LIN28B Activation by PRL-3 Promotes Leukemogenesis and a ... · sion of PRL-3 in human AML cells...

11
Oncogenes and Tumor Suppressors LIN28B Activation by PRL-3 Promotes Leukemogenesis and a Stem Celllike Transcriptional Program in AML Jianbiao Zhou 1,2 , Zit-Liang Chan 1 , Chonglei Bi 1 , Xiao Lu 1 , Phyllis S.Y.Chong 1 , Jing-Yuan Chooi 2 , Lip-Lee Cheong 1 , Shaw-Cheng Liu 1 , Ying Qing Ching 1 , Yafeng Zhou 1 , Motomi Osato 1 , Tuan Zea Tan 1 , Chin Hin Ng 3 , Siok-Bian Ng 1,4,6 , Shi Wang 7 , Qi Zeng 5 , and Wee-Joo Chng 1,2,3 Abstract PRL-3 (PTP4A3), a metastasis-associated phosphatase, is also upregulated in patients with acute myeloid leukemia (AML) and is associated with poor prognosis, but the underlying molecular mechanism is unknown. Here, constitutive expres- sion of PRL-3 in human AML cells sustains leukemogenesis in vitro and in vivo. Furthermore, PRL-3 phosphatase activity dependently upregulates LIN28B, a stem cell reprogramming factor, which in turn represses the let-7 mRNA family, inducing a stem celllike transcriptional program. Notably, elevated levels of LIN28B protein independently associate with worse survival in AML patients. Thus, these results establish a novel signaling axis involving PRL-3/LIN28B/let-7, which confers stem celllike properties to leukemia cells that is important for leukemogenesis. Implications: The current study offers a rationale for targeting PRL-3 as a therapeutic approach for a subset of AML patients with poor prognosis. Mol Cancer Res; 15(3); 294303. Ó2016 AACR. Introduction The 5-year overall survival of acute myeloid leukemia (AML) remains about 30% to 40% and signicantly poorer in patients older than 65 years (1, 2). Leukemia stem cells (LSC), also called leukemia-initiating cells, a distinct subpopulation of AML cells with self-renewal capacity, initiate and sustain the development of the bulk leukemic population (35). LSC has been considered as the root source for the relapse and treatment failure of AML (6, 7). The identication of the molecular mechanisms underlying the transformation of LSCs offers a potential opportunity to eradicate leukemia (8). Protein tyrosine phosphatase of regenerating liver 3 (PRL-3, encoded by PTP4A3 gene) is a member of the VH1-like protein tyrosine phosphatase (PTP) family with dual specicity (9). Elevated expression of PRL-3 was detected in a variety of metastatic and primary tumor tissues (10). PRL-3 activates the PI3K/AKT pathway (11) and Src-ERK1/2 pathways (12), thus promoting epithelialmesenchymal transition (EMT) and tumor angiogenesis in solid tumors. PRL-3 protein is detected in approximately 50% and 90% of bone marrow samples from patients with AML and multiple myeloma, respectively (1316). Importantly, several studies have independently conrmed that the high expression of PRL-3 is associated with poor survival in AML (1517). We previously demonstrated that LEO1, a component of RNA polymerase IIassociated factor (PAF) complex, was induced by PRL-3 in AML (18). PAF complex plays an essential role in MLL-rearranged leukemia (19). These ndings collectively indicate that dysregu- lated PRL-3 is involved in the development of AML. However, the molecular mechanisms underlying the role of PRL-3 in AML are not well understood. Here, we report that LIN28B is a key oncogenic target in PRL-3positive AML. Increased LIN28B in turn downregulates let-7 mRNA family. The PRL-3/LIN28B/let-7 axis takes part in the transformation of LSCs and leukemogenesis of AML. Antagoniz- ing PRL-3 may be of clinical benet in the treatment of AML and eradication of LSCs. Materials and Methods Cell lines and cell culture AML cell lines MOLM-14, HL60, TF-1, and TF-1a were grown in RPMI1640 (Invitrogen) supplemented with 10% FBS (JRH Bios- ciences Inc.) at a density of 210 10 5 cells/ml in a humid incubator with 5% CO 2 at 37 C. Additional human IL3 (Pepro- Tech) was added into growth medium at 5 ng/mL to support TF-1 1 Cancer Science Institute of Singapore, National University of Singapore, Centre for Translational Medicine, Singapore, Republic of Singapore. 2 Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore. 3 Department of Haematology-Oncology, National University Cancer Institute, NUHS, Singapore, Republic of Singapore. 4 Department of Pathology, National University Hospital, Singapore, Republic of Singapore. 5 Institute of Molecular and Cell Biology, A STAR (Agency for Science, Technology and Research), Singapore, Republic of Singapore. 6 Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore. 7 Department of Pathology, National University Hospital, National University Health System, Singapore. Note: Supplementary data for this article are available at Molecular Cancer Research Online (http://mcr.aacrjournals.org/). Corresponding Author: Wee-Joo Chng, Department of Haematology-Oncolo- gy, National University Cancer Institute, NUHS, 1E, Kent Ridge Road, NUHS Tower Block, Level 10, Singapore 119228. Phone: 656-772-4612; Fax: 656-777- 5545; E-mail: [email protected] doi: 10.1158/1541-7786.MCR-16-0275-T Ó2016 American Association for Cancer Research. Molecular Cancer Research Mol Cancer Res; 15(3) March 2017 294 on February 4, 2021. © 2017 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from Published OnlineFirst December 23, 2016; DOI: 10.1158/1541-7786.MCR-16-0275-T

Transcript of LIN28B Activation by PRL-3 Promotes Leukemogenesis and a ... · sion of PRL-3 in human AML cells...

Page 1: LIN28B Activation by PRL-3 Promotes Leukemogenesis and a ... · sion of PRL-3 in human AML cells sustains leukemogenesis in vitro and in vivo. Furthermore, PRL-3 phosphatase activity

Oncogenes and Tumor Suppressors

LIN28B Activation by PRL-3 PromotesLeukemogenesis and a Stem Cell–likeTranscriptional Program in AMLJianbiao Zhou1,2, Zit-Liang Chan1, Chonglei Bi1, Xiao Lu1, Phyllis S.Y. Chong1,Jing-Yuan Chooi2, Lip-Lee Cheong1, Shaw-Cheng Liu1, Ying Qing Ching1,Yafeng Zhou1, Motomi Osato1, Tuan Zea Tan1, Chin Hin Ng3, Siok-Bian Ng1,4,6,Shi Wang7, Qi Zeng5, and Wee-Joo Chng1,2,3

Abstract

PRL-3 (PTP4A3), a metastasis-associated phosphatase, is alsoupregulated in patients with acute myeloid leukemia (AML)and is associated with poor prognosis, but the underlyingmolecular mechanism is unknown. Here, constitutive expres-sion of PRL-3 in human AML cells sustains leukemogenesisin vitro and in vivo. Furthermore, PRL-3 phosphatase activitydependently upregulates LIN28B, a stem cell reprogrammingfactor, which in turn represses the let-7 mRNA family, inducinga stem cell–like transcriptional program. Notably, elevated

levels of LIN28B protein independently associate with worsesurvival in AML patients. Thus, these results establish a novelsignaling axis involving PRL-3/LIN28B/let-7, which confersstem cell–like properties to leukemia cells that is importantfor leukemogenesis.

Implications: The current study offers a rationale for targetingPRL-3 as a therapeutic approach for a subset of AML patients withpoor prognosis. Mol Cancer Res; 15(3); 294–303. �2016 AACR.

IntroductionThe 5-year overall survival of acute myeloid leukemia (AML)

remains about 30% to 40% and significantly poorer in patientsolder than 65 years (1, 2). Leukemia stem cells (LSC), also calledleukemia-initiating cells, a distinct subpopulation of AML cellswith self-renewal capacity, initiate and sustain thedevelopment ofthe bulk leukemic population (3–5). LSC has been considered asthe root source for the relapse and treatment failure of AML (6, 7).The identification of the molecular mechanisms underlying thetransformation of LSCs offers a potential opportunity to eradicateleukemia (8).

Protein tyrosine phosphatase of regenerating liver 3 (PRL-3,encoded by PTP4A3 gene) is a member of the VH1-like protein

tyrosine phosphatase (PTP) family with dual specificity (9).Elevated expression of PRL-3 was detected in a variety ofmetastatic and primary tumor tissues (10). PRL-3 activates thePI3K/AKT pathway (11) and Src-ERK1/2 pathways (12), thuspromoting epithelial–mesenchymal transition (EMT) andtumor angiogenesis in solid tumors.

PRL-3 protein is detected in approximately 50% and 90% ofbone marrow samples from patients with AML and multiplemyeloma, respectively (13–16). Importantly, several studies haveindependently confirmed that the high expression of PRL-3 isassociated with poor survival in AML (15–17). We previouslydemonstrated that LEO1, a component of RNA polymerase II–associated factor (PAF) complex, was induced by PRL-3 in AML(18). PAF complex plays an essential role in MLL-rearrangedleukemia (19). These findings collectively indicate that dysregu-lated PRL-3 is involved in the development of AML. However, themolecular mechanisms underlying the role of PRL-3 in AML arenot well understood.

Here, we report that LIN28B is a key oncogenic target in PRL-3–positive AML. Increased LIN28B in turn downregulates let-7mRNA family. The PRL-3/LIN28B/let-7 axis takes part in thetransformation of LSCs and leukemogenesis of AML. Antagoniz-ing PRL-3 may be of clinical benefit in the treatment of AML anderadication of LSCs.

Materials and MethodsCell lines and cell culture

AML cell linesMOLM-14, HL60, TF-1, and TF-1a were grown inRPMI1640 (Invitrogen) supplemented with 10% FBS (JRH Bios-ciences Inc.) at a density of 2–10 � 105 cells/ml in a humidincubator with 5% CO2 at 37�C. Additional human IL3 (Pepro-Tech) was added into growthmedium at 5 ng/mL to support TF-1

1Cancer Science Institute of Singapore, National University of Singapore, Centrefor Translational Medicine, Singapore, Republic of Singapore. 2Department ofMedicine, Yong Loo Lin School of Medicine, National University of Singapore,Singapore, Republic of Singapore. 3Department of Haematology-Oncology,National University Cancer Institute, NUHS, Singapore, Republic of Singapore.4Department of Pathology, National University Hospital, Singapore, Republic ofSingapore. 5Institute ofMolecular andCell Biology, A�STAR (Agency for Science,Technology and Research), Singapore, Republic of Singapore. 6Department ofPathology, Yong Loo Lin School of Medicine, National University of Singapore,Singapore. 7Department of Pathology, National University Hospital, NationalUniversity Health System, Singapore.

Note: Supplementary data for this article are available at Molecular CancerResearch Online (http://mcr.aacrjournals.org/).

Corresponding Author: Wee-Joo Chng, Department of Haematology-Oncolo-gy, National University Cancer Institute, NUHS, 1E, Kent Ridge Road, NUHSTower Block, Level 10, Singapore 119228. Phone: 656-772-4612; Fax: 656-777-5545; E-mail: [email protected]

doi: 10.1158/1541-7786.MCR-16-0275-T

�2016 American Association for Cancer Research.

MolecularCancerResearch

Mol Cancer Res; 15(3) March 2017294

on February 4, 2021. © 2017 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst December 23, 2016; DOI: 10.1158/1541-7786.MCR-16-0275-T

Page 2: LIN28B Activation by PRL-3 Promotes Leukemogenesis and a ... · sion of PRL-3 in human AML cells sustains leukemogenesis in vitro and in vivo. Furthermore, PRL-3 phosphatase activity

cell growth. Bone marrow blast cells (>90%) from newly diag-nosed AML patients were obtained at National University Hos-pital in Singapore with informed consent. This study wasapproved by the Institutional Review Board of National Univer-sity of Singapore.

Cell viability assaysLeukemic cells were seeded in 96-well culture plates at a density

of 2 � 104 viable cells/100 mL/well in triplicates. CellTiter-GloLuminescent Cell Viability Assay (CTG assay, Promega) was usedto determine the cell growth and viability as described previously(20). Each experiment was in triplicate.

Affymetrix microarray and gene set enrichment analysesTF1-pEGFP and TF1-hPRL-3 cells were harvested, and total

RNA was extracted using the RNeasy Mini Kit, according tothe manufacturer's instructions (Qiagen). Gene expressionprofiling was performed using Affymetrix U133 Plus 2.0 genechip (Affymetrix) according to the manufacturer's protocol.The scanned data were processed using MicroArray Suite ver-sion 5.0 (Affymetrix). The detailed microarray analysis andsubsequent gene set enrichment analysis (GSEA) are describedin Supplementary Materials and Methods. Expression data(GSE64872) were deposited in the Gene Expression Omnibus.

Western blot analysisPreparation of the cell lysate and immunoblotting were per-

formed using standard techniques as described previously (21),except for PRL-3 analysis, and 16% gel was used. Anti-PRL3antibody was clone 318 (1:1,000) as reported previously (22).Anti-LIN28B antibody was purchased from Santa Cruz Biotech-nology (Cat. #sc-130802). Anti-b-actin (1:3,000, Sigma-Aldrich)was used as loading control.

Lentivirus LIN28B-shRNA infectionScramble shRNA control and two human LIN28B specifically

pLKO.1 lentiviral shRNAswere purchased fromOpenBiosystems.TF1-hPRL-3 or TF-1a cells (3�106)weremixedwith concentratedviral supernatant and 8 mg/mL of polybrene (Millipore) andcentrifuged at 2,500 rpm for 90minutes at 30�C. After additionalincubation at 37�C for 4 hours, themediumwas changed to freshcomplete medium. Two days later, cells were submitted forprotein extraction, followed by Western blot analysis of LIN28Band PRL-3.

mRNA mimics and inhibitorsPredesigned miRCURY LNA microRNA Mimics to let-7a

(#470408), let-7b (#470775), mRNA mimic negative control(#479903), specific miRCURY LNA microRNA Inhibitors to let-7a (#4101777), let-7b (#4100945), and control inhibitor(#199006) were purchased from Exiqon. Briefly, cells weretransfected with 30 nmol/L mRNA mimics or mRNA inhibitorsor their negative controls by electroporation with Neon Trans-fection System (Thermo Fisher Scientific) and were maintainedin a humidified incubator at 37�C in 5%CO2. Cell proliferationassays were carried out at days 1, 2, 4, and 6 after transfection.For colony-forming assays, colony numbers were counted atday 7.

Colony formation assay and serial replating assayTF1-pEGFP and TF1-hPRL-3 cells were washed twice with 1�

PBS, and cell viability was determined by Typan blue exclusionmethod. After that, about 20,000 vial cells from each cell linewere plated in MethoCult medium without cytokines (H4230,STEMCELL Technologies) in 6-well plates and cultured for 7 daysat 37�C in a 5%CO2 incubator. Colonies consisting of more than50 cells were counted under an invertedmicroscope. A total offiverandom 4 � 10 magnification fields were selected, and averagenumber of colonies of each sample was calculated. The experi-ments were duplicated. In serial replating assay, the colonynumberwas counted, and then subjected to replating every 7days.

Limited serial dilution and bone marrow transplantationThe protocol was reviewed and approved by Institutional

Animal Care and Use Committee in compliance with the Guide-lines on the Care and Use of Animals for Scientific Purpose.Detailed information is provided in Supplementary Materialsand Methods.

Statistical analysisStudent t test (two-tailed paired) was used for examining the

statistical difference for in vitro cell line experiments, such as cellviability assay and colony-forming assay; P values of <0.05 wereconsidered to be significant. Data were presented as mean � SD.Multivariate survival analyses were conducted using R version3.1.2 and survival package version 2.37-7. Protein expressionof LIN28B and clinicopathologic parameters were first discretizedinto binary categories prior to Cox regression analysis. Kaplan–Meier analyses were conducted using GraphPad Prism version5.04 (GraphPad Software). Statistical significance of the Kaplan–Meier analysis was calculated by log-rank test (P < 0.05).

ResultsPRL-3 promotes AML maintenance and progression in vivo

As reported inprevious studies (15, 18),we established apair ofstable, isogenic cell lines, TF1-pEGFP and TF1-hPRL3, by trans-fecting pEGFP (vector control) and pEGFP-hPRL-3 vectors intoTF-1 cells, respectively, and showed that PRL-3 promoted cyto-kine-independent growth of AML cells. Here, we further deter-mined the in vivo function of PRL-3 on leukemogenesis by usingthis pair of TF1 cells in a bone marrow transplantation xenograftassay. NOD/SCID mice inoculated with TF1-hPRL3 cells via tailvein injection developed leukemia-like symptoms with enlargedspleen and liver and succumbed to the diseasewithin 3months. Incontrast, mice implanted with TF1-pEGFP did not display anysign of sickness, and all survived till the end of the experimentalperiod (Fig. 1A). We next performed serial bone marrow trans-plantation experiments. Bone marrow cells harvested from theprimary recipients were transplanted into secondary recipients.Compared with primary leukemia, there was a significant shortersurvival in the secondary recipients (Fig. 1A, P < 0.01). We thenperformed tertiary transplantation. Tertiary recipients succumbedto the disease even earlier than the secondary recipients (Fig. 1A,P < 0.001). Histologic analysis revealed that leukemic cells exten-sively infiltrated into the liver and spleen in diseased mice(Fig. 1B). Moreover, the expressions of PRL-3 and LIN28B weresignificantly elevated in engrafted mouse bone marrow cellsharvested from secondary and tertiary recipients, while their

Activation of LIN28B by PRL-3 Promotes Leukemogenesis

www.aacrjournals.org Mol Cancer Res; 15(3) March 2017 295

on February 4, 2021. © 2017 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst December 23, 2016; DOI: 10.1158/1541-7786.MCR-16-0275-T

Page 3: LIN28B Activation by PRL-3 Promotes Leukemogenesis and a ... · sion of PRL-3 in human AML cells sustains leukemogenesis in vitro and in vivo. Furthermore, PRL-3 phosphatase activity

expression of let-7 was significantly lower as compared with theprimary generation of mouse bone marrow cells (Fig. 1C). Thesedata bespeak a good correlation between the expressions of thesethree genes and aggressiveness of the disease.

Together, these studies demonstrate that PRL-3 promotes AMLmaintenance and progress in an animal model.

PRL-3 induces a "stemness" transcriptional programin AML cells

To gain insight into the role of PRL-3 in leukemogenesis,we conducted an mRNA expression microarray analysis of TF1-pEGFP and TF1-hPRL3 cell lines. Using 2-fold as the cut-off level,89 genes showed increased expression and24genes haddecreased

expression in TF1-hPRL3 cells compared with TF1-pEGFP cells(Supplementary Table S1). The top 10 up- and downregulatedgenes were shown in Supplementary Table S2. It is noteworthythat PRL-3 induced expression of many LSC or cancer stem cell–related genes, including LIN28B, KIT, IGF2BP1 (IGF2 mRNAbinding protein 1), LAPTM4B, FYN, EIF4E3, Caveolin 1, amongothers, while reducing the expression of cell surface markers ofmore differentiated cells, such as CD36 and CD38 (Supplemen-tary Table S1). LIN28B ranked as the third most elevated geneafter PTP4A3 (PRL-3) and MT1G in TF-hPRL3 cells (Supplemen-tary Table S2). It has been well established that LIN28B is a stemcell reprogramming factor and a marker for cancer stem cell (23).qRT-PCR analysis validated the changes of selected genes, includ-ing LIN28B, LAPTM4B, IGF2BP1, FYN, RAB27B, KIT, CD36, andCD38 (Supplementary Fig. S1), and increased LIN28B in TF1-hPRL3 cells was confirmed by additional RT-PCR and Westernblot assays (Fig. 2A). GSEA of this microarray result revealed asignificant enrichment of genes identified in a previously definedgene expression signature of leukemia stem cell (LSC SIGNA-TURE_SAITO, P < 0.001) and hematopoietic stem cell (HSCSIGNATURE_EPPERT, P < 0.001; Fig. 2B). This differentiallyexpressed gene list was uploaded to the MetaCore algorithm forgene cluster analysis, and the result showed LIN28B to act as afocal point in the regulation of many genes such as miRNA let-7family (Supplementary Fig. S2).

Notably, the let-7 miRNA family members play a "gatekeeper"role of embryonic stem cell pluripotency and act as tumorsuppressors in a wide range of human malignancies (24). LIN28was identified to selectively repress the expressionof let-7miRNAsor directly bindmRNA (23, 25, 26). Therefore, we quantitated thelevel of let-7 miRNAs in TF1-pEGFP and TF1-hPRL3 cells. Withthe exception of let-7c, the other let-7miRNAmembers, includinglet-7a, let-7b, let-7d, let-7f, let-7g, let-7i, and miR-98, were allsignificantly repressed in TF1-hPRL3 cells relative to TF1-pEGFPcells (Fig. 2C). To rule out the effect of cell type specificity, weoverexpressed PRL-3 into other AML cell line, HL60, and mea-sured the expression of LIN28B and let-7a miRNA. Similarly, weobserved that HL60-hPRL3 cells had significant higher expressionof PRL-3 and LIN28B, but lower let-7a as compared with HL60-pEGFP cells (Fig. 2D). In summary, our data indicate that PRL-3enforces a stem-like transcription program in AML cells, driven bychanges of LIN28B and let-7 miRNAs.

PRL-3–expressing AML cells have leukemia-initiatingcapabilities

It has been well established that leukemia-initiating cells areenriched in CD34þCD38� subpopulation of AML cells. Lineageanalysis demonstrated that the percentage of CD34þCD38� cellswas 8-fold higher in TF1-hPRL3 cell lines than in TF1-pEGFP celllines (Fig. 3A; Supplementary Fig. S3A).We also found that PRL-3was more highly expressed on CD34þCD38� AML thanCD34þCD38þ AML cells, as well as CD34þCD38� cells fromhealthy controls, suggesting PRL-3 was upregulated in AML-ini-tiating cells (Fig. 3B; Supplementary Fig. S3B and S3C). To assessthe functional involvement of PRL-3 in leukemia-initiatingcells, we used serial replating assay to assess another intrinsiccharacteristic of leukemia-initiating cells, self-renewal capacity, inTF1-hPRL3 cells. As expected, TF1-hPRL3 cells could be seriallyreplated in basic methylcellulose without additional cytokines(Fig. 3C), whereas no colonies could be observed in TF1-pEGFPcells. To exclude possible artefacts accompanied by clone

1.5

1.0

0.5

0.00 20 40 60 80 100

Days postinoculation

Fra

ctio

n s

urv

ival

P < 0.001

TF1-pEGFPTF1-hPRL3 (Primary)

TF1-hPRL3 (Secondary)

TF1-hPRL3 (Tertiary)

A

B

C

TF1-pEGFP Mice TF1-hPRL3 Mice

Liver

Spleen

3.0

2.5

2.0

1.5

1.0

0.5

0.0Primary Secondary Tertiary

PRL-3 LIN28B let-7a

Rel

ativ

e ex

pre

ssio

n (

fold

ch

ang

e)

Figure 1.

PRL-3 promotes AML maintenance and progression in vivo. A, Survival analysisof immunodeficient mice transplanted with TF1-pEGFP, TF1-hPRL3 cells andsecondary and tertiary transplantation from primary recipient mice ofTF1-hPRL3. Each group consisted of 10 mice. Survival curves were constructedaccording to the Kaplan–Meier method. B, Representative images of H&Estaining of spleen and liver tissues harvested from TF1-pEGFP mice andTF1-hPRL3 mice. C, qRT-PCR analysis of PRL-3, LIN28B, and let-7a expression inthe bone marrow cells of recipient mice. Error bars indicate the mean � SDof 3 mice per group. � , P < 0.05; �� , P < 0.01.

Zhou et al.

Mol Cancer Res; 15(3) March 2017 Molecular Cancer Research296

on February 4, 2021. © 2017 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst December 23, 2016; DOI: 10.1158/1541-7786.MCR-16-0275-T

Page 4: LIN28B Activation by PRL-3 Promotes Leukemogenesis and a ... · sion of PRL-3 in human AML cells sustains leukemogenesis in vitro and in vivo. Furthermore, PRL-3 phosphatase activity

selection, we established another pair of isogenic TF1-FUGW andTF1-FUGW-PRL3 after transduced TF-1 cells with either FUGWcontrol lentivirus or FUGW-PRL3 lentivirus and selection. Inaccordance with the previous report, TF1-FUGW-PRL3 cellsgained cytokine-independent growth advantage, whereas TF1-FUGW cells failed to do (Supplementary Fig. S4). We nextassessed the replating capacity of this pair of isogenic cells.In agreement with the results from TF1-pEGFP and TF1-hPRL3,TF1-FUGW-PRL3 cells could be serially replated, whereas TF1-FUGW control cells could not form colonies (Supplementary Fig.S4). To address the question of whether TF1-hPRL3 cellsmaintainleukemia-initiating cell properties in vivo, we conducted limitingdilution transplantation and serial BMT assays. The ability of TF1-hPRL3 cells to propagate leukemia in immunodeficient mice wasevaluated using a 5-time cell dilution (50,000, 10,000, 2,000, and400 cells). Leukemic symptoms development characterized byhepatosplenomegaly was observed in all mice injected with50,000 and 10,000 cells (5mice/group), and 4 of 5mice receiving2,000 cells (Supplementary Table S3; Supplementary Fig. S5A).

Consistently, we also observed significantly higher expression ofPRL-3 and LIN28B and decreased expression of let-7a miRNA inbonemarrow cells harvested from diseasedmice (SupplementaryFig. S5B). In contrast, none of the mice (n¼ 5) transplanted with400 cells developed disease up to 6 months postinoculation(Supplementary Table S3; Supplementary Fig. S5A). So the esti-mated frequency of leukemia-initiating cell in TF1-hPRL3 cellscould be 1 in 1,771 [95% confidence interval (CI), 712–4,410)cells according to the analysis with ELDA software. In contrast,mice (n¼ 5) receiving 50,000 TF1-pEGFP cells were all healthy atthe end of experiments (Supplementary Table S3). Collectively,these results demonstrate a functional role for PRL-3 on trans-formation of leukemia-initiating cells in vitro and in vivo.

LIN28B is a key downstream target of PRL-3Through our microarray study, LIN28B is one of the most

significantly upregulated gene in TF1-hPRL3 compared withTF1-pEGFP. The C(X)5R motif within the P-loop of the PRL-3protein possess the PTP enzymatic activity. To examine the

Figure 2.

Gene expression changes driven by PRL-3 indicate acquisition of "stemness" properties in AML cells. A, Validation of increased expression of LIN28B on mRNAand protein levels by RT-PCR and Western blot (including densitometric quantification), respectively. M, 100 bp DNA ladder; lane 1, TF1-pEGFP; lane 2,TF1-hPRL3. B, GSEA analysis revealed that the TF1-hPRL3 cell expression profile significantly enriched previously reported LSC and HSC signatures. ES,enrichment score; NES, normalized enrichment score; FDR: False Discovery Rate. C, qRT-PCR analysis of the human let-7miRNA family in TF1-pEGFP and TF1-hPRL3cells. D, qRT-PCR analysis of PRL-3, LIN28B, and let-7a expression in HL60 AML cells transfected with either pEGFP or pEGFP-hPRL3 plasmid. Error barsindicate the mean � SD of three independent experiments. �� , P < 0.01.

Activation of LIN28B by PRL-3 Promotes Leukemogenesis

www.aacrjournals.org Mol Cancer Res; 15(3) March 2017 297

on February 4, 2021. © 2017 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst December 23, 2016; DOI: 10.1158/1541-7786.MCR-16-0275-T

Page 5: LIN28B Activation by PRL-3 Promotes Leukemogenesis and a ... · sion of PRL-3 in human AML cells sustains leukemogenesis in vitro and in vivo. Furthermore, PRL-3 phosphatase activity

importance of PRL-3 phosphatase activity in the regulation ofLIN28B, we transfected a catalytic domain mutant of PRL-3(C104S) into TF-1 cells and checked LIN28B protein level. West-ern blotting analysis showed that the substitution of C104 withserine effectively diminished thePRL-3–mediatedupregulationofLIN28, suggesting a phosphatase activity–dependent mechanismof regulation of LIN28B (Supplementary Fig. S6A). To verify therelevance of LIN28B overexpression in clinical samples, we exam-ined the expression of PRL-3 and LIN28B in CD34þCD38� cellsisolated from 28 primary AML samples using quantitative RT-PCR. As shown in Fig. 4A, there was a significant positive corre-lation between the expression levels of PRL-3 and LIN28B (Pear-son r¼ 0.51; two-tailed P¼ 0.007). Furthermore, as LIN28B playsan important role in repressing miRNAs and cellular reprogram-ming (27),we therefore focusedon the biological significance andregulatory mechanisms of LIN28B in the transformation of LSCsinduced by PRL-3. We used two independent LIN28B-shRNAs toselectively knockdown LIN28B expression in TF1-hPRL3 cells,and then performed serial replating assays. As shown in Fig. 4B,significant suppression of LIN28B protein was achieved by bothshRNAs. The knockdown efficacy of shRNA2 was higher thanshRNA1. Scrambled shRNA–treated TF1-hPRL3 cells could bereplated and the numbers of the colonies increased with eachround, whereas the colony number of LIN28B-shRNAs trans-duced cells was markedly diminished in each round of plating(Fig. 4C). Importantly, the significant reduction in the number ofcolonies correlated with the level of suppression of LIN28 expres-sion (Fig. 4B), indicating theon-target effect fromspecific shRNAs.We used lentiviral transduction method to specifically knock-down PRL-3 in MOLM-14 cells and then performed serial replat-

ing assays. Next, stable shRNA-mediated knockdown led to stablereductions in PRL-3 expression by 80%(Supplementary Fig. S6B).These cells and cells transduced with scramble shRNA were thenplated in methylcellulose for CFU in vitro assays. We observedsignificant reduced number of colonies in PRL-3 shRNA-expres-sing cells than scramble shRNA-expressing cells (SupplementaryFig. S6C). Collectively, these observations imply that inhibition ofPRL-3 and LIN28B effectively diminishes the replating capacity ofhuman AML cells.

Previously, a subclone of TF-1 cell line, TF-1a, was generated.TF-1a can proliferate continuously without cytokines anddeveloptumor in nude mice, in contrast to its parental line (28). Immu-nophenotypically, TF-1a cells are more immature and primitive(CD34þ/CD38�) than the parental TF-1 cells (CD34þ/CD38þ;ref. 28). TF-1a cells and TF1-hPRL3 cells share similarities in thesefeatures, which are indicators of self-renewal and uncontrolledproliferative capacity. In accordance with results obtained fromTF1-hPRL3 cells, high expression of PRL-3 and LIN28B proteinwas found in TF-1a cells compared with TF-1 cells (Fig. 4D). It isworth pointing out that the expression of PRL-3 and LIN28B inTF1-hPRL3 cells was comparable with their endogenous levels inTF-1a cells (Fig. 4D), suggesting our observations were not arti-ficially produced. The different levels of PRL-3 and LIN28Bproteins between TF-1a and TF-1 might be the underlying causefor the abovementioned different characteristics between thesetwo cell lines. To exclude any possible artifacts caused by over-expression or nonspecific effect of the shRNA approach, we nextconducted a loss-of-function together with a rescue experiment inthe TF-1a cell line expressing endogenously high level of PRL-3and LIN28B. TF-1a cells transduced with PRL-3-shRNAs or

10

9

8

7

6

5

4

3

2

1

0

4.5

4.0

3.5

3.0

2.5

2.0

1.5

1.0

0.5

0.0TF1-pEGFP TF1-hPRL3

CD

34+

CD

38–

Cel

ls (

fold

cha

nges

)

Rel

ativ

e ex

pres

sion

of P

RL-

3

Col

ony

num

ber

(2,0

00 c

ells

)

CD38– CD38–CD38+ CD38+

AML (CD34+) Controls (CD34+)

1,200

1,000

800

600

400

200

01 2 3

Plate

A

C

B

Figure 3.

PRL-3 is responsible for thetransformation of leukemia-initiatingcells. A, FACS analysis ofCD34þCD38� population in TF1-pEGFP and TF1-hPRL3 cell lines.B, qRT-PCR analysis of PRL-3expression in CD34þCD38�,CD34þCD38þ fractions isolated fromAML patient bone marrows (n ¼ 6,mean � SD) and healthy control bonemarrow cells (n ¼ 6, mean � SD). C,Serial replating assay using TF1-hPRL3cells. Bar figures represent threeseparate experiments (mean � SD).� , P < 0.05; �� , P < 0.01.

Zhou et al.

Mol Cancer Res; 15(3) March 2017 Molecular Cancer Research298

on February 4, 2021. © 2017 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst December 23, 2016; DOI: 10.1158/1541-7786.MCR-16-0275-T

Page 6: LIN28B Activation by PRL-3 Promotes Leukemogenesis and a ... · sion of PRL-3 in human AML cells sustains leukemogenesis in vitro and in vivo. Furthermore, PRL-3 phosphatase activity

2,000

1,500

1,000

500

00 500 1,000 1,500

PRL-3 Expression

LIN

28B

Exp

ress

ion

N = 28

P = 0.007Pearson r = 0.51

900

800

700

600

500

400

300

200

100

0

Col

ony

num

ber

(2,0

00 c

ells

)

1 2 3

Scramble

Scramble

Scr

ambl

e

PR

L-3

shR

NA

PR

L-3

shR

NA

+

LIN

28B

shR

NA

LIN

28B

OE

Plate

Plate

LIN28B-shRNA 1

LIN28B-shRNA

LIN28B-shRNA 2

Scr

ambl

e

LIN

28B

shR

NA

1LI

N28

B s

hRN

A2

LIN28B

LIN28B

LIN28B

β-Actin

β-Actin

β-Actin

1.0 0.6 0.2

0.1 1.0 1.0

1.0 1.0 1.1

1.0 3.0 4.0

1.0 0.2 0.9 0.8

1.0 0.2 0.2 0.9

1.0 1.0 1.0 1.0

1.0 1.1 1.1

PRL-3

PRL-3

PRL-3-shRNA

PRL-3-shRNA + LIN28 OE

TF

-1

TF

-1a

TF

-hP

RL3

1,000

900

800

700

600

500

400

300

200

100

01 2 3

Col

ony

num

ber

(2,0

00 c

ells

)

A

C

E

F

D

B

Figure 4.

LIN28B plays a pivotal role in PRL-3–enhanced LSC phenotype. A, The correlation of LIN28B and PRL-3 expression in CD34þCD28� primary bone marrowcells from 28 patients with AML at diagnosis. B, Immunoblot and densitometric analyses showed that LIN28B-specific shRNAs downregulated LIN28Bprotein expression in TF1-hPRL3 cells. C, Quantification of colonies of indicated cells over three rounds of replating in methylcellulose medium (n ¼ 3, mean �SD). � , P < 0.05; �� , P < 0.01. D, Comparison of PRL-3 and LIN28B proteins in TF-1, TF-1a, and TF1-hPRL3 cell lines. E, Western blot and densitometricanalyses of PRL-3 and LIN28B protein levels in TF1-a cells receiving different treatments as indicated. OE, overexpression. F, Quantification of colonies ofindicated cell lines over three rounds of replating (n ¼ 3, mean � SD). The colony number of LIN28B overexpression was significantly higher thanthose of LIN28B-shRNA and PRL-3 shRNA (P < 0.05).

Activation of LIN28B by PRL-3 Promotes Leukemogenesis

www.aacrjournals.org Mol Cancer Res; 15(3) March 2017 299

on February 4, 2021. © 2017 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst December 23, 2016; DOI: 10.1158/1541-7786.MCR-16-0275-T

Page 7: LIN28B Activation by PRL-3 Promotes Leukemogenesis and a ... · sion of PRL-3 in human AML cells sustains leukemogenesis in vitro and in vivo. Furthermore, PRL-3 phosphatase activity

LIN28B-shRNAs produced significantly less number of coloniesin the serial replating assay, while replating activity of PRL-3-shRNA transduced cells was restored to near normal level by theoverexpression of LIN28B (Fig. 4E and F). In conclusion, thesedata support that LIN28B is essential for PRL-3–mediated replat-ing capacity of leukemic cells.

Repression of let-7 miRNAs is required for the PRL-3/LIN28B-promoted leukemogenesis

The RNA-binding protein, LIN28B, functions to repress let-7miRNAs via blocking the microprocessor complex (29). As it iswell established that let-7 is an important tumor suppressor gene,which inhibits numerous oncogenes, such as RAS, c-MYC, NFkB,and HMGA (30, 31), we decided to examine whether the repres-sion of let-7 miRNAs is required for PRL-3/LIN28B-inducedleukemogenesis. Ectopic expression of let-7a and let-7b ledto decreased cell colony-forming capacity of TF1-hPRL3 cells(Fig. 5A, P < 0.05) and proliferation (Fig. 5B, P < 0.01). Concor-dantly, antagomiRs to let-7a and let-7b significantly increasedcolony-forming capacity of TF1-hPRL3 cells (Fig. 5C, P < 0.05)and cell proliferation (Fig. 5D, P < 0.01). To further validate theimportant role of let-7 miRNAs, we repeated the same experi-ments by replacing TF1-hPRL3 cell line with MOLM-14, expres-

sing high level of endogenous PRL-3 and LIN28B protein.Indeed, similar results were observed for MOLM-14 cells (Sup-plementary Fig. S7). Taken together, these evidences demonstratethat let-7 plays an essential role in the PRL-3/LIN28B-mediatedleukemogenesis.

LIN28Boverexpression independently predicts poor survival ofpatients with AML

To examine the clinical importance of LIN28B in AML, weused IHC to determine LIN28B protein expression in bonemarrow samples from 159 patients with AML at diagnosis.LIN28B staining was calculated as the number of LIN28B-positive blast cells divided by the total number of blast cells inat least 10 fields and then expressed as a percentage. Scoring ofthe tissue microarray was completed independently by twoclinically qualified pathologists who were blinded to the clinicalinformation. We observed that LIN28B mainly localizes innuclear of leukemia blast cells. We classified the LIN28B stainingpattern as follows: negative, <20% leukemia cells with nuclearstaining, 20%–<50% leukemia cells with nuclear staining, 50%–

<75% leukemia cells with strong nuclear staining, and �75%leukemia cells with strong nuclear staining. The scoring resultswere shown in Supplementary Table S4, and representative

600

500

400

300

200

100

0

60

50

40

30

20

10

0NC miRNA let-7a let-7b Day 0 Day 2 Day 4 Day 6

Col

ony

num

ber

(2,0

00 c

ells

)

Col

ony

num

ber

(2,0

00 c

ells

)

Cel

l num

ber

(×10

5 )

Cel

l num

ber

(×10

5 )

Vector ctrl

let-7a

let-7b

1,000

900

800

700

600

500

400

300

200

100

0

n.s.

No inhibitor Ctrl Inhibitor let-7a Inhibitor let-7b Inhibitor Day 0 Day 2 Day 4 Day 6

90

80

70

60

50

40

30

20

10

0

No inhibitor

let-7a Inhibitorlet-7b Inhibitor

Ctrl Inhibitor

A

C D

B

Figure 5.

Role of let-7 miRNAs in PRL-3/LIN28B-mediated leukemogenesis. A and B, Impact of let-7a and let-7b on colony forming (A) and cell proliferation (B) ofTF1-hPRL3 cells. Ambion Pre-miR let-7a, let-7b, and a negative control (NC) miRNA (30 nmol/L each) were transfected into TF1-hPRL3 cells by electroporationwith Neon Transfection System. C and D, Effect of inhibitors specific to let-7a and let-7b (30 nmol/L each) on colony forming (C) and cell proliferation (D)of TF1-hPRL3 cells. The results were shown as mean � SD of three independent experiments. � , P < 0.05; �� , P < 0.01.

Zhou et al.

Mol Cancer Res; 15(3) March 2017 Molecular Cancer Research300

on February 4, 2021. © 2017 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst December 23, 2016; DOI: 10.1158/1541-7786.MCR-16-0275-T

Page 8: LIN28B Activation by PRL-3 Promotes Leukemogenesis and a ... · sion of PRL-3 in human AML cells sustains leukemogenesis in vitro and in vivo. Furthermore, PRL-3 phosphatase activity

LIN28B staining images were presented in Fig. 6A. Notably, thefive normal bone marrow samples from healthy controls dem-onstrated very little LIN28B expression (Fig. 6A).

The survival data were available for 108 patients. We classifiedpatients with LIN28B scoring 50% and above as the "LIN28Bhigh" group and patients with negative or LIN28B scoring lessthan 50% as the "LIN28B low" group. The comparison of variousclinical features between LIN28B high and low groups was sum-marized in Supplementary Table S5.We then compared event-freesurvival between these two groups of patients. On the basis of theKaplan–Meier survival analysis, the group of patients who wereLIN28B high had amuch shorter event-free survival than patientswhowere LIN28B low (Fig. 6B, P¼ 0.0159). Multivariate analysisrevealed that LIN28B protein expression was an independentprognostic factor of FLT3 status, NPM mutation, and cytogeneticrisk. The HR of LIN28B expression was 3.96 (95% CI, 2.15–7.30;P < 0.001; Supplementary Table S6). Overall, high expression ofLIN28B protein was a novel, independent predictor for poorsurvival in our cohort.

DiscussionThe phosphatase of regenerating liver (PRL) family contains

three members, PRL-1, PRL-2, and PRL-3. Among the PRL family,PRL-3 has been most frequently studied in cancer, followed byPRL-2 and PRL-1. PRL-2 has been reported to increase EPO andIL3 response in hematopoietic cells (32) and play critical roles inregulating HSC self-renewal (33). A previous study reported thatgenetic disruption of PRL-3 reduces clonogenicity and growth ofCD133þ mouse colon cancer cells in an in vitro culture system(34). CD133 is also a specific marker for human hematopoieticstem/progenitor cells (35, 36). The observations from these

published studies indicate a possibility that aberrant expressionof PRL family in hematopoietic cells may promote the transfor-mation of LSC in AML. The association between PRL-3 expressionand poor prognosis has been widely confirmed in a number ofsolid tumors and AML (10, 13, 15–17), arguing for the role ofPRL-3 as a biomarker or a therapeutic target in stratifying patientsfor personalized therapy (9, 33, 37, 38). Thus, it is of clinicalimportance to further understand the molecular mechanism bywhich PRL-3 is regulated and contributes to leukemogenesis. Inthis study, we demonstrate that PRL-3 contributes to the leukemicphenotype of AML and plays an important role in the transfor-mation of LSCs via the LIN28B/let-7 regulatory axis (graphicsummary in Fig. 7).

Our global gene expression analysis showed the upregula-tion of several stemness factors, such as LIN28B, c-KIT, andIGF2BP1, and the downregulation of cell surface markers ofmore mature cells, such as CD36 and CD38, by PRL-3. c-KITfunctions as an important regulator of HSC proliferation andself-renewal through binding to soluble steel factor (39). Gain-of-function mutations in c-KIT have been characterized in AMLand other types of cancers (40). GSEA analysis further revealedthat our gene expression profile significantly enriched withpreviously reported LSC and HSC gene signatures (41, 42),suggesting PRL-3 induces a stem cell-like transcriptional pro-gram in leukemia cells.

Serial transplantation in mouse model and serial replatingassay provided functional evidence that PRL-3 confers stemcell–like properties in AML cells and mediates their transforma-tion into leukemia-initiating cells. We further demonstrated thatincreased expression is dependent on the phosphatase activity ofPRL-3, and LIN28B is essential for this transformation process.LIN28B is a stemness factor and an miRNA regulator, whichinhibits the biogenesis of let-7 miRNA family (25, 26). A dysreg-uated LIN28B/let-7 circuit has been implicated in the develop-ment of many types of malignancies, including T-cell acutelymphoblastic leukemia (43), neuroblastoma (44), and livercancer (45). Interestingly, MUC1-C, a transmembrane oncopro-tein, induces activation of LIN28B and downregulation of let-7 innon–small cell lung cancer, thus stimulating EMT and stem cellself-renewal (46). Here, we demonstrate that the LIN28B/let-7axis plays a key role in PRL-3–induced leukemia. The colony-

AML#1 AML#2 Ctrl

1.0

0.5

0.00 1,000 2,000 3,000 4,000

Fra

ctio

n s

urv

ival

Days

P = 0.0159

LIN28B Low

LIN28B High

A

B

Figure 6.

The clinical relevance of LIN28B expression in patients with AML. A,Representative IHC images of AML#1 (LIN28B: strong positive), AML#2 (LIN28B:negative), and 1 healthy control (LIN28B: weak positive). B, Impact of LIN28Bprotein expression on outcome of patients with AML. Survival curves wereconstructed according to the Kaplan–Meier method. LIN28B low: staining <50%(n ¼ 67); LIN28B high: staining �50% (n ¼ 41); P ¼ 0.0158.

Figure 7.

Schematic description of the role of PRL3/LIN28B/let-7 axis in the pathogenesis

of AML. ", increased expression; #, decreased expression; activate; ?, inhibit.

Activation of LIN28B by PRL-3 Promotes Leukemogenesis

www.aacrjournals.org Mol Cancer Res; 15(3) March 2017 301

on February 4, 2021. © 2017 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst December 23, 2016; DOI: 10.1158/1541-7786.MCR-16-0275-T

Page 9: LIN28B Activation by PRL-3 Promotes Leukemogenesis and a ... · sion of PRL-3 in human AML cells sustains leukemogenesis in vitro and in vivo. Furthermore, PRL-3 phosphatase activity

forming capacity of leukemic cells is alleviated after knockingdown LIN28B or upon ectopic expression of let-7 miRNAs, whileoverexpression of LIN28B partially rescues the decreased colonyforming capacity caused by viral-mediated silencing of PRL-3 inserial replating assays. By studying a cohort of clinical samples, weshowed the clinical significance of LIN28B expression in bonemarrow samples of AML patients. The shared gene signatures ofLSCs between our expression profile and others derived fromclinical AML samples further underscores the clinical relevance ofour findings (41, 42).

Leukemia-initiating or stem cells can arise from malignanttransformation of normal hematopoietic stem cells or the dereg-ulation of genes that induce stem cell–like properties in moremature progenitor cells. Our investigation has uncovered a noveland critical regulatory PRL-3/LIN28B/let-7 network, which playsan important role in transformation of LSCs and development ofAML and identifies AML patients with poor outcomes. We havepreviously shown that PRL-3 is deregulated in myeloid malig-nancies by STAT activation by upstream oncogenic kinases, suchas FLT3 (e.g., in FLT3-ITD AML; refs. 13, 21, 47). Our resultstherefore provide potential mechanistic insights to the highrelapse rates and poor outcome of FLT3-ITD AML. This novelregulatory network may represent the "Achilles' heel" of LSCs inAML, and its therapeutic disruption could lead to the eliminationof LSCs in a molecular subset of AML in which PRL-3/LIN28Bappears pivotal for the transformation.

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

Authors' ContributionsConception and design: J. Zhou, W.-J. ChngDevelopment of methodology: J. Zhou, J.-Y. Chooi, W.-J. ChngAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): J. Zhou, Z.-L. Chan, C. Bi, X. Lu, J.-Y. Chooi,S.-C. Liu, Y.Q. Ching, Y. Zhou, C.H. Ng, S.-B. Ng, S. WangAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): J. Zhou, Z.-L. Chan, C. Bi, J.-Y. Chooi, M. Osato,T.Z. Tan, W.-J. ChngWriting, review, and/or revision of the manuscript: J. Zhou, W.-J. ChngAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): C. Bi, P.S.Y. Chong, J.-Y. Chooi, L.-L. Cheong,Y.Q. Ching, Q. ZengStudy supervision: W.-J. Chng

AcknowledgmentsThe authors thank the Singapore National Research Foundation and the

Ministry of Education under the Research Center of Excellence Program andNational Medical Research Council (NMRC) for financial support.

Grant SupportThis work was supported by the Singapore National Research Founda-

tion and the Ministry of Education under the Research Center of Excel-lence Program (to W.-J. Chng) and NMRC Clinician-Scientist IRG grantCNIG11nov38 (to J. Zhou). W.-J. Chng was also supported by NMRCClinician Scientist Investigator Award.

The costs of publication of this articlewere defrayed inpart by the payment ofpage charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received August 11, 2016; revised October 29, 2016; accepted November 18,2016; published OnlineFirst December 23, 2016.

References1. Burnett A,WetzlerM, Lowenberg B. Therapeutic advances in acutemyeloid

leukemia. J Clin Oncol 2011;29:487–94.2. Zhou J, Ching YQ, Chng WJ. Aberrant nuclear factor-kappa B activity in

acute myeloid leukemia: from molecular pathogenesis to therapeutictarget. Oncotarget 2015;6:5490–500.

3. Bonnet D, Dick JE. Human acute myeloid leukemia is organized as ahierarchy that originates from a primitive hematopoietic cell. Nat Med1997;3:730–7.

4. Yip BH, So CW. Mixed lineage leukemia protein in normal and leukemicstem cells. Exp Biol Med 2013;238:315–23.

5. Gilliland DG, Jordan CT, Felix CA. The molecular basis of leukemia.Hematology Am Soc Hematol Educ Program 2004:80–97.

6. Wiseman DH, Greystoke BF, Somervaille TC. The variety of leukemic stemcells in myeloid malignancy. Oncogene 2014;33:3091–8.

7. Zhou J, ChngWJ. Identification and targeting leukemia stem cells: The pathto the cure for acutemyeloid leukemia.World J StemCells 2014;6:473–84.

8. Misaghian N, Ligresti G, Steelman LS, Bertrand FE, Basecke J, Libra M, et al.Targeting the leukemic stem cell: the Holy Grail of leukemia therapy.Leukemia 2009;23:25–42.

9. Campbell AM, Zhang ZY. Phosphatase of regenerating liver: a novel targetfor cancer therapy. Expert Opin Ther Targets 2014;18:555–69.

10. Al-Aidaroos AQ, Zeng Q. PRL-3 phosphatase and cancer metastasis. J CellBiochem 2010;111:1087–98.

11. Wang H, Quah SY, Dong JM, Manser E, Tang JP, Zeng Q. PRL-3 down-regulates PTEN expression and signals through PI3K to promote epithelial-mesenchymal transition. Cancer Res 2007;67:2922–6.

12. Ming J, Liu N, Gu Y, Qiu X, Wang EH. PRL-3 facilitates angiogenesis andmetastasis by increasing ERK phosphorylation and up-regulating thelevels and activities of Rho-A/C in lung cancer. Pathology 2009;41:118–26.

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

signaling and implicated in anti-AML therapy. PLoS One 2011;6:e19798.

14. Fagerli UM, Holt RU, Holien T, Vaatsveen TK, Zhan F, Egeberg KW, et al.Overexpression and involvement inmigrationby themetastasis-associatedphosphatase PRL-3 in human myeloma cells. Blood 2008;111:806–15.

15. Park JE, YuenHF,Zhou JB, Al-Aidaroos AQ,GuoK,Valk PJ, et al.Oncogenicroles of PRL-3 in FLT3-ITD induced acute myeloid leukaemia. EMBOMolMed 2013;5:1351–66.

16. Qu S, Liu B, Guo X, Shi H, Zhou M, Li L, et al. Independent oncogenic andtherapeutic significance of phosphatase PRL-3 in FLT3-ITD-negative acutemyeloid leukemia. Cancer 2014;120:2130–41.

17. BeekmanR, ValkhofM, Erkeland SJ, Taskesen E, RockovaV, Peeters JK, et al.Retroviral integration mutagenesis in mice and comparative analysis inhumanAML identify reduced PTP4A3 expression as a prognostic indicator.PLoS One 2011;6:e26537.

18. ChongPS, Zhou J, Cheong LL, Liu SC,Qian J,Guo T, et al. LEO1 is regulatedby PRL-3 and mediates its oncogenic properties in acute myelogenousleukemia. Cancer Res 2014;74:3043–53.

19. Muntean AG, Tan J, Sitwala K, Huang Y, Bronstein J, Connelly JA, et al. ThePAF complex synergizes with MLL fusion proteins at HOX loci to promoteleukemogenesis. Cancer Cell 2010;17:609–21.

20. Zhou J, Pan M, Xie Z, Loh SL, Bi C, Tai YC, et al. Synergistic antileukemiceffects betweenABT-869 and chemotherapy involve downregulation of cellcycle-regulated genes and c-Mos-mediated MAPK pathway. Leukemia2008;22:138–46.

21. Zhou J, Chong PS, Lu X, Cheong LL, Bi C, Liu SC, et al. Phosphatase ofregenerating liver-3 is regulated by signal transducer and activatorof transcription 3 in acute myeloid leukemia. Exp Hematol 2014;42:1041–52.

22. Li J, Guo K, Koh VW, Tang JP, Gan BQ, Shi H, et al. Generation of PRL-3-and PRL-1-specific monoclonal antibodies as potential diagnostic markersfor cancer metastases. Clin Cancer Res 2005;11:2195–204.

Zhou et al.

Mol Cancer Res; 15(3) March 2017 Molecular Cancer Research302

on February 4, 2021. © 2017 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst December 23, 2016; DOI: 10.1158/1541-7786.MCR-16-0275-T

Page 10: LIN28B Activation by PRL-3 Promotes Leukemogenesis and a ... · sion of PRL-3 in human AML cells sustains leukemogenesis in vitro and in vivo. Furthermore, PRL-3 phosphatase activity

23. Zhou J, Ng SB, Chng WJ. LIN28/LIN28B: an emerging oncogenic driver incancer stem cells. Int J Biochem Cell Biol 2013;45:973–8.

24. Fabbri M, Garzon R, Andreeff M, Kantarjian HM, Garcia-Manero G, CalinGA. MicroRNAs and noncoding RNAs in hematological malignancies:molecular, clinical and therapeutic implications. Leukemia 2008;22:1095–105.

25. Heo I, Joo C, Kim YK, Ha M, Yoon MJ, Cho J, et al. TUT4 in concert withLin28 suppresses microRNA biogenesis through pre-microRNA uridyla-tion. Cell 2009;138:696–708.

26. Viswanathan SR, Daley GQ, Gregory RI. Selective blockade of microRNAprocessing by Lin28. Science 2008;320:97–100.

27. Thornton JE, Gregory RI. How does Lin28 let-7 control development anddisease? Trends Cell Biol 2012;22:474–82.

28. HuX,Moscinski LC,Hill BJ, ChenQ,Wu J, Fisher AB, et al. Characterizationof a unique factor-independent variant derived from human factor-depen-dent TF-1 cells: a transformed event. Leuk Res 1998;22:817–26.

29. Piskounova E, Polytarchou C, Thornton JE, LaPierre RJ, Pothoulakis C,Hagan JP, et al. Lin28A and Lin28B inhibit let-7 microRNA biogenesis bydistinct mechanisms. Cell 2011;147:1066–79.

30. Wang LD, Rao TN, Rowe RG, Nguyen PT, Sullivan JL, PearsonDS, et al. Therole of Lin28b in myeloid and mast cell differentiation and mast cellmalignancy. Leukemia 2015;29:1320–30.

31. Bussing I, Slack FJ, Grosshans H. let-7 microRNAs in development, stemcells and cancer. Trends Mol Med 2008;14:400–9.

32. Akiyama S, Dhavan D, Yi T. PRL-2 increases Epo and IL-3 responses inhematopoietic cells. Blood Cells Mol Dis 2010;44:209–14.

33. Kobayashi M, Chen S, Gao R, Bai Y, Zhang ZY, Liu Y. Phosphatase ofregenerating liver in hematopoietic stem cells and hematological malig-nancies. Cell Cycle 2014;13:2827–35.

34. Cramer JM, ZimmermanMW,ThompsonT,HomanicsGE, Lazo JS, LagasseE. Deletion of Ptp4a3 reduces clonogenicity and tumor-initiation ability ofcolitis-associated cancer cells in mice. Stem Cell Res 2014;13:164–71.

35. Bhatia S, Reister S, Mahotka C, Meisel R, Borkhardt A, Grinstein E. Controlof AC133/CD133 and impact on human hematopoietic progenitor cellsthrough nucleolin. Leukemia 2015;29:2208–20.

36. Takahashi M, Matsuoka Y, Sumide K, Nakatsuka R, Fujioka T, Kohno H,et al. CD133 is a positivemarker for a distinct class of primitive human cord

blood-derived CD34-negative hematopoietic stem cells. Leukemia 2014;28:1308–15.

37. Guo K, Li J, Tang JP, Tan CP, Hong CW, Al-Aidaroos AQ, et al. Targetingintracellular oncoproteins with antibody therapy or vaccination. Sci TranslMed 2011;3:99ra85.

38. Guo K, Li J, Tang JP, Tan CP, Wang H, Zeng Q. Monoclonal antibodiestarget intracellular PRL phosphatases to inhibit cancer metastases in mice.Cancer Biol Ther 2008;7:750–7.

39. Kent D, Copley M, Benz C, Dykstra B, Bowie M, Eaves C. Regulation ofhematopoietic stem cells by the steel factor/KIT signaling pathway. ClinCancer Res 2008;14:1926–30.

40. Masson K, Ronnstrand L. Oncogenic signaling from the hematopoi-etic growth factor receptors c-Kit and Flt3. Cell Signal 2009;21:1717–26.

41. Eppert K, Takenaka K, Lechman ER, Waldron L, Nilsson B, van Galen P,et al. Stem cell gene expression programs influence clinical outcome inhuman leukemia. Nat Med 2011;17:1086–93.

42. Saito Y, Kitamura H, Hijikata A, Tomizawa-Murasawa M, Tanaka S,Takagi S, et al. Identification of therapeutic targets for quiescent,chemotherapy-resistant human leukemia stem cells. Sci Transl Med2010;2:17ra9.

43. Rao S, Lee SY, Gutierrez A, Perrigoue J, Thapa RJ, Tu Z, et al. Inactivation ofribosomal protein L22 promotes transformation by induction of thestemness factor, Lin28B. Blood 2012;120:3764–73.

44. Molenaar JJ, Domingo-Fernandez R, Ebus ME, Lindner S, Koster J, DrabekK, et al. LIN28B induces neuroblastoma and enhances MYCN levels vialet-7 suppression. Nat Genet 2012;44:1199–206.

45. Nguyen LH, RobintonDA, SeligsonMT,Wu L, Li L, RakhejaD, et al. Lin28bis sufficient to drive liver cancer and necessary for its maintenance inmurine models. Cancer Cell 2014;26:248–61.

46. AlamM, Ahmad R, Rajabi H, Kufe D. MUC1-C induces the LIN28B–>LET-7–>HMGA2 axis to regulate self-renewal in NSCLC. Mol Cancer Res2015;13:449–60.

47. Zhou J, Cheong LL, Liu SC, Chong PS, Mahara S, Bi C, et al. The pro-metastasis tyrosine phosphatase, PRL-3 (PTP4A3), is a novel mediator ofoncogenic function of BCR-ABL in human chronic myeloid leukemia. MolCancer 2012;11:72.

www.aacrjournals.org Mol Cancer Res; 15(3) March 2017 303

Activation of LIN28B by PRL-3 Promotes Leukemogenesis

on February 4, 2021. © 2017 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst December 23, 2016; DOI: 10.1158/1541-7786.MCR-16-0275-T

Page 11: LIN28B Activation by PRL-3 Promotes Leukemogenesis and a ... · sion of PRL-3 in human AML cells sustains leukemogenesis in vitro and in vivo. Furthermore, PRL-3 phosphatase activity

2017;15:294-303. Published OnlineFirst December 23, 2016.Mol Cancer Res   Jianbiao Zhou, Zit-Liang Chan, Chonglei Bi, et al.  

like Transcriptional Program in AML−CellLIN28B Activation by PRL-3 Promotes Leukemogenesis and a Stem

  Updated version

  10.1158/1541-7786.MCR-16-0275-Tdoi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mcr.aacrjournals.org/content/suppl/2017/03/03/1541-7786.MCR-16-0275-T.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mcr.aacrjournals.org/content/15/3/294.full#ref-list-1

This article cites 46 articles, 12 of which you can access for free at:

  Citing articles

  http://mcr.aacrjournals.org/content/15/3/294.full#related-urls

This article has been cited by 3 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://mcr.aacrjournals.org/content/15/3/294To request permission to re-use all or part of this article, use this link

on February 4, 2021. © 2017 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst December 23, 2016; DOI: 10.1158/1541-7786.MCR-16-0275-T