond 20% of the tested cases) in breast cancer.Intriguingly, we also detected 5 miRNAs with obvious genome

copy gains. Among the 11 frequently lostmiRNAs, we identifiedmiR-128-2 as the most frequently lost miRNA (deleted in 75%cases; Fig. 1A), even higher than let-7g (60% deletion rate). Thisobservation attracted our interest to determine the specificfunctional roles and regulatory mechanisms of miR-128-2.

miR-128-2 is frequently downregulated together withARPP21 in human breast cancer and is associated withpoor survival outcome

miR-128-2 is an intronic gene, which resides within the 17thintron of the ARPP21 gene on human chromosome 3p22.3 (Fig.1B). The 21-kDa cAMP-regulated phosphoprotein (ARPP21),also known as Regulator of Calmodulin Signaling, which isenriched in brain, has been described as a candidate TSGand is frequently deleted in mammary carcinoma (16). We firstdetected the expression of ARPP21 using quantitative reversetranscription (qRT)-PCRandobserveda significant reduction inbreast cancer (n ¼ 21) compared with normal specimens (n ¼15; Fig. 1C). To assess whether deregulation of miR-128-2expression is due to loss of DNA copy number, we nextexamined the expression levels of pri-miR-128-2 and maturemiR-128.We found that bothpri-miR-128-2 andmaturemiR-128were remarkably downregulated (Fig. 1D and E), and theirexpressions were significantly concordant with that of the hostgene ARPP21 (Supplementary Fig. S1G and H), indicating thatmiR-128-2 is often co-expressed with ARPP21.

To further explore the clinicopathologic implications ofmiR-128 in the progression of breast cancer, we determinedthe specific abundance of mature miRNA-128in breast cancerspecimens (n ¼ 85) and normal breast tissue specimens (n ¼18) by in situ hybridization (ISH). We observed significantlydecreased expression of miR-128 in mammary carcinomacompared with normal specimens (P ¼ 0.039, Fig. 1F andG). In normal mammary tissue miR-128 was predominantlylocalized to carcinoma cells with limited expression in stroma.Moreover, we further analyzed the relationships between miR-128 expression and clinicopathologic characteristics of breastcancer and the results are summarized in SupplementaryTable S2. A statistically significant association was observedbetween lower expression of miR-128 and clinical stage (P ¼0.036), lymph node metastasis (P ¼ 0.020) and Ki-67 labelingindex (P ¼ 0.027).

Wenext examined the relationship betweenmiR-128 expres-sion and survival in the breast cancer patient cohortwith 5-yearfollow-up (n ¼ 85) using Kaplan–Meier survival analyses. Wefound that patients with lower expression levels of miR-128 exhibited significantly worse survival outcomes comparedwith thosewithhigher expression levels ofmiR-128 (Fig. 1HandI, Supplementary Table S3). Thus, these results showed thatmiR-128-2 is frequently downregulated together with ARPP21in human breast cancer and the expression level of miR-128may be a potential prognostic indicator for patient survival.

Forced miR-128 expression reduces oncogenicity,migration and invasion of mammary carcinoma cells invitro

To determine the functionality of miR-128 in breast cancercells, we first carried out a preliminary screen for expression

miR-128-2 as a Tumor Suppressor in Breast Cancer

www.aacrjournals.org Cancer Res; 72(22) November 15, 2012 6037

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 3: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

*

ID Chromo

some Change of

genome copy Freque

ncy, %

hsa-mir-128-2 3p22.3 Deletion 75%

hsa-let-7g 3p21.1 Deletion 60%

hsa-mir-4792 3p24.2 Deletion 35%

hsa-mir-26a-1 3p22.2 Deletion 35%

hsa-mir-5193 3p21.31 Deletion 35%

hsa-mir-3938 3p14.3 Deletion 35%

hsa-mir-885 3p25.3 Deletion 25%

hsa-mir-138-1 3p21.32 Deletion 25%

hsa-mir-564 3p21.31 Deletion 25%

hsa-mir-135a-1 3p21.1 Deletion 20%

hsa-mir-4273 3p12.3 Deletion 20%

hsa-mir-4270 3p25.1 Amplification 80%

hsa-mir-4444-2 3p12.3 Amplification 70%

hsa-mir-425 3p21.31 Amplification 65%

hsa-mir-191 3p21.31 Amplification 65%

hsa-mir-5688 3p12.1 Amplification 25%

A B

C

D

ARPP21

3p22.3

Chr3

Exon 17 Exon 18 Intron 17

hsa-miR-128-2

Months after surgery

20 30 40 50 60 70

1.0

0.8

0.6

0.4

0.2

0.0

P = 0.017

Pro

babili

ty

OS

miR-128 positive

miR-128 negative

Months after surgery

P = 0.023

Pro

babili

ty

RFS

miR-128 positive

miR-128 negative

F

I

Normal

ISH with LNA-miR-128 probe

G

Group n miR-128 expression

Negative, n (%) Positive, n (%)

Normal 18 6 (33.3) 12 (66.7)

Breast cancer 85 51(60.0) 34 (40.0)*

Mammary carcinoma

E

H

0

100

200

300

0

100

200

300

400

Normal N = 15

Breast cancer N = 21

Re

lative

exp

ressio

n o

f R

ela

tive e

xp

ressio

n o

f

AR

PP

21

/GA

PD

H

pri-m

iR-1

28-2

/pre

- β-a

ctin

**

0

1

2

3

4

5

Rela

tive e

xpre

ssio

n o

f

miR

-128/U

6

Normal N = 15

Breast cancer N = 21

*

1.0

0.8

0.6

0.4

0.2

0.0

0 10 20 30 40 50 60 70

NormalN = 15

Breast cancerN = 21

Qian et al.

Cancer Res; 72(22) November 15, 2012 Cancer Research6038

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 4: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

levels of ARPP21, pri-miR-128-2 and mature miR-128 in 2mammary epithelial cell lines and 8 mammary carcinoma celllines by qRT-PCR (Fig. 2A, and Supplementary Fig. S2A). Weobserved remarkably higher expression levels of ARPP21, pri-miR-128-2 and mature miR-128 in 2 immortalized but other-wise normal HMECs, in contrast to lower or undetectableexpression levels inmammary carcinoma cell lines. In addition,we observed a strong correlation between the expression levelof mature miR-128 and ARPP21 (Pearson coefficient¼ 0.8749)and a further stronger correlation between the expression levelof pri-miR-128-2 and ARPP21 (Pearson coefficient ¼ 0.9689;Supplementary Fig. S2B and C).We next assessed the potential effect of miR-128 on the

phenotype of theMDA-MB-231 cells by transfecting a syntheticmiRNA mimic (Fig. 2B). Forced miR-128 expression decreasedboth the total cell number and colony formationnumber in softagar by approximately 60% compared with cells transfectedwith scrambled control oligonucleotides (Fig. 2B). Smallercolonies and a 2-fold decrease in branching colonies wereconsistently observed with MDA-MB-231 miR-128 cells cul-tured in 3-dimensional matrigel (Fig. 2C). In addition, forcedexpression of miR-128 in MDA-MB-231 cells produced a "cob-ble stone" like epithelial cellular morphology in monolayeradherent culture as compared with the control cells (Fig. 2D).In colony-scattering assays, we observed a significantly largerproportion of compact cells (3-fold) and smaller proportion ofscattered cells (3-fold) with forced expression of miR-128 inMDA-MB-231 cells (Fig. 2E). Finally, forced expression of miR-128 in MDA-MB-231 cells led to a retarded wound closing (Fig.2F) and reduction in cell migration by 3-fold and invasion by2.5-fold compared with the control cells (Fig. 2G).Conversely, miR-128 depletion in mammary epithelial MCF-

10A cells by transfection of specific ASO (Supplementary Fig.S2D) significantly increased both the total cell number andcolony formation number in soft agar compared with cellstransfected with control oligonucleotides (Supplementary Fig.S2D). Moreover, miR-128 depletion inMCF-10A cells increasedthe number of branching colonies by 3-fold in matrigel (Sup-plementary Fig. S2F). Furthermore, MCF-10A cells withreduced expression of miR-128 exhibited a spindle like anda more scattered, elongated, and mesenchymal cellular mor-phology in monolayer adherent culture compared with thecontrol cells (Supplementary Fig. S2E). In colony-scatteringassays, we observed a 4-fold larger proportion of scattered cellsand 2-fold smaller proportion of compact cells in MCF-10Acells with diminished expression of miR-128 (SupplementaryFig. S2G). Finally, miR-128 depletion in MCF-10A cells resultedin a more rapid wound closing (Supplementary Fig. S2H), andan obvious augmentation in cell migration by 2-fold and

invasion by 2.5-fold compared with the control cells (Supple-mentary Fig. S2I).

To determine whether miR-128 impacts on EMT, we exam-ined the expression of epithelial and mesenchymal markers byWestern blotting. MDA-MB-231 cells transfected withmiR-128expressed higher levels of the epithelial markers, E-cadherinand plakoglobin (g-catenin) and lower levels of the mesenchy-mal markers, FN1 and vimentin. Conversely, MCF-10A cellstransfected with miR-128 ASO expressed lower levels of epi-thelial markers and higher levels of mesenchymal markers(Supplementary Fig. S2J).

To exclude cell type specific effects, the parallel experimen-tal approaches were deployed inMCF-7 with forced expressionof miR-128 and conversely with HBL-100 with reduced expres-sion of miR-128. We observed concordant effects in the dif-ferent 2 cell lines (Supplementary Fig. S3A-L). Besides, we usedTERT immortalized HMEC cells as another model of normalimmortalized mammary epithelial cells (17). We infectedTERT-HMEC cells with pBabe-miR-128 sponge or controlretroviruses and carried out colony formation assay. Weobserved larger colonies and a 3-fold increase in colony num-ber of TERT-HMEC cells transfected with pBabe-miR-128sponge compared with control cells (Supplementary Fig.S3M). Thus, these data indicate that forced expression ofmiR-128 dramatically alters the morphology and impairsoncogenicity and invasiveness of mammary carcinoma cellsin vitro.

Forced miR-128 expression impairs mammarycarcinoma xenograft growth in vivo

To further determine the function of miR-128 in mammarycarcinoma cells in vivo, we constructed 1 retroviral vectorexpressing miR-128 by harboring pri-miR-128-2, which wastransduced in MDA-MB-231 cells, and another vector inhibit-ing miR-128 expression by inserting a cassette of bulged miR-128 sequences as described (18), which was transduced innontumorigenic MCF-10A. The 2 stable cell lines, denotedMDA-MB-231 pri-miR-128-2 exhibited elevated miR-128expression by 2.8-fold and MCF-10A miR-128 sponge reducedmiR-128 expression by 4-fold, respectively (Fig. 3A).

We next injected 5 � 106 MDA-MB-231 pri-miR-128-2 cellsand MDA-MB-231 control cells subcutaneously into the flanksof female immunodeficient nude mice. Both groups formedpalpable and measurable tumors, however, with a 5.2-foldincrease of miR-128 expression, tumors formed by MDA-MB-231 pri-miR-128-2 were smaller than those formed bycontrol cells (Fig. 3B and C). Tumors formed by MDA-MB-231 pri-miR-128-2 cells were well confined and differentiated(upper right panel, Fig. 3D), whereas those formed by MDA-

Figure 1. miR-128 andARPP21 are frequently lost and inversely associatedwith poor prognosis. A, list of aberrantmicroRNAs (deleted or amplified) on humanchromosome 3p in breast carcinoma determined by genomic real-time PCR. B, schematic diagram of the genomic locus of hsa-miR-128-2 (Homo sapiens-miR-128-2).C, expression level of ARPP21 in breast cancer specimens (n ¼ 21) and benign breast disease tissues (Normal; n ¼ 15) were analyzed byqRT-PCR. GAPDH was used as an internal control. D, expression level of pri-miR-128-2 in breast cancer specimens (n ¼ 21) and benign breast diseasetissues (n¼15)weremeasuredbyqRT-PCR. b-actinwasused as an internal control. E,miR-128 expressionwasdeterminedbyqRT-PCR in breast carcinomaspecimens (n¼ 21) and benign breast disease tissues (n¼ 15). U6 (U6 small nuclear RNA) was used as an internal control. F, expression level of miR-128 wasexamined inmammary carcinoma specimens and normalmammary specimens. One representative example of tissuemicroarray sections analyzed by in situhybridization is shown. G, expression of miR-128 in human breast cancer specimens and normal breast tissues. H and I, correlations between the expressionlevel of mature miR-128 and 5-year patient overall survival (OS) analysis or relapse-free survival (RFS) by Kaplan–Meier analysis.

miR-128-2 as a Tumor Suppressor in Breast Cancer

www.aacrjournals.org Cancer Res; 72(22) November 15, 2012 6039

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 5: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

A

0

0.2

0.4

0.6

0.8

1

1.2

1.4

1.6

1.8

MCF-10A HBL100 ZR75-1 SKBR3 T47D MCF-7 BT549 MDA-MB

-468

MDA-MB

-231

MDA-MB

-435s

HMECs

Breast cancer cell lines

P = 0.002 R

ela

tive e

xpre

ssio

n o

f

miR

-128/U

6

B

C E

D

G

Control miR-128

0 h

22 h

MDA-MB-231

F

0

0.2

0.4

0.6

0.8

1

1.2

1.4

Migration Invasion

Control

miR-128

Fold

-mig

ration/invasio

n

*** ***

100× 200×

Co

ntr

ol

miR

-128

400×

Monolayer adherent morphology

3D Matrigel assay

0

50

100

150

200

Control miR-128

Num

ber

of

bra

nchin

g

colo

nie

s p

er

200 c

ells

**

100×

200×

Control miR-128

Perc

enta

ge o

f colo

ny

types

0%

20%

40%

60%

80%

100%

120%

Control miR-128

Compact Loose Scattered

**

**

Colony-scattering assay

Wound healing assay Transwell assay

Fold

-change

**

** *** 0

1

98

miR-128 Expression

Total cell number

Colony

formation

Control

miR-128

124

MDA-MB-231

Qian et al.

Cancer Res; 72(22) November 15, 2012 Cancer Research6040

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 6: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

MB-231 control cells were poorly encapsulated and highlyinvasive (upper left panel, Fig. 3D). Furthermore, local infil-tration into muscle, and tumor emboli in blood vessels, wereobserved in tumors formed by control cells (upper leftpanel, Fig. 3D). Moreover, pulmonary metastases were readilydetectable in the lungs of host mice 3.5 weeks after subcuta-neous injectionwithMDA-MB-231 control cells. In contrast, nometastases were detected in mice injected with MDA-MB-231pri-miR-128-2 cells (Fig. 3D). In addition, Ki-67 or PCNAstaining and TUNEL or Caspase-3 assays were carried out ontumor sections, and the MDA-MB-231 pri-miR-128-2 tumorsexhibited fewer Ki-67-labeled or PCNA-labeled cells (Fig. 3E,Supplementary Fig. S4A) and higher percentage of TUNEL-positive or Caspase-3-labeled cells (Fig. 3F, Supplementary Fig.S4B) compared with MDA-MB-231 control tumors.Alternatively, 5 � 106 MCF-10A miR-128 sponge cells and

MCF-10A control cells were subcutaneously inoculated intothe flanks of host mice. Consistent with prior reports (19), thenontumorigenic MCF-10A cell line is not capable of initiatingtumor formation. However, MCF-10A miR-128 sponge cellsreadily formed visible andmeasurable tumors (SupplementaryFig. S4C and D), suggesting that miR-128 depletion promotedtumor formation in vivo.

Forced expression of miR-128 represses mammarycarcinoma stem cell-like behaviorDysregulated cancer stem cells have been postulated to be

crucial for cancer initiation and progression (20). By extension,we wondered whether miR-128 could influence cancer stemcell-like behavior. We therefore transiently transfected miR-128 mimics into MDA-MB-231 cells and miR-128 ASO intoMCF-10A cells. We then cultured these cells in conditionedmammosphere culture media in ultra-low attachment 6-wellplates. MDA-MB-231 cells with forced expression of miR-128exhibited both significantly decreased number (43%) and sizeof mammospheres compared with control cells (Fig. 4A).Conversely, we observed a slightly yet significant increase(2.6-fold) in mammospheric growth of MCF-10A cells withmiR-128 depletion (Fig. 4A). To exclude the possibility that theincrease inmammosphere number arose from the aggregationof quiescent cells, we carried out self-renewal assays. Weobserved decreased numbers of secondary and tertiary mam-mospheres generated from MDA-MB-231 cells with increasedmiR-128 expression, whereas MCF-10A cells with decreasedmiR-128 expression exhibited increased numbers of secondaryand tertiary mammospheres compared with their correspond-

ing control cells (Fig. 4B). Recent studies have identifiedALDH1 enzymatic activity as a potential marker for breastcancer stem cells (21). To this end, we carried out the ALDE-FLUOR assay to determine whether miR-128 modulated theALDH1 positive cell population. Forced expression of miR-128in MDA-MB-231 markedly reduced the percentage of ALDH1positive cells by 3-fold whereas depletion of miR-128 expres-sion increased the percentage of ALDH1 positive cells byapproximately 3.5-fold (Fig. 4C). Moreover, we have sortedMDA-MB-231-ALDH1þ and MDA-MB-231-ALDH1- popula-tions, and observed decreased expression of miR-128, CD44and CD133 and increased expression of CD24 in the MDA-MB-231-ALDH1þ population comparing with MDA-MB-231-ALDH1- population (Supplementary Fig. S4E and F). Wenext determined whether miR-128 modulated the CD44þ

CD24neg/low population. Forced expression of miR-128 inMDA-MB-231 reduced the percentage of CD44þCD24neg/low

population by 1.5-fold whereas depletion of miR-128 expres-sion increased the percentage of CD44þCD24neg/low populationby 1.3-fold (Supplementary Fig. S4G).

To ascertain the influence of miR-128 on tumor-initiatingcapability in vivo, we subcutaneously injected MDA-MB-231-pri-miR-128-2, MCF-10A miR-128 sponge cells and their cor-responding control cells at a series of limiting dilutions from5 � 105 to 5 � 103 cells into nude mice and identified theirability to form visible tumors. After an incubation period of 6weeks, as summarized in Fig. 4D, in mice injected with 5� 104

cells, the tumor incidence derived fromMDA-MB-231 pri-miR-128-2 cells (4/6) was lower than control cells (6/6). Strikingly,in mice injected with 5 � 103 cells, 4/6 mice inoculatedwith MDA-MB-231 control cells formed tumors, whereas notumor was detected in mice inoculated withMDA-MB-231 pri-miR-128-2 cells. Conversely, no tumors were observed in miceinjected with any numbers of MCF-10A control cells or 5� 103

MCF-10AmiR-128 sponge cells. However, tumors were formedin mice injected with 5 � 105 MCF-10A miR-128 sponge cells(2/6) and 5 � 104 MCF-10A miR-128 sponge cells (1/6),suggesting that depletion of miR-128 expression confers nor-mal mammary epithelial MCF-10A cell with tumor-initiatingcapacity. Collectively, these observations indicate thatmiR-128dramatically interfere withmammary carcinoma stem cell-likebehavior, both in vitro and in vivo.

To determine a potential association between miR-128 andALDH1 expression in patients with breast cancer, we carriedout qRT-PCR on a cohort of 24 breast cancer specimens todetermine the expression levels of miR-128 and ALDH1mRNA.

Figure 2. Forced expression of miR-128 alters the morphology and reduces migration and invasion of MDA-MB-231 cells in vitro. A, expression levels of miR-128 in 10humanmammary epithelial or carcinomacell lineswereanalyzedbyqRT-PCR.The resultwasnormalized toU6small nuclearRNA.B, left, expressionlevels of miR-128 inMDA-MB-231 cells transiently transfectedwithmiR-128mimic. Middle, total cell number assays. 5� 103MDA-MB-231 cells transfectedwithmiR-128or control were seededwith 10%FBSand counted after 6 days. Right, soft agar assay. 2� 103 cellswere seeded in 0.35%agarose and coloniesformed were counted after incubation for 14 days. C, branching morphogenesis after cell growth in 3D Matrigel for 10 days was observed undera phase-contrast microscope. The number of branching colonies per 200 cells was counted. D, monolayer adherent morphology of MDA-MB-231 cells withforced expression of miR-128. Cultures were imaged at approximately 40% confluence and at�100,�200, and�400 magnifications. E, 1� 103 MDA-MB-231-miR-128 cells or control cells were seeded in 10-cm petri dish. Colonies formed by each cell type, were categorized and counted as per the extent ofcolony scattering. Percentages of each colony category in the total counts were plotted as indicated. F, wound-healing assay of MDA-MB-231 cellstransfected with miR-128 mimic or scrambled sequence oligonucleotides. Magnification, �100. G, transwell migration assay and Matrigel invasion assay ofMDA-MB-231 cells transfectedwithmiR-128mimic or scrambled sequences. A representative experiment is shown in triplicate along asmean�SEM inA, B,C, E, and G. ��, P < 0.01; ���, P < 0.001 (Student t test).

miR-128-2 as a Tumor Suppressor in Breast Cancer

www.aacrjournals.org Cancer Res; 72(22) November 15, 2012 6041

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 7: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

We observed that the expression of miR-128 was inverselyassociated with ALDH1 mRNA (Pearson coefficient ¼�0.4065; Fig. 4E). In addition, we carried out IHC staining ona cohort of 47 breast cancer specimens to determine the

expression levels of ALDH1. Combined with the expressionlevels of miR-128 by ISH, we observed that the expression ofmiR-128 was inversely and significantly associated withALDH1 expression (Pearson coefficient ¼ �0.327, P ¼ 0.025).

A B

C

MDA-MB-231 pBabe

MDA-MB-231 pri-miR-128-2

D

Fat Pad Injection tumor growth curve T

um

or

volu

me (

mm

3)

(Week)

***

***

***

***

***

0

500

1,000

1,500

2,000

2,500

3,000

3,500

4,000

0 0.5 1 1.5 2 2.5 3 3.5

MDA-MB-231 pBabe

MDA-MB-231 pri-miR-128-2

Rela

tive e

xpre

ssio

n o

f

miR

-128/U

6

**

0

0.5

1

1.5

2

2.5

3

3.5

pBabe pBabe-pri-

miR-128-2

Control miR-128

Sponge

**

MDA-MB-231 MCF-10A

MDA-MB-231 pBABE MDA-MB-231 pBabe-pri-miR-128-2

Pri

ma

ry t

um

or

200×

400×

Lu

ng

meta

sta

sis

200×

400×

50 μm

20 μm

Ki-67 staining E

***

Positiv

e c

ells

(%

)

0

10

20

30

40

50

60

pBabe pri-miR-128-2

pBabe pri-miR-128-2

TUNEL

F

Positiv

e c

ells

(%

) ***

0

5

10

15

20

25

30

pBabe pri-miR-128-2

pBabe pri-miR-128-2

Figure 3. Forced expression ofmiR-128 impairs MDA-MB-231cells xenograft growthand inducesapoptosis in vivo. A, qRT-PCRanalysis of the expression level ofmiR-128 in MDA-MB-231 cellsinfected with pBabe-pri-miR-128-2 or control vector and MCF-10Acells infected with pBabe-miR-128Sponge or scrambled sequenceoligonucleotide. The result wasnormalized to U6 small nuclearRNA. B, tumor growth curve ofMDA-MB-231 control or MDA-MB-231-pri-miR-128-2 cellsimplanted s.c. into the flanks offemale nude mice. C,representative images of tumorsformed in nude mice that receivedsubcutaneous injections of MDA-MB-231 cells transfected with theindicated vectors. D, H&E stainedsections of tumors and lungsisolated from mice that receivedsubcutaneous injection of MDA-MB-231-pri-miR-128-2 or controlcells at week 4. Abundant localinfiltration into the muscle andtumor emboli in blood vesselswere visualized in the margins ofthe primary tumor nodule (upper,red arrow), whereas pulmonarymetastases were readilydetectable in the lungs (bottom,red arrow). Images were taken at�200 and �400 magnification. E,cell proliferation was assessed byKi-67 staining on the sections oftumors. F, apoptosis wasmeasured by TUNEL labeling. Arepresentative experiment isshown in triplicate along asmean � SEM in A, E, and F.��, P < 0.01; ���, P < 0.001(Student t test).

Qian et al.

Cancer Res; 72(22) November 15, 2012 Cancer Research6042

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 8: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

A B

CMDA-MB-231

**

Control miR-128

ALD

H1+

ve c

ells

(%

)

0

50

100

150

MCF10A

**

Control miR-128 ASO

ALD

H1+

ve c

ells

(%

)

0

100

200

300

400

500

Control miR-128 ASO

MDA-MB-231

0

0.2

0.4

0.6

0.8

1

1.2

Control miR-128

Fold

-mam

mosphere

num

bers

*

MCF10A

0

0.5

1

1.5 2

2.5

3

3.5

Control miR-128 ASO

Fold

-mam

mosphere

num

bers

*

MDA-MB-231

G2 G3 G1

*** *

Fold

-mam

mosphere

num

bers

0

0.5

1

1.5

2 Control miR-128

***

MCF-10A

**

0

1

2

3

4

5

6 Control

miR-128 ASO

G2 G3 G1

Fold

-mam

mosphere

num

bers

*

*

0.9%

DEAB

0.0%

Control

0.3%

DEAB

0.0%

miR-128

DEAB 0.0%

0.4%

Control

DEAB

1.6%

0.1%

miR-128 ASO

Control miR-128

D

Cells

Tumors incidence/number of injections

5×106 5×105 5×104 5×103

MDA-MB-231 pBabe 8/8 6/6 6/6 4/6

MDA-MB-231 pri-miR-128-2 8/8 6/6 4/6 0/6

MCF-10A control 0/8 0/6 0/6 0/6

MCF-10A miR-128 Sponge 4/8 2/6 1/6 0/6

R2 = 0.1652

P = 0.02

Pearson coefficient = – 0.4065

Relative expression of miR-128/U6

Rela

tive e

xpre

ssio

n o

f A

LD

H1/G

AP

DH

0

20

40

60

80

100

120

140

160

0 1 2 3 4 5 6

E

Figure 4. miR-128 represses theCSC-like population inmammary carcinoma cells. A,MDA-MB-231 andMCF-10A cells with forced or depleted expression ofmiR-128 were seeded in ultra low attachment plates in mammospheric growth media. After 14 days, growth was measured by AlamarBlue. Representativeimages ofmammosphere generated by either forced expression or depleted expression ofmiR-128 cells ofMDA-MB-231 andMCF-10A. Bar, 50 mm. B,miR-128 inhibited the self-renewal potential of MDA-MB-231 cells (upper), whereas depletion of miR-128 enhanced the self-renewal potential of MCF-10A cells(bottom).MDA-MB-231andMCF-10Acellswith forced or depletedexpressionofmiR-128were grownundermammospheric condition andwere sequentiallycultured from first generation (G1) till third generation (G3) and growth compared with respective control cells. C, miR-128 modulates the ALDH1þcell population in MDA-MB-231 and MCF-10A cells with forced or depleted expression of miR-128. Cells were incubated with ALDEFLUOR substrate(BAAA) and fluorescencewasmeasured by flow cytometry. Cells with DEAB (diethylaminobenzaldehyde), a specific inhibitor of ALDH1, were used as control.D, tumor Incidence of MDA-MB-231 stably transfected with pri-miR-128-2 or MCF-10A stably transfected with miR-128 Sponge and then injectedinto hostmice in limiting dilutions. E,miR-128 is inversely associatedwith ALDH1 in breast cancer samples. Pearson's correlation coefficient was determinedbetween the expression levels of ALDH1 and miR-128 by qRT-PCR in a cohort of 24 breast cancer specimens. A representative experiment is shownin triplicate along as mean � SEM in A, B, and C. �, P < 0.05; ��, P < 0.01(Student t test).

miR-128-2 as a Tumor Suppressor in Breast Cancer

www.aacrjournals.org Cancer Res; 72(22) November 15, 2012 6043

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 9: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

miR-128 directly targets a cohort of stem cell factorsTo determine the mechanism by which miR-128 exerts

tumor suppressor properties, we initially employed 3 algo-rithms to predict the potential mRNA targets of miR-128—MiRanda (22), Pictar (23), and TargetScan (6). On the basis ofthe sequences in their 30 UTRs, 98 mRNAs were predicted to betargets by all of the 3 programs (Fig. 5A, Supplementary TableS4). To further narrow the possible downstream effectors ofmiR-128, we next screened for the mRNA expression profile ofan array of genes, which have been widely known in cancerinitiation and progression, by qRT-PCR in 2 sets of paired cells.(MDA-MB-231 cells transfected with either miR-128 mimics orcontrol and MCF-10A cells transfected with either miR-128ASO or scrambled sequence control). Interestingly, as shown inthe heat map (Fig. 5B), the data derived from the 2 comple-mentary screening systems were highly concordant, and indi-cated that miR-128 modulates numerous key genes function-ally involved in neoplastic progression (Supplementary TableS5). By combination of the 2 strategies, we identified a cohort of6 genes (BMI1, CSF1, KLF4, LIN28A, NANOG, and SNAIL), all ofwhich are known transcriptional regulators implicated instem cell development (24) and contain 30UTR elements thatare partially complementary to miR-128 (Supplementary Fig.S5A-G).

To confirm whether the 6 candidate genes are direct targetsof miR-128, we cloned full-length fragments of their 30UTRsinto the luciferase reporter plasmid psi-CHECK2. We thentransfected themiR-128mimicwith different 30UTRconstructsinto MDA-MB-231 cells and observed that miR-128 markedlyrepressed the relative luciferase activities of the wild-type30UTR of BMI1, CSF1, KLF4, LIN28A, NANOG, and SNAIL (Fig.5C). Furthermore, mutation of the partially complementarymiR-128 site(s) in these 30UTRs abrogated responsiveness tomiR-128 (Fig. 5C). In addition, we measured the effects of miR-128 on expression levels of the 6 targets. Forced expression ofmiR-128 sharply reduced the protein levels of all 6 targets by60% to 90%, whereas inhibition of miR-128 resulted in anobvious increase by 4-fold to 10-fold (Fig. 5D). To furtherascertain whether miR-128 represses these 6 targets in vivo,we examined their expression by IHC staining on formalin-fixed paraffin-embedded sections derived from tumorsformed in the xenograft studies. The 6 proteins were expressedat low, almost undetectable, levels in tumors derived fromMDA-MB-231 pri-miR-128-2 cells compared with their controlcounterparts, respectively (Fig. 5E). Thus, BMI1, CSF1, KLF4,LIN28A, NANOG, and SNAIL are all bona fide targets of miR-128.

miR-128 diminishes mammary carcinoma cell tumor-initiating capacity via AKT and STAT3

Given our identification of these 6 stem cell factors ascoordinate targets of miR-128, we reasoned there may becommon signaling pathways involved in mediating the effectsof miR-128. BMI1 has been reported to act as part of thepolycomb repressor complex, which plays a role in stem cellrenewal and malignant transformation through activation ofAKT pathway (25, 26). CSF1 is known as macrophage colonystimulating factor 1, which regulates progenitor cell differen-

tiation by AKT phosphorylation (27). KLF4, LIN28A, andNANOG are all well-known stem cell factors and reported topossess fundamental roles in iPS reprogramming, in whichAKT and STAT3 pathways are activated to promote theinduction of normal somatic cells to pluripotent stem cells(28, 29). SNAIL (SNAI) is a zinc finger transcriptional repressor,which has been described to induce EMT via repression of E-Cadherin and activation of several signaling pathways, includ-ing STAT3 and AKT (30, 31). Consequently, we examined theexpressions levels of AKT and STAT3 by Western blotting.Diminished p-AKT and p-STAT3 activities were observed inMDA-MB-231-miR-128 cells, whereas elevated p-AKT and p-STAT3 activities were detected in MCF-10A cells with miR-128depletion compared with their respective control cells (Fig.6A). Moreover, we carried out IHC staining to examine levels ofp-AKT and p-STAT3 on sections of xenograft tumors. Weobserved decreased p-AKT and p-STAT3 in MDA-MB-231-pri-miR-128-2 cells compared with control cells (Fig. 6B),showing that miR-128 deprives mammary carcinoma tumor-initiating suppression via AKT and STAT3 phosphorylationboth in vitro and in vivo.

To further show that p-AKT and p-STAT3 activities wererequired for the tumor-initiating capability of miR-128, MCF-10A miR-128 sponge cells were treated with either the specificAKT inhibitor IV and/or the STAT3 inhibitor III, WP1066, orspecific siRNAs targeting AKT and/or STAT3. We observedmarkedly diminished p-AKT and p-STAT3 activities (Fig. 6C)and expression of AKT and STAT3 (Supplementary Fig. S6C)in MCF-10A miR-128 sponge cells, which subsequently abro-gated miR-128 depletion–promoted tumor-initiating capacity(Fig. 6D, Fig. S6D). Thus, miR-128 modulates mammary car-cinoma tumor-initiating capability via PI3K/AKT and STAT3pathways.

miR-128-2 is directly repressed by SNAIL and attenuatedvia TGF-b

We next undertook to determine howmiR-128 expression isspecifically regulated. Given our observation that miR-128 wasexpressed at low levels in breast cancer cells and directlyrepressed multiple stem cell factors, we hypothesized whetherany transcription factors might regulate miR-128 expression.To this end, we searched conserved transcription factor bind-ing sites by use of rVista 2.0 (32), and identified 2 conserved E-boxmotifs, 1 at -991 bp (E-box 1, CACATG) and the other at -26bp (E-box 2, CACATG) relative to the transcription start site(þ1) of the humanmiR-128-2 stem-loop, respectively (Fig. 6E).SNAIL family (SNAIL, SLUG, and SMUC), ZEB (ZEB1 andZEB2), and the bHLH (E47 and TWIST1) families have beenreported to bind to E-box sequences (CANNTG) present in thepromoters of genes (30). To further determine which factorscould impact on miR-128-2 expression, we constructed a 3-kbfragment upstream of human miR-128-2 stem-loop andinserted the fragment into the luciferase reporter plasmidpGL3. We cotransfected this plasmid together with differentvectors expressing SNAIL, SLUG or TWIST1. We observedreduced luciferase activity by SNAIL and a milder decreaseby SLUG, whereas no change was observed with TWIST1(Supplementary Fig. S6A).

Qian et al.

Cancer Res; 72(22) November 15, 2012 Cancer Research6044

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 10: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

A

C

Rela

tive r

enill

a/firefly

lucifera

se

activity

0

0.2

0.4

0.6

0.8

1

1.2

1.4

psi-

Check2

BMI1

3'UTR

BMI1

3'UTR Mut

CSF1

3'UTR

Control

miR-128

** * * *

NS

NS NS NS

CSF1

3'UTR Mut

KLF4

3'UTR

KLF4

3'UTR Mut

NANOG

3'UTR

NANOG

3'UTR Mut

0

0.2

0.4

0.6

0.8

1

1.2

1.4

psi-

Check2

LIN28A

3’UTR

Control

miR-128

*

NS NS

NS NS

*

Rela

tive r

enill

a/firefly

lucifera

se

activity

LIN28A

Mut 1

LIN28A

Mut 2

LIN28A

Mut 1+2

SNAIL

3’UTR

SNAIL

3’UTR Mut

D

TargetScan

miRanda Pictar

435

131 220

98

Stem cell factors

BMI1, CSF1, KLF4, LIN28A, NANOG, SNAIL

60 191

41

B

E

pB

ab

e

pri

-miR

- 128-2

BMI1 CSF1

KLF4

pB

ab

e

pri

-miR

-128-2

LIN28A NANOG SNAIL

MDA-MB-231 with

miR-128 mimics MCF-10A with

miR-128 ASO

MDA-MB-231 with

miR-128 mimics MCF-10A with

miR-128 ASO

LIN28A

β-ACTIN

Control miR-128 Control miR-128 ASO

CSF1

BMI1

KLF4

NANOG

SNAIL

MDA-MB-231 MCF-10A

26kD

43kD

72kD

37kD

58kD

39kD

34kD

Figure 5. miR-128 directly targets a cohort of stem cell factors. A, schematic displaying in silico analyses that led to identification of 98 putativemiR-128 targetgenes by 3 computational algorithms. B, heatmap depicting themRNA expression profile of an array of genes involved in cancer initiation and progression byqRT-PCR in 2 sets of paired cells (MDA-MB-231 cells transfected with either miR-128 mimics or control and MCF-10A cells transfected with either miR-128ASO or scrambled sequence control). Red squares correspond to upregulation of mRNA levels, whereas green squares correspond to downregulation ofmRNA levels. C, luciferase activity of luciferase reporters with wild-type or mutant 30UTRs were carried out after cotransfection with miR-128 mimic ornegative control in MDA-MB-231 cells. The luciferase activity of each sample was normalized to Firefly luciferase activity. D, the protein levels weremeasuredbyWestern blotting after transfectionwithmiR-128mimicsor control inMDA-MB-231 cells (left) or after transfectionwithmiR-128ASOor control inMCF-10A cells (right). b-Actin was used as an internal control. E, the protein levels of targets were detected by immunohistochemical staining on sections ofxenograft tumors formed by MDA-MB-231-pri-miR-128-2 or control cells, at week 4 after subcutaneous transplantation. Images were taken at �400magnification. Bar, 20 mm. A representative experiment is shown in triplicate along as mean � SEM in C. �, P < 0.05; ��, P < 0.01; NS, not significant(Student t test).

miR-128-2 as a Tumor Suppressor in Breast Cancer

www.aacrjournals.org Cancer Res; 72(22) November 15, 2012 6045

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 11: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

B A

C D

AKT p-AKT STAT3 p-STAT3

pB

ab

e

pri

- miR

-128-2

AKT Inhibitor IV :

miR-128 ASO :

STAT3 Inhibitor III :

** ***

Fold

-mam

mosphe

re n

um

bers

0

0.5

1

1.5

2

2.5

3

3.5

4 ** **

F G

E

Rela

tive f

irefly/

renill

a

lucifera

se

activity

NS NS NS

*

*

0

0.2

0.4

0.6

0.8

1

1.2

pGL3 miR-128-2 Mut 1

pcDNA3.1 pc-Snail

Mut 2 Mut 1+2

164bp

205bp

170bp

Inp

ut

Sn

ail

IgG

Inp

ut

Sn

ail

IgG

Vector pc-Snail

No

Ab

No

Ab

EBox1

EBox2

Control

hsa-miR-128-2

stem loop E-Box 1

CACATG

E-Box 2

CACATG Control

–26 +1 –991 –2116

H

I

*

*

Rela

tive e

xpre

ssio

n o

f m

iRN

A/m

RN

A

**

**

0

0.2

0.4

0.6

0.8

1

1.2

miR-128 ARPP21 pri-miR-128-2

Control

pc-Snail

TGF-β:

miR-128 :

**

**

J K

L

Rela

tive e

xpre

ssio

n o

f

miR

-128/U

6

TGF-β: (Days) 0

0.2

0.4

0.6

0.8

1

1.2

0 3 6 9 12

** **

* *

0

1

2

3

4

5

6

7

8

9

10

pc-Snail :

miR-128 :

Fold

-mam

mosphe

re n

um

bers

*** **

M

TGF-β :

miR-128 :

Fold

-mam

mosphe

re n

um

bers

** **

0

2

4

6

8

10

12

14

200180160140

50454035302.01.51.00.50.0

0

0.2

0.4

0.6

0.8

1

1.2

Rela

tive e

xpre

ssio

n o

f

miR

-128/U

6

Rela

tive e

xpre

ssio

n o

f

miR

-128/U

6

SB431542

Control

TGF-β

**

NS

AKT

p-AKT

STAT3

p-STAT3

MCF-10A

AKT Inhibitor IV

III

:

miR-128 ASO :

STAT3 Inhibitor :

β-ACTIN

60kD

43kD

60kD

86kD

86kD

miR-128 ASO

AKT

p-AKT

STAT3

p-STAT3

Control miR-128 Control

MDA-MB-231 MCF-10A

β-ACTIN

60kD

43kD

60kD

86kD

86kD

Figure 6. miR-128 affects cellular pathways via AKT and STAT3 phosphorylation and is directly repressed by SNAIL and responsive to TGF-b. A, Westernblotting showing downregulation of AKT andSTAT3 phosphorylation inMDA-MB-231 cells with forced expression ofmiR-128 and increased AKT andSTAT3

Qian et al.

Cancer Res; 72(22) November 15, 2012 Cancer Research6046

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 12: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

We next cotranfected the SNAIL expressing vector withdifferent constructs, which contained 3-kb fragment upstreamof human miR-128-2 stem-loop, into MCF-10A cells andobserved that SNAIL significantly repressed the relative lucif-erase activity of miR-128-2 (Fig. 6F). In addition, mutations ofeither of the 2 E-Boxmotifs abrogated responsiveness to SNAIL(Fig. 6F), indicating that 2 E-Box sites are necessary andfunctional for SNAIL regulation of miR-128. Conversely, deple-tion of SNAIL by specific siRNA increased the activity ofluciferase construct of miR-128-2, whereas mutations in eitherof the 2 E-Box sites abolished the increase (Supplementary Fig.S6B). We next carried out ChIP assays to validate whetherSNAIL controls miR-128-2 expression by binding to the 2putative binding sites. With an amplicon at a distance of2,116 base pairs upstream of the human miR-128-2 stem-loopas negative control, ChIP revealed that SNAIL bound to both E-box 1 and E-box 2 (Fig. 6G). Concordantly, forced expression ofSNAIL decreased the expression levels of mature miR-128, pri-miR-128-2 and ARPP21 (Fig. 6H).We further examined the mammosphere-forming ability

of the MCF-10A cells with forced expression of SNAIL and/ormiR-128. MCF-10A cells with forced expression of SNAILformed 8-fold more mammospheres than MCF-10A controlcells, and the increased numbers of mammospheres wasabrogated by transfection with miR-128 (Fig. 6I). Thus, thesefindings indicate that SNAIL enhances tumor-initiatingcapability of mammary epithelial cells by repressing miR-128-2 expression through directly binding to the 2 E-Boxsites.We further sought to determine whether certain growth

factors secreted into the tumor microenvironment may con-tribute to promotion of mammary carcinoma tumor-initiatingcapacity through repression of miR-128 expression. Previousstudies revealed that TGF-b is a potent inducer of SNAIL,which can endow mammary epithelial cells with stem cell-likecharacteristics (33, 34). Treatment of MCF-10A cells with TGF-b resulted in reduced miR-128 expression (Fig. 6J), sustainedover time (Fig. 6K). In addition, to verify the specificity of TGF-b treatment, we used SB431542, a specific TGF-b1 receptorkinase inhibitor (35). We observed that SB431542 abrogated

the TGF-b induced reduction in miR-128 expression (Fig. 6L),suggesting that TGF-b decreased miR-128 expression byenhancing expression levels of SNAIL. Functionally, weobserved that TGF-b-treated MCF-10A cells exhibited an 11-fold increase in mammosphere formation compared withuntreated control cells (Fig. 6M). The increase in mammo-sphere numbers stimulated by TGF-b was abolished by forcedexpression of miR-128 (Fig. 6M). Thus, TGF-b promotes mam-mary epithelial cell oncogenic transformation, to some extent,by miR-128-2 downregulation.

DiscussionGenome instability has been proposed as one of the

hallmarks of cancer, which generates random mutationsand confers selective advantage on cancer cells, enablingtheir outgrowth and eventually increasing cancer risk (36).The fact that 50% of human miRNAs reside in or aroundfragile chromosomal sites prompted us to uncover thesebreakpoint-associated miRNAs and clarify their functions. Inthis study, we have identified 16 miRNAs, which are localizedat chromosome 3p and often aberrantly expressed in humanbreast cancer. Of the 11 miRNAs, which are subject tochromosome loss, several of them have been well documen-ted. Our group has observed that let-7g, located on chromo-some 3p21.1, is remarkably reduced in invasive ductalcarcinoma and may be considered as a prognostic biomark-er for breast cancer (10). In addition, miR-26a-1 and miR-135a-1 have been reported to be frequently lost in a variety ofhuman malignancies, including breast, prostate, liver, brain,pancreatic, colon, and nasopharyngeal carcinoma. More-over, miR-885-5p is downregulated upon loss of the3p25.3 region in neuroblastoma (37). Intriguingly, we alsofound that 5 miRNAs (Fig. 1A) were amplified in breastcancer compared with normal breast tissues. miR-191 hasbeen validated to be amplified and is viewed as a candidateoncogenic miRNA in liver (38), stomach (39), and pancreas(40). LOH comprises not only monosomy, caused by loss of 1allele, but also homozygous disomy and homozygous poly-somies, caused by the concurrent loss of 1 allele andendomultiplication of the other allele (41). Thus, we have

phosphorylation in MCF-10A cells with depletion of miR-128 compared with their control cells, respectively. B, immunohistochemical staining revealedreduced AKT and STAT3 phosphorylation on sections of xenograft tumors formed by MDA-MB-231-pri-miR-128-2 (bottom) compared with control cells(upper). Imageswere taken at�400magnification. Bar, 20mm.C,Western blotting showing decreasedAKT andSTAT3 phosphorylation inmiR-128-depletedMCF-10A cells treated with the specific AKT inhibitor IV and/or STAT3 inhibitor III. D, in vitro quantification of mammosphere formation inmiR-128-depletedMCF-10A cells (2� 103) treatedwith the specific AKT inhibitor IV and/or STAT3 inhibitor III. E, bioinformatics analyses of predicted bindingsites for SNAIL at promoter of miR-128. Schematic representation of the 2.2-kb regulatory region upstream of the human miR-128-2 stem-loop. TwoE-boxmotifs were predicted, 1 at -991 bp (E-box 1) and the other at -26 bp (E-box 2) relative to the transcription start site (þ1) of humanmiR-128-2 stem-loop.F, luciferase assays for promoter activity. MCF-10A cells were cotransfected with different promoter constructs (wide-type or mutant) and vector expressingSNAIL. All cells were cotransfected with a plasmid expressing Renilla luciferase as an internal control. G, ChIP assay in MCF-10A cells transfected with avector expressing SNAIL. PCR was carried out with primers specific for human miR-128 E-box 1 and E-box 2, respectively. Amplicon at a distance of 2,116basepairs upstreamof the humanmiR-128-2 stem-loopwasusedas negative control. H, qRT-PCRanalysis for expression levels ofmaturemiR-128,pri-miR-128-2 and ARPP21 after transfection with a vector expressing SNAIL in MCF-10A cells. U6 snRNA and GAPDH served as internal controls, respectively.I, in vitro quantification of mammosphere formation by MCF-10A cells (2 � 103) cotransfected with pc-SNAIL or control and miR-128 mimic. J, relativeexpression of miR-128 in MCF-10A cells treated with recombinant TGF-b (2.5 ng/mL) for 12 days and transfected with or without miR-128 mimic. K, relativeexpression of miR-128 in MCF-10A cells treated with recombinant TGF-b (2.5 ng/mL) for 3, 6, 9, 12 days, respectively. L, TGF-b depletion of miR-128expression was mediated through TGF-b1 receptor. MCF-10A cells were treated with TGF-b in the presence or absence of 5 mmol/L SB431542, a specificTGF-b1 receptor kinase inhibitor. M, in vitro quantification of mammosphere formation by MCF-10A cells (2� 103) described in J. U6 small nuclear RNA wasused as an internal control in J, K, and L. A representative experiment is shown in triplicate along as mean � SEM in D and F–M. �, P < 0.05; ��, P < 0.01;���, P < 0.001; NS, not significant (Student t test).

miR-128-2 as a Tumor Suppressor in Breast Cancer

www.aacrjournals.org Cancer Res; 72(22) November 15, 2012 6047

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 13: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

identified a series of aberrant miRNAs at chromosome 3p,which exhibits frequent LOH and a discontinuous pattern ofallele loss in breast cancer. Further endeavors will unraveltheir precise roles in the pathogenesis of breast cancer.

Among the eleven downregulated miRNAs, miR-128-2 wasthe most frequently deleted (75% deletion) in our cohort.Previous reports have identified miR-128 as a TSG. miR-128was first identified to be enriched in brain and frequentlyreduced in glioma cells (42), and dysregulation of miR-128confered a variety of malignant hallmarks on neural cells,including sustained proliferative signaling (43), resistingapoptosis (44), activating invasion and metastasis (45), andpromoting self-renewal (25). A recent study has proposedthat reduced miR-128 in breast tumor-initiating cells pro-duced chemotherapeutic resistance (46). Consistently, weobserved that forced expression of miR-128 in invasive MDA-MB-231 cells inhibited self-renewal and tumor-initiatingcapacities whereas depletion of miR-128 endowed normalmammary epithelial MCF-10A cells with a malignant phe-notype and initiated tumor formation in vivo. Moreover, wefurther provided proof that miR-128 could directly target acohort of stem cell factors that have comprehensive roles instem cell development. In addition, we showed that miR-128-2 is directly regulated by SNAIL, suggesting a potentialmechanism of how miR-128-2 is precisely controlled inembryogenesis and tumorigenesis. Finally, we observedmiR-128 is involved in the TGF-b signaling pathway, inwhich TGF-b promotes breast cancer initiation and pro-gression, partially, via miR-128-2 repression. Thus, we haveextended our current knowledge by delineating the role ofmiR-128 in tumor-initiation, implying miR-128 is a candidatemolecular target for clinical prognosis or therapy.

It is of interest to note miR-128 could directly target aseries of 6 genes: BMI1, CSF1, KLF4, LIN28A, NANOG, andSNAIL, all of which are critical determinants of normal stemcell maintenance and renewal (24), and contribute to tumorformation. One explanation as to why depletion of miR-128triggers oncogenic transformation in normal mammaryMCF-10A cells is due to the liberation of these 6 fundamentalfactors, which subsequently modulate PI3K/AKT and STAT3pathways enabling cells to undergo oncogenic transforma-tion. Our hypothesis is further supported by the observationsthat miR-128 modulates numerous key genes functionallyinvolved in EMT and neoplastic progression (SupplementaryTable S5).

The SNAIL family of zinc-finger transcrip-tion factors con-sist of SNAIL1 (SNAIL), SNAIL2 (SLUG), and SNAIL3 (SMUC),all potent inducers of EMT and important predictors of breastcancer metastatic potential and recurrence (47). Expression ofSNAIL is precisely regulated by a complex signaling network atthe transcriptional and posttranscriptional level. Here, ourfindings supplement this network with the novel descriptionof 2 positive feedback loops (Fig. 7). First, SNAIL directly bindsto E-Box sites upstream of miR-128-2 and represses miR-128-2expression, which in turn further increases the expression ofSNAIL via direct targeting by miR-128-2. Secondly, SNAILindirectly enhances the expression of BMI1, CSF1, KLF4,LIN28A, and NANOG by inhibiting miR-128-2, which cooper-

atively increases the activity of AKT and STAT3. It has beenreported that AKT upregulates the expression of SNAIL with(48) or without (49) the activation of NF-kB. The STAT3pathway was also recently been reported to increase theexpression of SNAIL (50). Thus, we have indentified anotherpositive feedback loop between SNAIL and the AKT or STAT3cascade. Furthermore, TGF-b pathway has been showed toinduce EMT and endow mammary epithelial cells with stemcell-like traits mainly via transcriptionally inducing SNAIL ex-pression (33, 34). Our results show that TGF-b promotes EMTand breast cancer initiation at least in part bymiR-128-2 down-regulation, revealing a cascade (TGF-b!TGF-b1 Receptor!SNAIL!miR-128-2!BMI1/CSF1/KLF4/LIN28A/NANOG!pSTAT3/pAKT!EMT/Stem cell-like traits) exists in the breastcancer oncogenic transformation triggered by TGF-b.

Collectively, in this study, our data reveal that decreasedmiR-128-2 expression due to allele loss on chromosome 3p22.3can endow nontumorigenic mammary epithelial MCF-10Awith malignant phenotypes and stem cell-like behaviors bothin vitro and in vivo via de-repression of a cohort of stem cellfactors and activation of the PI3K/AKT and STAT3 pathways.In addition, miR-128-2 is downregulated by SNAIL andrepressed in the TGF-b signaling pathway. The SNAIL/miR-128 axis provides a novel understanding of the mechanism ofmammary epithelial oncogenic transformation.

Disclosure of Potential Conflicts of InterestsNo potential conflicts of interest were disclosed.

SNAIL

hsa-miR-128-2

BMI1

p-STAT3

Oncogenic transformation

TGF-β1 Receptor

TGF-β

p-AKT

CSF1 KLF4 LIN28A NANOG

NF-κB

Figure 7. Proposedmodel for the critical roles ofmiR-128 in themediationofmammary epithelial oncogenic transformation andprogression. TGF-bspecifically represses miR-128 expression via phosphorylation of TGF-b1 receptor to increase SNAIL expression. SNAIL further direct binds toE-Box sites and downregulates miR-128-2, which in turn represses acohort of stem cell factors, including BMI1, CSF1, KLF4, LIN28A, andNANOG as direct targets and subsequently promotes oncogenictransformation of mammary epithelial cells via activation of p-STAT3 andp-AKT pathways. Two positive feedback loops are proposed asindicated, 1 between SNAIL and miR-128 and the other between SNAILand p-STAT3 and p-AKT activities via affecting SNAIL transcription orposttranscriptional processing.

Qian et al.

Cancer Res; 72(22) November 15, 2012 Cancer Research6048

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 14: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

Authors' ContributionsConception and design: P.-X. Qian, P.E. Lobie, T. ZhuDevelopment of methodology: P.-X. Qian, A. Banerjee, Z.S. WuAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): P.-X. Qian, A. Banerjee, X. Zhang, H. Wang, W. Zhang,P.E. Lobie, T. ZhuAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): P.-X. Qian, A. Banerjee, Z.S.Wu, V. Pandey,W. Zhang,P.E. Lobie, T. ZhuWriting, review, and/or revision of the manuscript: P.-X. Qian, A. Banerjee,P.E. Lobie, T. ZhuAdministrative, technical, or material support (i.e., reporting or organiz-ingdata, constructingdatabases):V. Pandey, X.-F. Lv, S. Tan, P.E. Lobie, T. ZhuStudy supervision: P.E. Lobie, T. Zhu

AcknowledgmentsWe thank Muh-Hwa Yang and Kou-Juey Wu (Institute of Clinical Med-

icine, National Yang-Ming University, Taipei, Taiwan) for kindly providing the

SNAIL, SLUG and TWIST1-expression plasmids. TERT-HMEC cell line waskindly gifted from Hines WC (University of New Mexico School of Medicine,New Mexico).

Grant SupportThis work was supported by the National Key Scientific Program of China

(2012CB934002, 2010CB912804), Chinese Academy of Sciences (XDA01040410),National Natural Science Foundation of China (30971492, 81101597), ChineseAcademy of Sciences Visiting Professorship for Senior International Scientist(2010TZS03), and Cancer Science Institute of Singapore.

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicate thisfact.

Received April 23, 2012; revised July 31, 2012; accepted August 14, 2012;published OnlineFirst September 26, 2012.

References1. Albertson DG, Collins C, McCormick F, Gray JW. Chromosome aber-

rations in solid tumors. Nat Genet 2003;34:369–76.2. Stratton MR. Exploring the genomes of cancer cells: progress and

promise. Science 2011;331:1553–8.3. Yang X, Lippman ME. BRCA1 and BRCA2 in breast cancer. Breast

Cancer Res Treat 1999;54:1–10.4. Yang Q, Yoshimura G, Mori I, Sakurai T, Kakudo K. Chromosome 3p

and breast cancer. J Hum Genet 2002;47:453–9.5. MaitraA,Wistuba II,WashingtonC, VirmaniAK,AshfaqR,MilchgrubS,

et al. High-resolution chromosome 3p allelotyping of breast carcino-mas and precursor lesions demonstrates frequent loss of heterozy-gosity and a discontinuous pattern of allele loss. Am J Pathol 2001;159:119–30.

6. Bartel DP. MicroRNAs: target recognition and regulatory functions.Cell 2009;136:215–33.

7. Calin GA, Croce CM.MicroRNA signatures in human cancers. Nat RevCancer 2006;6:857–66.

8. Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S,et al. HumanmicroRNAgenes are frequently locatedat fragile sites andgenomic regions involved in cancers. Proc Natl Acad Sci U S A2004;101:2999–3004.

9. Lu J, Getz G, Miska EA, Alvarez-Saavedra E, Lamb J, Peck D, et al.MicroRNA expression profiles classify human cancers. Nature2005;435:834–8.

10. Qian P, Zuo Z, Wu Z, Meng X, Li G, Zhang W, et al. Pivotal role ofreduced let-7g expression in breast cancer invasion and metastasis.Cancer Res 2011;71:6463–74.

11. Chen LC,MatsumuraK, DengG, KurisuW, LjungBM, LermanMI, et al.Deletion of two separate regions onchromosome3p in breast cancers.Cancer Res 1994;54:3021–4.

12. Matsumoto S, Kasumi F, Sakamoto G, Onda M, Nakamura Y, EmiM. Detailed deletion mapping of chromosome arm 3p in breastcancers: a 2-cM region on 3p14.3–21.1 and a 5-cM region on3p24.3–25.1 commonly deleted in tumors. Genes ChromosomesCancer 1997;20:268–74.

13. Pandey V, Perry JK, Mohankumar KM, Kong XJ, Liu SM, Wu ZS,et al. Autocrine human growth hormone stimulates oncogenicityof endometrial carcinoma cells. Endocrinology 2008;149:3909–19.

14. Tang JZ, Zuo ZH, Kong XJ, Steiner M, Yin Z, Perry JK, et al. Signaltransducer and activator of transcription (STAT)-5A and STAT5Bdifferentially regulate humanmammarycarcinomacell behavior. Endo-crinology 2010;151:43–55.

15. Kozomara A, Griffiths-Jones S. miRBase: integrating microRNAannotation and deep-sequencing data. Nucl Acids Res 2011;39:D152–7.

16. Sinha S, Singh RK, Alam N, Roy A, Roychoudhury S, Panda CK.Frequent alterations of hMLH1 and RBSP3/HYA22 at chromosomal

3p22.3 region in early and late-onset breast carcinoma: clinical andprognostic significance. Cancer Sci 2008;99:1984–91.

17. Trujillo KA, HinesWC, Vargas KM, Jones AC, Joste NE, BisoffiM, et al.Breast field cancerization: isolation and comparison of telomerase-expressing cells in tumor and tumor adjacent, histologically normalbreast tissue. Mol Cancer Res 2011;9:1209–21.

18. Gentner B, Schira G, Giustacchini A, AmendolaM, Brown BD, PonzoniM, et al. Stable knockdown of microRNA in vivo by lentiviral vectors.Nat Methods 2009;6:63–6.

19. Soule HD, Maloney TM, Wolman SR, Peterson WD Jr, Brenz R,McGrath CM, et al. Isolation and characterization of a spontaneouslyimmortalized human breast epithelial cell line, MCF-10. Cancer Res1990;50:6075–86.

20. Reya T, Morrison SJ, ClarkeMF,Weissman IL. Stem cells, cancer, andcancer stem cells. Nature 2001;414:105–11.

21. Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, BrownM, et al. ALDH1 is a marker of normal and malignant human mammarystem cells and a predictor of poor clinical outcome. Cell Stem Cell2007;1:555–67.

22. John B, Enright AJ, Aravin A, Tuschl T, Sander C, Marks DS. HumanMicroRNA targets. PLoS Biol 2004;2:e363.

23. Krek A, Grun D, Poy MN, Wolf R, Rosenberg L, Epstein EJ, et al.Combinatorial microRNA target predictions. Nat Genet 2005;37:495–500.

24. Jaenisch R, Young R. Stem cells, the molecular circuitry of pluripo-tency and nuclear reprogramming. Cell 2008;132:567–82.

25. Godlewski J, Nowicki MO, Bronisz A, Williams S, Otsuki A, Nuovo G,et al. Targeting of the Bmi-1 oncogene/stem cell renewal factor bymicroRNA-128 inhibits glioma proliferation and self-renewal. CancerRes 2008;68:9125–30.

26. Lukacs RU, Memarzadeh S, Wu H, Witte ON. Bmi-1 is a crucialregulator of prostate stem cell self-renewal and malignant transfor-mation. Cell Stem Cell 2010;7:682–93.

27. Papakonstanti EA, Zwaenepoel O, Bilancio A, Burns E, Nock GE,Houseman B, et al. Distinct roles of class IA PI3K isoforms in primaryand immortalised macrophages. J Cell Sci 2008;121:4124–33.

28. Huang J, Chen T, Liu X, Jiang J, Li J, Li D, et al. More synergeticcooperation of Yamanaka factors in induced pluripotent stem cellsthan in embryonic stem cells. Cell Res 2009;19:1127–38.

29. Okita K, Yamanaka S. Intracellular signaling pathways regulatingpluripotency of embryonic stem cells. Curr Stem Cell Res Ther2006;1:103–11.

30. Peinado H, Olmeda D, Cano A. Snail, Zeb and bHLH factors in tumourprogression: an alliance against the epithelial phenotype? Nat RevCancer 2007;7:415–28.

31. Dang H, Ding W, Emerson D, Rountree CB. Snail1 induces epithelial-to-mesenchymal transition and tumor initiating stem cell character-istics. BMC Cancer 2011;11:396.

miR-128-2 as a Tumor Suppressor in Breast Cancer

www.aacrjournals.org Cancer Res; 72(22) November 15, 2012 6049

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 15: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

32. Loots GG, Ovcharenko I. rVISTA 2.0: evolutionary analysis oftranscription factor binding sites. Nucl Acids Res 2004;32:W217–21.

33. Miettinen PJ, Ebner R, Lopez AR, Derynck R. TGF-beta inducedtransdifferentiation of mammary epithelial cells to mesenchymalcells: involvement of type I receptors. J Cell Biol 1994;127:2021–36.

34. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, et al. Theepithelial-mesenchymal transition generates cells with properties ofstem cells. Cell 2008;133:704–15.

35. Inman GJ, Nicolas FJ, Callahan JF, Harling JD, Gaster LM, Reith AD,et al. SB-431542 is a potent and specific inhibitor of transforminggrowth factor-beta superfamily type I activin receptor-like kinase(ALK) receptors ALK4, ALK5, and ALK7. Mol Pharmacol 2002;62:65–74.

36. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation.Cell 2011;144:646–74.

37. Afanasyeva EA, Mestdagh P, Kumps C, Vandesompele J, Ehemann V,Theissen J, et al. MicroRNA miR-885-5p targets CDK2 and MCM5,activates p53 and inhibits proliferation and survival. Cell Death Differ2011;18:974–84.

38. Elyakim E, Sitbon E, Faerman A, Tabak S, Montia E, Belanis L, et al.hsa-miR-191 is a candidate oncogene target for hepatocellular car-cinoma therapy. Cancer Res 2010;70:8077–87.

39. Shi X, Su S, Long J, Mei B, Chen Y. MicroRNA-191 targets N-deacetylase/N-sulfotransferase 1 and promotes cell growth in humangastric carcinoma cell line MGC803. Acta Biochim Biophys Sin(Shanghai) 2011;43:849–56.

40. Kent OA, Mullendore M, Wentzel EA, Lopez-Romero P, Tan AC,Alvarez H, et al. A resource for analysis of microRNA expression andfunction in pancreatic ductal adenocarcinoma cells. Cancer Biol Ther2009;8:2013–24.

41. Tsuda H. Gene and chromosomal alterations in sporadic breastcancer: correlation with histopathological features and implicationsfor genesis and progression. Breast Cancer 2009;16:186–201.

42. Smirnova L, Grafe A, Seiler A, Schumacher S, Nitsch R, Wulczyn FG.Regulation of miRNA expression during neural cell specification. Eur JNeurosci 2005;21:1469–77.

43. Zhang Y, Chao T, Li R, Liu W, Chen Y, Yan X, et al. MicroRNA-128inhibits glioma cells proliferation by targeting transcription factorE2F3a. J Mol Med (Berl) 2009;87:43–51.

44. Adlakha YK, Saini N. MicroRNA-128 downregulates Bax and inducesapoptosis in human embryonic kidney cells. Cell Mol Life Sci2011;68:1415–28.

45. Evangelisti C, FlorianMC,Massimi I, Dominici C, Giannini G, Galardi S,et al. MiR-128 up-regulation inhibits Reelin and DCX expression andreduces neuroblastoma cell motility and invasiveness. FASEB J2009;23:4276–87.

46. Zhu Y, Yu F, Jiao Y, Feng J, TangW, Yao H, et al. ReducedmiR-128 inbreast tumor-initiating cells induces chemotherapeutic resistance viaBmi-1 and ABCC5. Clin Cancer Res 2011;17:7105–15.

47. WuY, Zhou BP. Snail: more than EMT. Cell Adh Migr 2010;4:199–203.48. Julien S, Puig I, Caretti E, Bonaventure J, Nelles L, van Roy F, et al.

Activation of NF-kappaB by Akt upregulates Snail expression andinduces epithelium mesenchyme transition. Oncogene 2007;26:7445–56.

49. Grille SJ, Bellacosa A, Upson J, Klein-Szanto AJ, van Roy F,Lee-Kwon W, et al. The protein kinase Akt induces epithelial mesen-chymal transition and promotes enhancedmotility and invasiveness ofsquamous cell carcinoma lines. Cancer Res 2003;63:2172–8.

50. Yadav A, Kumar B, Datta J, Teknos TN, Kumar P. IL-6 promotes headand neck tumor metastasis by inducing epithelial-mesenchymal tran-sition via the JAK-STAT3-SNAIL signaling pathway. Mol Cancer Res2011;9:1658–67.

Qian et al.

Cancer Res; 72(22) November 15, 2012 Cancer Research6050

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507

Page 16: Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 ... · Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3 Targets Multiple Stem Cell Factors to Promote Transformation of

2012;72:6036-6050. Published OnlineFirst September 26, 2012.Cancer Res   PengXu Qian, Arindam Banerjee, Zheng-Sheng Wu, et al.   Mammary Epithelial CellsTargets Multiple Stem Cell Factors to Promote Transformation of Loss of SNAIL Regulated miR-128-2 on Chromosome 3p22.3

  Updated version

  10.1158/0008-5472.CAN-12-1507doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2012/09/26/0008-5472.CAN-12-1507.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/72/22/6036.full#ref-list-1

This article cites 50 articles, 14 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/72/22/6036.full#related-urls

This article has been cited by 5 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://cancerres.aacrjournals.org/content/72/22/6036To request permission to re-use all or part of this article, use this link

on May 13, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 26, 2012; DOI: 10.1158/0008-5472.CAN-12-1507