9
MOLECULAR AND CELLULAR BIOLOGY, Sept. 2011, p. 3584–3592 Vol. 31, No. 17 0270-7306/11/$12.00 doi:10.1128/MCB.05821-11 Copyright © 2011, American Society for Microbiology. All Rights Reserved. MicroRNA-146a Inhibits Glioma Development by Targeting Notch1 Jie Mei, 1 Robert Bachoo, 2 and Chun-Li Zhang 1 * Department of Molecular Biology 1 and Department of Neurology, 2 University of Texas Southwestern Medical Center, Dallas, Texas 75390-9148 Received 16 June 2011/Returned for modification 20 June 2011/Accepted 23 June 2011 Dysregulated epidermal growth factor receptor (EGFR) signaling through either genomic amplification or dominant-active mutation (EGFR vIII ), in combination with the dual inactivation of INK4A/ARF and PTEN, is a leading cause of gliomagenesis. Our global expression analysis for microRNAs revealed that EGFR activation induces miR-146a expression, which is further potentiated by inactivation of PTEN. Unexpectedly, overexpres- sion of miR-146a attenuates the proliferation, migration, and tumorigenic potential of Ink4a/Arf / Pten / Egfr vIII murine astrocytes. Its ectopic expression also inhibits the glioma development of a human glioblastoma cell line in an orthotopic xenograft model. Such an inhibitory function of miR-146a on gliomas is largely through downregulation of Notch1, which plays a key role in neural stem cell maintenance and is a direct target of miR-146a. Accordingly, miR-146a modulates neural stem cell proliferation and differentiation and reduces the formation and migration of glioma stem-like cells. Conversely, knockdown of miR-146a by microRNA sponge upregulates Notch1 and promotes tumorigenesis of malignant astrocytes. These findings indicate that, in response to oncogenic cues, miR-146a is induced as a negative-feedback mechanism to restrict tumor growth by repressing Notch1. Our results provide novel insights into the signaling pathways that link neural stem cells to gliomagenesis and may lead to new strategies for treating brain tumors. Gliomas are the most frequently observed brain tumors, with glioblastoma multiforme (GBM) being the most common and aggressive form in adults (35). Despite major therapeutic improvements made by combining neurosurgery, chemother- apy, and radiotherapy, the prognosis and survival rate for pa- tients with GBM is still extremely poor (7). The deadly nature of GBM originates from explosive growth and invasive behav- ior, which are fueled by dysregulation of multiple signaling pathways. Epidermal growth factor receptor (EGFR) activa- tion, in cooperation with loss of tumor suppressor functions, such as mutations in Ink4a/Arf and Pten genes, constitutes a lesion signature for GBM (4). Such dysregulated genetic path- ways are sufficient to transform neural stem cells (NSCs) or astrocytes into cancer stem-like cells. This gives rise to high- grade malignant gliomas with a pathological phenotype resem- bling human GBM (5, 59). However, the downstream events underlying these genetic dysregulations in gliomagenic cells have not been fully elucidated. MicroRNAs are 20- to 22-nucleotide noncoding RNA mol- ecules that have emerged as key players in controlling NSC self-renewal and differentiation (11, 57). Aberrant expression of miRNAs, such as miR-21, miR-124, and miR-137, is linked to glioma formation (49). miR-199b-5p and miR-34a impair cancer stem-like cells through the negative regulation of sev- eral components of the Notch pathway in brain tumors (18, 21). The Notch pathway is an evolutionarily conserved signal- ing pathway that plays an important role in neurogenesis (3, 10, 23). Upon binding to its ligand, Delta, the Notch intracellular domain (NICD; the activated form of Notch) is released from the membrane by presenilin/-secretase-mediated cleavage and translocates to the nucleus. In the nucleus, NICD forms a complex with Rbpj and activates the expression of several transcriptional repressors, such as Hes1 and Hes5, which in- hibit neurogenesis (15, 27, 29). Thus, activation of the Notch pathway is essential to maintain both developing and adult NSCs (36). This property of the Notch pathway enables it to promote glioma growth (30), and its inhibition by drugs could abolish glioma stem-like cells and reduce tumorigenesis (17). We show here that miR-146a is specifically induced as a converging downstream target of EGFR and PTEN signaling in immortalized Ink4a/Arf / astrocytes. We further demon- strate that miR-146a acts as a native safeguarding mechanism to restrict the formation of glioma stem-like cells and glioma growth by directly controlling the expression of Notch1. MATERIALS AND METHODS Cell culture, MTT, and anchorage-independent growth assays. We isolated primary NSCs by mechanical dissociation using 1-ml pipettes from embryonic day 14.5 (E14.5) mouse forebrains in growth medium consisting of Dulbecco modified Eagle plus F-12 (DMEM/F12) medium, 1 mM L-glutamine, N2 (Invit- rogen), 20 ng of EGF/ml, and 20 ng of FGF2 (Peprotech)/ml. These cells were cultured as free-floating neurospheres under 37°C and 5% CO 2 . For differenti- ation, we exposed NSCs to DMEM/F12 medium with N2 supplement, further supplemented with 5 M forskolin (FSK) and 0.5% fetal bovine serum (FBS), 1 M retinoic acid (RA) and 0.5% (vol/vol) FBS, or 0.5% (vol/vol) FBS. This was done in 60-mm dishes or eight-well chamber slides coated with laminin (5 g/ml) and poly-L-ornithine (10 g/ml). The Ink4a/Arf / , Ink4a/Arf / Egfr vIII , and Ink4a/Arf / Pten / Egfr vIII astrocytes or human U87 glioma cells were cul- tured in DMEM containing 10% FBS. Glioma stem-like cells from malignant astrocytes or U87 cells were enriched by culturing in NSC medium with growth factors. Cell growth was determined by using the CellTiter 96 nonradioactive cell proliferation assay kit (MTT assay; Promega) according to the manufacturer’s instructions. For anchorage-independent growth by soft agar assay, 2,000 Ink4a/ Arf / Pten / Egfr vIII astrocytes transduced with either wild-type or seed re- * Corresponding author. Mailing address: Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9148. Phone: (214) 648-1670. Fax: (214) 648-1488. E-mail: chun [email protected]. Published ahead of print on 5 July 2011. 3584 on February 13, 2018 by guest http://mcb.asm.org/ Downloaded from

MicroRNA-146a Inhibits Glioma Development by Targeting Notch1

Embed Size (px)

Citation preview

Page 1: MicroRNA-146a Inhibits Glioma Development by Targeting Notch1

MOLECULAR AND CELLULAR BIOLOGY, Sept. 2011, p. 3584–3592 Vol. 31, No. 170270-7306/11/$12.00 doi:10.1128/MCB.05821-11Copyright © 2011, American Society for Microbiology. All Rights Reserved.

MicroRNA-146a Inhibits Glioma Developmentby Targeting Notch1�

Jie Mei,1 Robert Bachoo,2 and Chun-Li Zhang1*Department of Molecular Biology1 and Department of Neurology,2 University of

Texas Southwestern Medical Center, Dallas, Texas 75390-9148

Received 16 June 2011/Returned for modification 20 June 2011/Accepted 23 June 2011

Dysregulated epidermal growth factor receptor (EGFR) signaling through either genomic amplification ordominant-active mutation (EGFRvIII), in combination with the dual inactivation of INK4A/ARF and PTEN, isa leading cause of gliomagenesis. Our global expression analysis for microRNAs revealed that EGFR activationinduces miR-146a expression, which is further potentiated by inactivation of PTEN. Unexpectedly, overexpres-sion of miR-146a attenuates the proliferation, migration, and tumorigenic potential of Ink4a/Arf�/� Pten�/�

EgfrvIII murine astrocytes. Its ectopic expression also inhibits the glioma development of a human glioblastomacell line in an orthotopic xenograft model. Such an inhibitory function of miR-146a on gliomas is largelythrough downregulation of Notch1, which plays a key role in neural stem cell maintenance and is a direct targetof miR-146a. Accordingly, miR-146a modulates neural stem cell proliferation and differentiation and reducesthe formation and migration of glioma stem-like cells. Conversely, knockdown of miR-146a by microRNAsponge upregulates Notch1 and promotes tumorigenesis of malignant astrocytes. These findings indicate that,in response to oncogenic cues, miR-146a is induced as a negative-feedback mechanism to restrict tumor growthby repressing Notch1. Our results provide novel insights into the signaling pathways that link neural stem cellsto gliomagenesis and may lead to new strategies for treating brain tumors.

Gliomas are the most frequently observed brain tumors,with glioblastoma multiforme (GBM) being the most commonand aggressive form in adults (35). Despite major therapeuticimprovements made by combining neurosurgery, chemother-apy, and radiotherapy, the prognosis and survival rate for pa-tients with GBM is still extremely poor (7). The deadly natureof GBM originates from explosive growth and invasive behav-ior, which are fueled by dysregulation of multiple signalingpathways. Epidermal growth factor receptor (EGFR) activa-tion, in cooperation with loss of tumor suppressor functions,such as mutations in Ink4a/Arf and Pten genes, constitutes alesion signature for GBM (4). Such dysregulated genetic path-ways are sufficient to transform neural stem cells (NSCs) orastrocytes into cancer stem-like cells. This gives rise to high-grade malignant gliomas with a pathological phenotype resem-bling human GBM (5, 59). However, the downstream eventsunderlying these genetic dysregulations in gliomagenic cellshave not been fully elucidated.

MicroRNAs are 20- to 22-nucleotide noncoding RNA mol-ecules that have emerged as key players in controlling NSCself-renewal and differentiation (11, 57). Aberrant expressionof miRNAs, such as miR-21, miR-124, and miR-137, is linkedto glioma formation (49). miR-199b-5p and miR-34a impaircancer stem-like cells through the negative regulation of sev-eral components of the Notch pathway in brain tumors (18,21). The Notch pathway is an evolutionarily conserved signal-ing pathway that plays an important role in neurogenesis (3, 10,23). Upon binding to its ligand, Delta, the Notch intracellular

domain (NICD; the activated form of Notch) is released fromthe membrane by presenilin/�-secretase-mediated cleavageand translocates to the nucleus. In the nucleus, NICD forms acomplex with Rbpj and activates the expression of severaltranscriptional repressors, such as Hes1 and Hes5, which in-hibit neurogenesis (15, 27, 29). Thus, activation of the Notchpathway is essential to maintain both developing and adultNSCs (36). This property of the Notch pathway enables it topromote glioma growth (30), and its inhibition by drugs couldabolish glioma stem-like cells and reduce tumorigenesis (17).

We show here that miR-146a is specifically induced as aconverging downstream target of EGFR and PTEN signalingin immortalized Ink4a/Arf�/� astrocytes. We further demon-strate that miR-146a acts as a native safeguarding mechanismto restrict the formation of glioma stem-like cells and gliomagrowth by directly controlling the expression of Notch1.

MATERIALS AND METHODS

Cell culture, MTT, and anchorage-independent growth assays. We isolatedprimary NSCs by mechanical dissociation using 1-ml pipettes from embryonicday 14.5 (E14.5) mouse forebrains in growth medium consisting of Dulbeccomodified Eagle plus F-12 (DMEM/F12) medium, 1 mM L-glutamine, N2 (Invit-rogen), 20 ng of EGF/ml, and 20 ng of FGF2 (Peprotech)/ml. These cells werecultured as free-floating neurospheres under 37°C and 5% CO2. For differenti-ation, we exposed NSCs to DMEM/F12 medium with N2 supplement, furthersupplemented with 5 �M forskolin (FSK) and 0.5% fetal bovine serum (FBS), 1�M retinoic acid (RA) and 0.5% (vol/vol) FBS, or 0.5% (vol/vol) FBS. This wasdone in 60-mm dishes or eight-well chamber slides coated with laminin (5 �g/ml)and poly-L-ornithine (10 �g/ml). The Ink4a/Arf�/�, Ink4a/Arf�/� EgfrvIII, andInk4a/Arf�/� Pten�/� EgfrvIII astrocytes or human U87 glioma cells were cul-tured in DMEM containing 10% FBS. Glioma stem-like cells from malignantastrocytes or U87 cells were enriched by culturing in NSC medium with growthfactors. Cell growth was determined by using the CellTiter 96 nonradioactive cellproliferation assay kit (MTT assay; Promega) according to the manufacturer’sinstructions. For anchorage-independent growth by soft agar assay, 2,000 Ink4a/Arf�/� Pten�/� EgfrvIII astrocytes transduced with either wild-type or seed re-

* Corresponding author. Mailing address: Department of MolecularBiology, University of Texas Southwestern Medical Center, Dallas, TX75390-9148. Phone: (214) 648-1670. Fax: (214) 648-1488. E-mail: [email protected].

� Published ahead of print on 5 July 2011.

3584

on February 13, 2018 by guest

http://mcb.asm

.org/D

ownloaded from

Page 2: MicroRNA-146a Inhibits Glioma Development by Targeting Notch1

gion-mutated miR-146a were plated and cultured in 12-well plates, as describedpreviously (47). Five weeks later, cell colonies in the plates were stained with 0.5ml of 0.005% crystal violet and counted under an inverted microscope. Weconducted each experiment in triplicate.

miRNA microarray and qRT-PCR. We extracted total RNAs from Ink4a/Arf�/� (I), Ink4a/Arf�/� EgfrvIII (IE), or Ink4a/Arf�/� Pten�/� EgfrvIII (IPE)astrocytes using the miReasy minikit (Qiagen). Biological replicates were col-lected. RNA quality was examined by Bioanalyzer (Illumina). Portions (5 �g) ofeach RNA sample were processed for labeling and hybridization to GeneChipmiRNA arrays (Affymetrix) by the Microarray Core Facility at UT SouthwesternMedical Center. After scanning and normalization to control probes, the arraydata was analyzed by VAMPIRE software to identify statistically significantdifferences in gene expression between sample groups (http://genome.ucsd.edu/microarray). For quantitative reverse transcription-PCR (qRT-PCR), 1 �g oftotal RNA was reverse transcribed by using NCode miRNA first-strand synthesisand qRT-PCR kits (Invitrogen). qRT-PCRs were performed in a 384-well plateusing an ABI 7900HT instrument. The PCR program consisted of 2 min at 50°Cand 10 min at 95°C, followed by 40 cycles of 15 s at 95°C and 60 s at 58°C. Primerquality was analyzed by dissociation curves. The expression of miR-146a andNotch1 was normalized to that of U6 and Hprt, respectively.

Lentivirus production and transduction. A genomic fragment encompassingthe miR-146a coding region or a seed region-mutated version was cloned by PCRinto pTomo vector (provided by Inder Verma at Salk Institute). To construct themiRNA sponge, we inserted the synthesized DNA fragment into the BamHI siteof a pCSC-SP-PW-IRES/GFP lentiviral vector and the empty vector as thecontrol. We produced the lentiviruses, determined their titers, and transducedcultured cells according to previously described methods (38, 48). The transduc-tion efficiency was monitored by determining the green fluorescent protein(GFP) expression. All experiments were performed using stably transduced cellswithin six passages.

Cell migration assay. Migration of malignant astrocytes in culture was deter-mined by the “scratch” assay. For this, cells were seeded into a six-well tissueculture dish and allowed to grow to 90% confluence in complete medium with10% FBS. Cells in monolayers were scratched in a single straight line using apipette tip (1 mm in diameter). Wounded monolayers were washed three timeswith culture medium to remove cell debris and then incubated for another 24 h.The migratory distance was measured under a microscope equipped with acamera. A Transwell migration assay was conducted essentially as previouslydescribed (43). Invasive behavior of glioma stem-like cells in vitro was examinedby measuring the migration ability of cultured neurospheres. Neurospheres withsimilar diameters were selected and plated onto six-well culture plates coatedwith laminin (5 �g/ml) and poly-L-ornithine (10 �g/ml) and cultured for 48 h inNSC growth medium. Cell migration and spreading was quantified by measuringthe distance between the edge of the neurosphere and the peripheries of radiallymigrating cells.

Luciferase reporter assay. The 3� untranslated region (3�UTR) of the Notch1gene, which contains one putative miR-146a targeting site, was amplified by PCRand inserted into the SacI and PmeI sites of the pMIR-REPORT vector (Am-bion). To express miR-146a, a 610-bp genomic fragment encompassing thecoding region was cloned by PCR and inserted into the HindIII and BamHI sitesof pCMV6 vector. A seed-sequence-mutated version of miR-146a was used as acontrol for all of the experiments. To create this mutant (pCMV6-miR-146a-mt),we replaced the seed sequence TGAGAACT with GCGGCCGC through site-directed mutagenesis using a QuikChange kit (Stratagene). Similarly, the bindingsite (AGTTCT) for miR-146a within the 3�UTR of the Notch1 gene was replacedwith ACGCGT to generate a control for the luciferase reporter assays. HEK293cells were transiently transfected using Fugene 6 reagent (Roche) in 48-wellplates. The following plasmids were used: pMIR-REPORT was used with eitherpCMV6-miR-146a or its mutant, pCMV6-miR-146a-mt, and pCMV-LacZ wasused as a control to monitor the transfection efficiency. The total plasmid amount(200 ng) was kept constant for each transfection with empty pCMV6 vector.After 48 h, the cells were lysed and assayed for luciferase and �-galactosidaseactivity using a Dual-Light system (Applied Biosystems). Relative reporter ac-tivities were determined by normalizing luminescence units to �-galactosidaseexpression.

Western blotting and immunocytochemistry. Protein expression was examinedby Western blotting, according to a standard procedure. Antibodies against thefollowing proteins were used: �-Actin (Sigma), 1:8,000; GFAP and p-AKT-T308(Cell Signaling Technology), 1:1,000; Sox2 (Chemicon), 1:1,000; Nestin (Aves),1:10,000; and Notch1 (Santa Cruz), 1:1,000. To partially block processing ofNotch1 protein by �-secretase, cells were treated with 10 �M DAPT {N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester} (Sigma) for 36 h.The blots were scanned and quantified by NIH software ImageJ. To examine the

proliferation and cell cycle exit, cultured cells were pulse-labeled with bromode-oxyuridine (BrdU) at 10 �M for 2 h and then fixed and processed for immuno-staining, as previously described (56). Antibodies against the following proteinswere used: BrdU (BD Pharmingen), 1:1,000; Ki67 (Novocastra), 1:500; cleavedcaspase 3 (Cell Signaling Technology), 1:300; and Tuj1 (Covance), 1:500).

Intracranial and subcutaneous cell transplantation model for tumorigenesis.NOD/SCID and nude mice (nu/nu) were purchased from Harlan Laboratories.They were housed in a pathogen-free facility under standard 12-h light/darkcycles and controlled temperature conditions and had free access to food andwater. Experimental protocols were approved by the Institutional Animal Careand Use Committee (IACUC) at the UT Southwestern Medical Center. Astro-cytes (5 � 105 cells) or neurospheres (2 � 105 cells) were respectively implantedinto the right caudoputamen of the brain (for orthotopic transplantation) orsubcutaneous tissues of 5 to 6 weeks old NOD/SCID or nude mice using a25-gauge needle. The injection sites were monitored frequently. Mice trans-planted with neurospheres were euthanized 3 weeks later. The tumors werefinely dissected, imaged, and measured according to the following formula:volume of tumor (in mm3) � (d2 � D)/2, where d and D are the shortest andlongest diameters, respectively. For survival analysis, the mice were euthanizedwhen they demonstrated severe health deterioration according to IACUC guide-lines.

Statistical analysis. Data are expressed as means � the standard deviations.Except for the survival studies, all data were analyzed by two-tailed Student ttests. The Kaplan-Meier survival curves were determined by using GraphPadPrism v5.0 (GraphPad Software, Inc.) and the log-rank test. A P value of 0.05was considered significant.

RESULTS

EGFR activation and PTEN inactivation synergistically in-duce expression of miR-146a. Loss of Ink4a/Arf results in im-mortal growth of primary murine astrocytes. Subsequentactivation of EGFR signaling through expression of a consti-tutively active EGFRvIII mutant transforms these astrocytesinto tumorigenic and neurosphere-forming glioma stem-likecells when cultured in defined medium for NSCs (5, 24). Invivo, concomitant activation of EGFR and inactivation ofInk4a/Arf and Pten produce rapid-onset and fully penetranthigh-grade gliomas that resemble GBM in humans (60), sug-gesting cooperative actions of these signaling pathways on cellproliferation. To confirm this, we performed MTT assays anddirect cell counts to assess the growth rate of Ink4a/Arf�/�,Ink4a/Arf�/� EgfrvIII, or Ink4a/Arf�/� Pten�/� EgfrvIII astro-cytes. Indeed, constitutive activation of EGFR confers Ink4a/Arf�/� astrocytes with a growth advantage that is further en-hanced by ablation of Pten (Fig. 1A and B).

To understand the downstream molecular events of this geneticcooperation and because of the emerging role of microRNA incancers, we performed global expression analysis of micro-RNAs in Ink4a/Arf�/�, Ink4a/Arf�/� EgfrvIII, or Ink4a/Arf�/�

Pten�/� EgfrvIII astrocytes. We identified a total of 19 uniquemiRNAs whose expression is altered by activation of EGFRalone or in combination with loss of Pten (Fig. 1C). Amongthese miRNAs, miR-146a, miR-182, miR-183, and miR-199a-3p are shown to be enriched in diverse cancers, whereasmiR-127 and miR-140 are downregulated in gliomas (32, 46).In contrast to the unchanged expression of miR-146b, miR-146a was significantly induced by EGFRvIII alone or in com-bination with ablation of Pten (Fig. 1C). The change inexpression was subsequently confirmed by qPCR using in-dependent RNA samples (Fig. 1D). We also examined theexpression of miR-146a after acute deletion of Pten throughCre-mediated recombination of floxed Pten alleles in im-mortalized astrocytes (Fig. 1E and F). Pten loss resulted in

VOL. 31, 2011 miR-146a RESTRICTS GLIOMAGENESIS 3585

on February 13, 2018 by guest

http://mcb.asm

.org/D

ownloaded from

Page 3: MicroRNA-146a Inhibits Glioma Development by Targeting Notch1

a 7-fold induction of miR-146a expression. This inductionwas further increased to 9-fold after activation of EGFR.These data indicate a cooperative nature of EGFR, PTEN,and INK4A/ARF signaling on cell proliferation and miR-146a expression.

miR-146a inhibits the proliferation and oncogenic potentialof malignant astrocytes. The induction of miR-146a by EGFRand/or PTEN signaling in Ink4a/Arf�/� astrocytes suggests thatmiR-146a may act as an onco-miR by transducing oncogenicsignals to control cell behavior. To test this hypothesis, weperformed overexpression experiments to examine whetherexogenous miR-146a could further enhance proliferation andtumorigenesis of malignant astrocytes. Due to the extremelylow efficiency of transient transfections in these astrocytes, weused a lentivirus-mediated expression system, in which thecytomegalovirus (CMV) promoter drives the expression ofboth miR-146a and GFP. As a control, we mutated eight nucle-otides within the seed region of miR-146a (Ctrl, Fig. 2A). Such amutation presumably abolishes the interactions of miR-146a withits targets. Examination of GFP expression showed that nearly100% of the cells were transduced by lentivirus. qRT-PCR anal-ysis demonstrated a 26-fold increase of wild-type miR-146a inInk4a/Arf�/� Pten�/� EgfrvIII astrocytes after lentiviral trans-duction (Fig. 2B). Since miR-146a had no effect on apoptosis(Fig. 2E), we examined cell growth by MTT assays and directcell counting. Interestingly, overexpression of miR-146a inhib-ited proliferation of Ink4a/Arf�/� Pten�/� EgfrvIII astrocytes(Fig. 2C and D), indicating that this microRNA may insteadfunction as a tumor suppressor. Indeed, overexpression ofmiR-146a resulted in a 25% reduction in the diameter (Fig.2F) and a 35% decrease in the number (Fig. 2G) of colonies in

an anchorage-independent growth assay, which is an in vitromodel for cellular transformation.

High-grade gliomas exhibit aggressive behavior, which ismanifested by rapid cellular migration under culture condi-tions in vitro (12, 50). Using a standard “scratch” assay after thecells become confluent in culture dishes, we found that en-hanced expression of miR-146a significantly reduced the rateof migration of Ink4a/Arf�/� Pten�/� EgfrvIII astrocytes com-pared to the same cells transduced with a control lentivirus(Fig. 2H). This observation was further confirmed by a Matri-gel transwell migration assay, which showed a 50% reductionof migrating malignant astrocytes upon ectopic expression ofmiR-146a (Fig. 2I). Finally, we examined the in vivo function ofmiR-146a during glioma development after cell transplanta-tion into the right caudoputamen of NOD/SCID mice. Forthis, we grafted an equal number (5 � 105) of Ink4a/Arf�/�

Pten�/� EgfrvIII astrocytes that were transduced with lentivi-ruses expressing either wild-type miR-146a or seed region-mutated miR-146a (Ctrl). Mice were monitored daily formorbidity. A subset of mice was also sacrificed around 3 weeksposttransplantation to examine tumor burden by histology(Fig. 3A). Ectopic expression of miR-146a but not the Ctrlsignificantly reduced the tumor burden and prolonged the sur-vival of mice that were transplanted with Ink4a/Arf�/� Pten�/�

EgfrvIII astrocytes (Fig. 3B). Together, these data suggest thatmiR-146a acts as a tumor suppressor of glioma development.

miR-146a inhibits the formation of glioma stem-like cellsfrom malignant astrocytes. Accumulating evidence suggeststhat self-renewable stem-like cells within the bulk of braintumors are the driving force for initiation and maintenance ofaggressive gliomas (13). These cells share common features

FIG. 1. Induction of miR-146a expression by EGFR and PTEN signaling in immortalized Ink4a/Arf�/� astrocytes. (A and B) Proliferation ofInk4a/Arf�/� (I), Ink4a/Arf�/� EgfrvIII (IE), or Ink4a/Arf�/� Pten�/� EgfrvIII (IPE) astrocytes, as determined by MTT assay (A) or by cell counting(B) (n � 3). (C) Heat map representation of differentially expressed miRNAs identified by microarray analysis. (D) qRT-PCR analysis wasperformed to confirm miR-146a expression induced by EGFR and PTEN signaling using independent RNA samples. (E) Effects of acute Pten losson the expression of miR-146a in Ink4a/Arf�/� Ptenf/f (IPf/f) or Ink4a/Arf�/� Ptenf/f EgfrvIII (IPf/fE) astrocytes. These cells were transduced withadenoviruses expressing either GFP or GFP-Cre. (F) Acute deletion of Pten in Ink4a/Arf�/� Ptenf/f and Ink4a/Arf�/� Ptenf/f EgfrvIII astrocytes.Astrocytes were transduced with adenoviruses expressing GFP (Ad-GFP) or GFP-Cre (Ad-GFP-Cre). Using primers located in the floxed exon5 of the Pten gene, a 157-bp PCR product can be detected in Ad-GFP-transduced but not Ad-GFP-Cre-transduced astrocytes. The expression ofHprt was used as a loading control. Each experiment was performed in triplicate, except for microarrays that were performed in duplicate. *, P 0.05; **, P 0.01; ***, P 0.001.

3586 MEI ET AL. MOL. CELL. BIOL.

on February 13, 2018 by guest

http://mcb.asm

.org/D

ownloaded from

Page 4: MicroRNA-146a Inhibits Glioma Development by Targeting Notch1

with NSCs, including the ability to form neurospheres and theexpression of stem cell markers, such as Sox2 and Nestin (5,21). In fact, neurosphere formation is routinely used to enrichglioma stem-like cells from primary human brain tumors (31,41). The finding that miR-146a inhibits tumorigenesis raisedthat possibility that it may negatively control the behavior ofglioma stem-like cells. We enriched these cells from Ink4a/Arf�/� Pten�/� EgfrvIII astrocytes by culturing them under se-rum-free conditions in the presence of 20 ng of EGF and 20 ngof FGF. After 7 days in culture, control-virus-transduced cellsreadily formed large free-floating spheres, robustly expressingNestin and Sox2 (Fig. 4A to D). In addition, these neuro-

spheres were able to differentiate into GFAP-positive astro-cytes and Tuj1-positive neurons in response to 1% FBS and 5�M FSK treatment, respectively (Fig. 4C). In sharp contrast,ectopic expression of miR-146a markedly diminished the abil-ity of Ink4a/Arf�/� Pten�/� EgfrvIII astrocytes to form neuro-spheres, as indicated by a 40% reduction of sphere size and a30% decline in sphere number (Fig. 4A). miR-146a alsoimpaired the self-renewal ability of these neurospheres, dem-onstrated by a significant reduction of secondary and tertiarysphere formation upon serial passages (Fig. 4B). Furthermore,exogenous miR-146a led to a considerable decrease in theexpression of Nestin and Sox2. This was accompanied by asharp increase in the level of GFAP expression, indicatingenhanced glial differentiation (Fig. 4D). The invasive behaviorof glioma stem-like cells is evidenced by rapid cellular migra-tion from neurospheres when attached to the coated plates inculture. We selected neurospheres with similar diameters fromeither control or miR-146a virus-transduced malignant astro-cytes and plated them onto laminin- and polyornithine-coatedchamber slides. After 48 h, control-virus-transduced cells rap-idly migrated from the edge of the attached spheres and spreadout onto the coated culture surfaces. In contrast, ectopic ex-pression of miR-146a caused a 38% reduction of migratorydistance from the edge of the spheres (Fig. 4E). These dataindicate that miR-146a controls not only the number of gliomastem-like cells but also their invasive behavior in vitro.

miR-146a targets Notch1. How does miR-146a impose aninhibitory function on glioma stem-like cells and tumorigene-sis? Using the Targetscan bioinformatics algorithm, wesearched for direct targets of miR-146a with an emphasis onthose genes that were shown to play a role in cancer stem cells,tumorigenesis or NSCs. Among several potential candidates,Notch1 is most prominent due to its established function inmaintaining NSCs (23, 27) (Fig. 5A). Notch1 also interactswith the EGFR and AKT pathways to positively regulate theproliferation of glioma stem-like cells and tumorigenesis (54).To confirm the regulation of Notch1 by miR-146a, we firstperformed a luciferase reporter assay by linking the 3�UTR of

FIG. 2. miR-146a inhibits the cellular transformation and migra-tion of malignant astrocytes. (A) Schema of miR-146a mutant (Ctrl).The seeding sequence of miR-146a is replaced with a NotI digestionsite. (B) qRT-PCR analysis of miR-146a expression in lentivirus-trans-duced astrocytes. Experiments were performed in triplicate. (C and D)Growth curves of Ink4a/Arf�/� Pten�/� EgfrvIII astrocytes transducedwith lentiviruses expressing either miR-146a or its mutant (Ctrl), asdetermined by MTT assay (C) or cell counting (D) (n � 3). (E) Apop-totic cells were detected by an antibody recognizing cleaved caspase 3,and nuclei were visualized by DAPI (4�,6�-diamidino-2-phenylindole)staining. (F and G) Ectopic expression of miR-146a restrains theclonogenic growth of Ink4a/Arf�/� Pten�/� EgfrvIII astrocytes in a soft-agar assay (n � 3). Representative images are shown (magnification,�100). (H) A scratch assay was performed to examine the effect ofmiR-146a on the migration of malignant astrocytes (n � 3). Repre-sentative images at 0 and 24 h are shown (magnification, �40).(I) Transwell migration assay of Ink4a/Arf�/� Pten�/� EgfrvIII astro-cytes transduced with lentiviruses expressing either miR-146a or itsmutant (Ctrl) (n � 3). *, P 0.05; **, P 0.01; ***, P 0.001.

FIG. 3. miR-146a decreases the tumor burden induced by intracra-nial transplantation of Ink4a/Arf�/� Pten�/� EgfrvIII murine astrocytes.(A) Representative histological analysis of brain gliomas in NOD/SCID mice 3 weeks posttransplantation. In each experiment, a seedregion-mutated version of miR-146a was used as a control (Ctrl).(B) Kaplan-Meier survival curve of mice transplanted with Ink4a/Arf�/� Pten�/� EgfrvIII astrocytes (n � 11 and 8 for the control andmiR-146a groups, respectively; P � 0.002 [log-rank test]).

VOL. 31, 2011 miR-146a RESTRICTS GLIOMAGENESIS 3587

on February 13, 2018 by guest

http://mcb.asm

.org/D

ownloaded from

Page 5: MicroRNA-146a Inhibits Glioma Development by Targeting Notch1

Notch1 to the firefly luciferase gene. The luciferase activity inCOS7 cells was significantly reduced (by 34 to 57%) when thereporter was cotransfected with increasing amount of plasmidsexpressing wild-type miR-146a. Such reduction is specific sincea control plasmid expressing seed region-mutated miR-146a(Ctrl) has little effect on reporter activity. Moreover, wild-typemiR-146a did not change the activity of a luciferase reporterwhen the binding site for miR-146a in 3�UTR of Notch1 wasmutated (Fig. 5B). Western blot analysis further showed thatectopic miR-146a in malignant astrocytes induced a dramaticreduction of Notch1 protein, especially the processed intracel-lular NICD, which is the most predominant form in these gliacells (Fig. 5C). When these cells were treated with DAPT, aninhibitor for �-secretase, to partially block Notch1 processing,

miR-146a significantly reduced the appearance of both full-length Notch1 and NICD (Fig. 5D). The inhibitory role ofmiR-146a on Notch1 is mainly through posttranscriptionalcontrol since it does not change the mRNA level of Notch1(Fig. 5E). Furthermore, phosphorylated AKT, a known down-stream target of Notch1 signaling (22, 58), was also markedlyreduced (Fig. 5C). It is known that individual microRNAs cantarget numerous mRNAs (6), raising the possibility thatNotch1 may not be the major target of miR-146a in gliomastem-like cells. We examined this possibility by performing arescue experiment. Interestingly, ectopic expression of NICDcompletely reversed the inhibitory effect of miR-146a on theformation of glioma stem-like cells. This was indicated by arespective 50 and 60% increase in the number and size ofneurospheres compared to miR-146a-expressing cells (Fig.5F). Importantly, overexpressing NICD did not change thelevel of miR-146a, suggesting that the rescue was not due todownregulation of miR-146a expression (data not shown).

miR-146a regulates NSC proliferation and differentiation.Notch signaling plays an essential role in maintaining NSCs (1,23). Its downregulation promotes neurogenesis during devel-opment or in the adult stage (40, 55). Our finding that miR-

FIG. 4. miR-146a reduces the formation of glioma stem-like cellsand their migration. (A) Glioma stem-like cells were established byculturing 5,000 Ink4a/Arf�/� Pten�/� EgfrvIII astrocytes in NSC me-dium. The number of spheres and their diameters were determined 7days later. A representative morphology of the spheres is shown in theleft panels at �100 magnification (n � 3). (B) Number counts ofprimary, secondary, and tertiary neurospheres established by culturing2,500 Ink4a/Arf�/� Pten�/� EgfrvIII murine astrocytes in NSC medium(n � 3). (C) In the absence of growth factor, neurospheres (derivedfrom Ink4a/Arf�/� Pten�/� EgfrvIII astrocytes) were differentiated intoGFAP-positive astrocytes and Tuj1-positive neurons in response to 1%FBS and 5 �M forskolin (FSK) treatments, respectively. (D) Ectopicexpression of miR-146a induces the glial marker GFAP and suppressesexpression of the stem cell markers Sox2 and Nestin. Actin was used asa loading control for Western blotting. Relative protein level is shown.(E) miR-146a inhibits the spreading and migration of glioma stem-likespheres. Representative photographs were taken at �100 magnifica-tion. Distance was measured from the edge of the sphere to theperiphery of the migrating cells (n � 3). *, P 0.05; **, P 0.01; ***,P 0.001.

FIG. 5. Notch1 is a direct target of miR-146a. (A) Sequence align-ments between 3�UTR of mouse Notch1 and miR-146a. (B) miR-146asuppresses the activity of a luciferase reporter that is linked to the3�UTR of Notch1, but not mutant 3�UTR of Notch1. After normaliza-tion to �-galactosidase expression, which was used as a control fortransfection efficiency, the data were presented as the ratio to that ofempty-vector-transfected cells. (C) miR-146a blocks the protein ex-pression of Notch1 and its downstream target, pAKT, in Ink4a/Arf�/�

Pten�/� EgfrVIII astrocytes. Protein loading was monitored by �-actin.The relative protein level is indicated. (D) Blocking Notch1 processingby DAPT, a �-secretase inhibitor, reveals the inhibitory effect of miR-146a on full-length Notch1 protein in murine astrocytes. (E) miR-146ahas no effect on the expression of Notch1 transcripts, as determined byqRT-PCR. (F) NICD rescues the inhibitory effect of miR-146a on theformation of glioma stem-like cells from Ink4a/Arf�/� Pten�/� EgfrVIII

astrocytes. *, P 0.05; ***, P 0.001.

3588 MEI ET AL. MOL. CELL. BIOL.

on February 13, 2018 by guest

http://mcb.asm

.org/D

ownloaded from

Page 6: MicroRNA-146a Inhibits Glioma Development by Targeting Notch1

146a targets Notch1 expression suggests that miR-146a mayregulate NSC behavior. Indeed, qRT-PCR analysis showed12-fold induction of miR-146a expression after 4 days ofculturing primary mouse E14.5 NSCs under differentiationconditions (Fig. 6A). Ectopic expression of miR-146a throughlentiviral transduction enhanced neuronal differentiation, indi-cated by a 5-fold increase of Tuj1� cells compared to control-virus-transduced NSCs under low EGF and FGF concentra-tions (1 ng/ml) (Fig. 6B and C). Exogenous miR-146a alsoinhibited NSC proliferation, as demonstrated by a 50% reduc-tion in either BrdU� or Ki67� cells (Fig. 6D and E). Suchreduction of proliferation was accompanied by increased cellcycle exit, which was shown by a higher ratio of BrdU� Ki67�

cells over the total number of BrdU� cells after 2 h of BrdUpulse (Fig. 6F). In contrast, miR-146a had no effect on apop-tosis, since an equal number of active caspase 3-positive cellswas observed (Fig. 6D). Together, these results indicate that

miR-146a potentiates neuronal differentiation of NSCs by pro-moting cell cycle exit.

miR-146a prolongs the survival of mice bearing human glio-blastoma cells. We next examined whether the biological roleof miR-146a is evolutionarily conserved in humans. Similar tomurine Notch1 gene, the 3�UTR of its human ortholog alsoharbors a potential binding site for miR-146a, albeit not aperfect match (Fig. 7A). Western blotting showed that ectopicmiR-146a markedly reduced the level of both the full-lengthand the active forms (NICD) of human Notch1 protein in U87glioblastoma cells (Fig. 7B). miR-146a also diminished theability of U87 cells to form glioma stem-like cells. This wasindicated by a 50% decline in sphere number, a 31% reduc-tion in sphere size (Fig. 7C), and a nearly 40% decrease in theappearance of secondary and tertiary spheres (data notshown). When U87 glioblastoma cells were transplanted intothe caudoputamen of NOD/SCID mice, miR-146a reduced thetumor burden and significantly extended the survival of tumor-bearing mice (Fig. 7D and E). Together, these data suggest

FIG. 6. miR-146a regulates normal NSCs. (A) Induction of miR-146a after culture of NSCs under differentiation conditions for 4 days.GF, growth factor bFGF and EGF; RA, retinoic acid; FSK, forskolin.(B and C) Promotion of neuronal differentiation of normal NSCs bymiR-146a. Neurons were stained with an antibody against Tuj1, andnuclei were visualized by DAPI staining. Representative photographswere taken at �400 magnification. (D to F) miR-146a inhibits NSCproliferation by inducing cell cycle exit. (D) Proliferating cells wereexamined by BrdU incorporation and Ki67 staining, which are shownin merged panels with phase-contrast images. Apoptotic cells werestained with an antibody for cleaved caspase 3 (Casp3). Representativephotographs were taken at �200 magnification. (E) Quantification ofproliferating NSCs (BrdU� or Ki67�). (F) miR-146a enhances cellcycle exit, which was measured by the ratio of BrdU� Ki67� cells tototal number of BrdU� cells. Experiments were performed in tripli-cate. **, P 0.01; ***, P 0.001.

FIG. 7. miR-146a promotes the survival of mice transplanted withhuman glioblastoma cells. (A) A recognition site for miR-146a in the3�UTR of human Notch1 gene. (B) miR-146a targets human Notch1.Protein levels were examined by Western blotting of lysates fromhuman U87 glioblastoma cells. Protein loading was monitored by �-ac-tin. The relative protein level is indicated. (C) miR-146a inhibits theformation of glioma stem-like cells from U87. These cells were en-riched in NSC medium. The numbers of spheres and their diameterswere measured 7 days later. (D and E) miR-146a promotes the survivalof tumor-bearing mice by reducing the glioma burden. (D) Histologicalanalysis of brain tumors 3 weeks after intracranial transplantation.(E) Kaplan-Meier survival curve of mice transplanted with human U87glioblastoma cells (n � 6 for each group; P � 0.006 [log-rank test]).

VOL. 31, 2011 miR-146a RESTRICTS GLIOMAGENESIS 3589

on February 13, 2018 by guest

http://mcb.asm

.org/D

ownloaded from

Page 7: MicroRNA-146a Inhibits Glioma Development by Targeting Notch1

that miR-146a function is evolutionarily conserved from miceto humans and is able to modulate the behavior of glioma cellsboth in vitro and in vivo.

Downregulation of miR-146a function enhances tumorigen-esis. miR-146a is induced by oncogenic EGFR and PTENsignaling (Fig. 1). However, its overexpression inhibits the be-havior of glioma stem-like cells and tumorigenesis by targetingNotch1. These seemingly paradoxical data suggest that miR-146a may act as a feedback mechanism to restrict tumorigen-esis (see below). To vigorously test this hypothesis, we used an“miRNA sponge”’ (14) to knock down miR-146a function inInk4a/Arf�/� Pten�/� EgfrvIII astrocytes. This sponge was de-signed to have an imperfect binding site near the miR-146aseed region, with a bulge from positions 10 to 13 (Fig. 8A).This design is presumed to be more effective and stable forinhibiting miR-146a function. We first evaluated its efficacy byusing a Notch1 3�UTR-containing luciferase reporter andfound that the sponge completely reversed the inhibitory effectof miR-146a on this reporter (Fig. 8B). We also examined thismiRNA sponge on the expression of Notch1 through Westernblotting. As expected, transduction of Ink4a/Arf�/� Pten�/�

EgfrvIII astrocytes with sponge-expressing lentiviruses totallyrescued Notch1 protein level that was inhibited by ectopicmiR-146a (Fig. 8C). Moreover, this sponge also enhanced theexpression of endogenous Notch1, most likely by releasing the

inhibition of miR-146a that is induced by EGFRvIII and Pten-loss (Fig. 8D). These data indicate that the use of a miRNAsponge is an effective and specific way to downregulate miR-146a function. We next examined the impact of knocking downmiR-146a on the behavior of glioma stem-like cells and tumor-igenesis. As expected if miR-146a has an inhibitory role, itsdownregulation promoted efficient formation of glioma stem-like cells from Ink4a/Arf�/� Pten�/� EgfrvIII astrocytes by sig-nificantly increasing the number and the size of neurospheres(Fig. 8E and F). When neurospheres (2 � 105 cells) fromlentivirus-transduced malignant astrocytes were grafted intothe flanks of female nude mice, knocking down miR-146afunction using a miRNA sponge caused a 80% increase intumor size by 28 days posttransplantation (Fig. 8G). Therefore,these results indicate that miR-146a is induced as a feedbackmechanism to restrict the oncogenic potential of EGFR andPTEN signaling.

DISCUSSION

We reveal here that constitutively active EGFRvIII cooper-ates with the loss of Pten to synergistically induce expression ofmiR-146a in Ink4a/Arf�/� astrocytes. Counterintuitively, up-regulation of miR-146a inhibits tumor growth and the forma-tion and migration of glioma stem-like cells by both malignant

FIG. 8. Knocking down miR-146a function promotes tumorigenesis. (A) Design of miRNA sponge. (B) Sponge suppresses miR-146a’s functionon activity of a luciferase reporter that is linked to the 3�UTR of the mouse Notch1 gene. The data are presented as the ratio to that of miR-146amutant-transduced cells. (C) miRNA sponge reverses miR-146a-mediated downregulation of Notch1 expression. The relative protein level isindicated. (D) Enhancing endogenous Notch1 expression by miRNA sponge in Ink4a/Arf�/� Pten�/� EgfrVIII astrocytes. (E and F) miRNA spongeagainst miR-146a promotes the formation of glioma stem-like cells from malignant astrocytes (n � 3). (G) Knocking down miR-146a function bysponge exacerbates the tumor burden. The volume of subcutaneous tumors was measured after microdissection (n � 5). *, P 0.05; **, P 0.01;***, P 0.001. (H) A diagram showing the miR-146a-involved signaling network.

3590 MEI ET AL. MOL. CELL. BIOL.

on February 13, 2018 by guest

http://mcb.asm

.org/D

ownloaded from

Page 8: MicroRNA-146a Inhibits Glioma Development by Targeting Notch1

murine Ink4a/Arf�/� Pten�/� EgfrvIII astrocytes and humanglioblastoma cells. We further show for the first time thatmiR-146a directly downregulates Notch1 and potentiates dif-ferentiation of normal NSCs. Knocking down miR-146a func-tion enhances glioma stem-like cell formation and exacerbatestumor burden. These data suggest that miR-146a integratesoncogenic cues to restrict tumor development as a feedbackmechanism (Fig. 8H).

Previously, miR-146a was shown to be induced by endotoxin(lipopolysaccharide) through two consensus NF-�B bindingsites in the promoter region (9). This region is highly homol-ogous between human and mouse, suggesting an evolutionarilyconserved regulatory mechanism for controlling miR-146a ex-pression. NF-�B is constitutively activated in glioma and manyother cancer cells, in which it promotes survival and metastaticpotential of these cells as well as tumorigenesis (28, 52). One ofthe key pathways that controls NF-�B activity in gliomas is thephosphatidylinositol 3-kinase (PI3K) pathway (16, 39), which isa converging point of EGFR- and PTEN-dependent signaltransduction. Activation of receptor tyrosine kinase EGFRleads to recruitment and activation of PI-3 kinase, which phos-phorylates phosphoinositol lipids to generate phosphatidylino-sitol-3-phosphates. These lipids in turn recruit and activateAKT in the plasma membrane through PDK1. The function ofPI3K is antagonized by PTEN, a lipid phosphatase that de-phosphorylates phosphatidylinositol 3,4,5-trisphosphate. Acti-vated AKT further phosphorylates IKK, thus promotingNF-�B activation. The synergistic induction of miR-146a byconstitutively active EGFRvIII and the inactivation of Pten inInk4a/Arf�/� astrocytes may reflect a convergence of these twosignaling pathways on NF-�B activity. Activated AKT alsopromotes Notch1 expression, which subsequently modulatestranscription of Egfr through p53 (44, 45). These data indicatethat miR-146a belongs to an integrated genetic circuit consist-ing of the PTEN, EGFR, NF-�B, and Notch pathways. Theresults from our present study reveal that miR-146a regulatesthe activity of this circuit by targeting Notch1 expression.

Recent studies indicate that certain miRNAs (such as miR-124, miR-137, miR-128, and miR-7) function as tumor sup-pressors (19, 49). These miRNAs are rarely expressed in glio-mas; however, their overexpression restrains proliferation andself-renewal of glioma stem-like cells by promoting neural dif-ferentiation (20, 21). miR-146a is unique in that it is signifi-cantly enriched in the human tissues of skin (melanoma), cer-vical, breast, pancreas and prostate cancers compared to thesame noncancerous tissues (42, 51, 53). Similarly, miR-146awas also upregulated in human glioblastoma tissues and inboth human and mouse primary glioma cell lines (32). In thisregard, increased expression of miR-146a can be viewed as abiomarker for cancers. Unexpectedly, our study reveals that,instead of promoting gliomagenesis through its upregulation,miR-146a rather plays an inhibitory role in restricting the for-mation of glioma stem-like cells and tumor burden. This resultis consistent with a demonstrated role of miR-146a in othercancers, such as pancreatic and breast cancers, where it inhibitscancer progression and invasion (8, 26, 33). A recent reportalso showed a positive correlation of miR-146a expression withthe survival time of gastric cancer patients (25). Furthermore,the overexpression of miR-146a inhibits the proliferation andsurvival of breast, prostate, and pancreatic cancer cells through

the downregulation of other targets in these cells, includingROCK1, EGFR, and MTA-2 (8, 33, 34). Our study addsNotch1 as a major target of miR-146a in glioma cells. Support-ing these cell culture models of tumorigenesis, it was recentlyreported that mice with a deletion of miR-146a spontaneouslydeveloped subcutaneous flank tumors (37). These data clearlyindicate that miR-146a serves as a native molecular brake foroncogenesis (2).

In summary, our current results and other emerging dataindicate that miR-146a constitutes an endogenous feedbacksystem to counteract the oncogenic potential of dysregulatedsignaling pathways, such as activation of EGFR and inactiva-tion of Pten in gliomas. By regulating multiple targets, includ-ing key neural stem cell factor Notch1, a miR-146a-mediatedinnate regulatory mechanism provides an opportunity to devisenovel therapeutic strategies against aggressive and deadlybrain tumors.

ACKNOWLEDGMENTS

We thank the Microarray Core Facility, Yuhua Zou and Wenze Niufor technical help, Rhonda Bassel-Duby and Eric Olson for discus-sions, Hiroyoshi Tanda for careful reading of the manuscript, PamelaJackson for administrative assistance, and Inder Verma (Salk Insti-tute) for providing pTomo lentiviral vector.

C.-L.Z. is a W. W. Caruth, Jr., Scholar in Biomedical Research. Thisstudy was supported by the Whitehall Foundation (2009-12-05), theWelch Foundation (I-1724-01), the National Institutes of Health(1DP2OD006484 and R01NS070981), and a Startup Fund from UTSouthwestern Medical Center to C.-L.Z.

REFERENCES

1. Ables, J. L., et al. 2010. Notch1 is required for maintenance of the reservoirof adult hippocampal stem cells. J. Neurosci. 30:10484–10492.

2. Ameres, S. L., and R. Fukunaga. 2010. Riding in silence: a little snowboard-ing, a lot of small RNAs. Silence 1:8.

3. Androutsellis-Theotokis, A., et al. 2006. Notch signaling regulates stem cellnumbers in vitro and in vivo. Nature 442:823–826.

4. Anonymous. 2008. Comprehensive genomic characterization defines humanglioblastoma genes and core pathways. Nature 455:1061–1068.

5. Bachoo, R. M., et al. 2002. Epidermal growth factor receptor and Ink4a/Arf:convergent mechanisms governing terminal differentiation and transforma-tion along the neural stem cell to astrocyte axis. Cancer Cell 1:269–277.

6. Bartel, D. P. 2009. MicroRNAs: target recognition and regulatory functions.Cell 136:215–233.

7. Behin, A., K. Hoang-Xuan, A. F. Carpentier, and J. Y. Delattre. 2003.Primary brain tumours in adults. Lancet 361:323–331.

8. Bhaumik, D., et al. 2008. Expression of microRNA-146 suppresses NF-�Bactivity with reduction of metastatic potential in breast cancer cells. Onco-gene 27:5643–5647.

9. Cameron, J. E., et al. 2008. Epstein-Barr virus latent membrane protein 1induces cellular MicroRNA miR-146a, a modulator of lymphocyte signalingpathways. J. Virol. 82:1946–1958.

10. Chambers, C. B., et al. 2001. Spatiotemporal selectivity of response toNotch1 signals in mammalian forebrain precursors. Development 128:689–702.

11. Cheng, L. C., E. Pastrana, M. Tavazoie, and F. Doetsch. 2009. miR-124regulates adult neurogenesis in the subventricular zone stem cell niche. Nat.Neurosci. 12:399–408.

12. Chuang, Y. Y., et al. 2004. Role of synaptojanin 2 in glioma cell migrationand invasion. Cancer Res. 64:8271–8275.

13. Das, S., M. Srikanth, and J. A. Kessler. 2008. Cancer stem cells and glioma.Nat. Clin. Pract Neurol. 4:427–435.

14. Ebert, M. S., J. R. Neilson, and P. A. Sharp. 2007. MicroRNA sponges:competitive inhibitors of small RNAs in mammalian cells. Nat. Methods4:721–726.

15. Ehm, O., et al. 2010. RBPJ�-dependent signaling is essential for long-termmaintenance of neural stem cells in the adult hippocampus. J. Neurosci.30:13794–13807.

16. Fan, X., et al. 2002. Genetic profile, PTEN mutation and therapeutic role ofPTEN in glioblastomas. Int. J. Oncol. 21:1141–1150.

17. Fan, X., et al. 2010. NOTCH pathway blockade depletes CD133-positiveglioblastoma cells and inhibits growth of tumor neurospheres and xenografts.Stem Cells 28:5–16.

VOL. 31, 2011 miR-146a RESTRICTS GLIOMAGENESIS 3591

on February 13, 2018 by guest

http://mcb.asm

.org/D

ownloaded from

Page 9: MicroRNA-146a Inhibits Glioma Development by Targeting Notch1

18. Garzia, L., et al. 2009. MicroRNA-199b-5p impairs cancer stem cells throughnegative regulation of HES1 in medulloblastoma. PLoS One 4:e4998.

19. Godlewski, J., H. B. Newton, E. A. Chiocca, and S. E. Lawler. 2010. MicroRNAs andglioblastoma; the stem cell connection. Cell Death Differ. 17:221–228.

20. Godlewski, J., et al. 2008. Targeting of the Bmi-1 oncogene/stem cell renewalfactor by microRNA-128 inhibits glioma proliferation and self-renewal. Can-cer Res. 68:9125–9130.

21. Guessous, F., et al. 2010. microRNA-34a is tumor suppressive in braintumors and glioma stem cells. Cell Cycle 9:1031–1036.

22. Guo, D., et al. 2009. Notch-1 regulates Akt signaling pathway and the ex-pression of cell cycle regulatory proteins cyclin D1, CDK2, and p21 in T-ALLcell lines. Leukoc. Res. 33:678–685.

23. Hitoshi, S., et al. 2002. Notch pathway molecules are essential for themaintenance, but not the generation, of mammalian neural stem cells. GenesDev. 16:846–858.

24. Holland, E. C., W. P. Hively, R. A. DePinho, and H. E. Varmus. 1998. Aconstitutively active epidermal growth factor receptor cooperates with dis-ruption of G1 cell-cycle arrest pathways to induce glioma-like lesions in mice.Genes Dev. 12:3675–3685.

25. Hou, Z., L. Xie, L. Yu, X. Qian, and B. Liu. MicroRNA-146a is down-regulated in gastric cancer and regulates cell proliferation and apoptosis.Med. Oncol., in press.

26. Hurst, D. R., et al. 2009. Breast cancer metastasis suppressor 1 up-regulatesmiR-146, which suppresses breast cancer metastasis. Cancer Res. 69:1279–1283.

27. Imayoshi, I., M. Sakamoto, M. Yamaguchi, K. Mori, and R. Kageyama.2010. Essential roles of Notch signaling in maintenance of neural stem cellsin developing and adult brains. J. Neurosci. 30:3489–3498.

28. Inoue, J., J. Gohda, T. Akiyama, and K. Semba. 2007. NF-�B activation indevelopment and progression of cancer. Cancer Sci. 98:268–274.

29. Kageyama, R., T. Ohtsuka, H. Shimojo, and I. Imayoshi. 2008. DynamicNotch signaling in neural progenitor cells and a revised view of lateralinhibition. Nat. Neurosci. 11:1247–1251.

30. Kanamori, M., et al. 2007. Contribution of Notch signaling activation tohuman glioblastoma multiforme. J. Neurosurg. 106:417–427.

31. Laks, D. R., et al. 2009. Neurosphere formation is an independent predictorof clinical outcome in malignant glioma. Stem Cells 27:980–987.

32. Lavon, I., et al. 2010. Gliomas display a microRNA expression profile rem-iniscent of neural precursor cells. Neurol. Oncol. 12:422–433.

33. Li, Y., et al. 2010. miR-146a suppresses invasion of pancreatic cancer cells.Cancer Res. 70:1486–1495.

34. Lin, S. L., A. Chiang, D. Chang, and S. Y. Ying. 2008. Loss of mir-146afunction in hormone-refractory prostate cancer. RNA 14:417–424.

35. Louis, D. N., et al. 2007. The 2007 WHO classification of tumours of thecentral nervous system. Acta Neuropathol. 114:97–109.

36. Louvi, A., and S. Artavanis-Tsakonas. 2006. Notch signaling in vertebrateneural development. Nat. Rev. Neurosci. 7:93–102.

37. Lu, L. F., et al. 2010. Function of miR-146a in controlling Treg cell-mediatedregulation of Th1 responses. Cell 142:914–929.

38. Marumoto, T., et al. 2009. Development of a novel mouse glioma modelusing lentiviral vectors. Nat. Med. 15:110–116.

39. Mischel, P. S., and T. F. Cloughesy. 2003. Targeted molecular therapy ofGBM. Brain Pathol. 13:52–61.

40. Oya, S., et al. 2009. Attenuation of Notch signaling promotes the differen-

tiation of neural progenitors into neurons in the hippocampal CA1 regionafter ischemic injury. Neuroscience 158:683–692.

41. Pellegatta, S., et al. 2006. Neurospheres enriched in cancer stem-like cellsare highly effective in eliciting a dendritic cell-mediated immune responseagainst malignant gliomas. Cancer Res. 66:10247–10252.

42. Philippidou, D., et al. 2010. Signatures of microRNAs and selected mi-croRNA target genes in human melanoma. Cancer Res. 70:4163–4173.

43. Piao, Y., L. Lu, and J. de Groot. 2009. AMPA receptors promote perivas-cular glioma invasion via �1 integrin-dependent adhesion to the extracellularmatrix. Neurol. Oncol. 11:260–273.

44. Purow, B. W., et al. 2008. Notch-1 regulates transcription of the epidermalgrowth factor receptor through p53. Carcinogenesis 29:918–925.

45. Rajasekhar, V. K., et al. 2003. Oncogenic Ras and Akt signaling contributeto glioblastoma formation by differential recruitment of existing mRNAs topolysomes. Mol. Cell 12:889–901.

46. Rao, S. A., V. Santosh, and K. Somasundaram. 2010. Genome-wide expres-sion profiling identifies deregulated miRNAs in malignant astrocytoma.Modern Pathol. 23:1404–1417.

47. Rich, J. N., et al. 2001. A genetically tractable model of human gliomaformation. Cancer Res. 61:3556–3560.

48. Scherr, M., et al. 2007. Lentivirus-mediated antagomir expression for spe-cific inhibition of miRNA function. Nucleic Acids Res. 35:e149.

49. Silber, J., et al. 2008. miR-124 and miR-137 inhibit proliferation of glioblas-toma multiforme cells and induce differentiation of brain tumor stem cells.BMC Med. 6:14.

50. Soroceanu, L., T. J. Manning, Jr., and H. Sontheimer. 1999. Modulation ofglioma cell migration and invasion using Cl� and K� ion channel blockers.J. Neurosci. 19:5942–5954.

51. Volinia, S., et al. 2006. A microRNA expression signature of human solidtumors defines cancer gene targets. Proc. Natl. Acad. Sci. U. S. A. 103:2257–2261.

52. Wang, H., W. Zhang, H. J. Huang, W. S. Liao, and G. N. Fuller. 2004.Analysis of the activation status of Akt, NF�B, and Stat3 in human diffusegliomas. Lab. Invest. 84:941–951.

53. Wang, X., et al. 2008. Aberrant expression of oncogenic and tumor-suppres-sive microRNAs in cervical cancer is required for cancer cell growth. PLoSOne 3:e2557.

54. Xu, P., et al. 2010. The oncogenic roles of Notch1 in astrocytic gliomas invitro and in vivo. J. Neuro-Oncol. 97:41–51.

55. Yoon, K., and N. Gaiano. 2005. Notch signaling in the mammalian centralnervous system: insights from mouse mutants. Nat. Neurosci. 8:709–715.

56. Zhang, C. L., Y. Zou, W. He, F. H. Gage, and R. M. Evans. 2008. A role foradult TLX-positive neural stem cells in learning and behaviour. Nature451:1004–1007.

57. Zhao, C., et al. 2010. MicroRNA let-7b regulates neural stem cell prolifer-ation and differentiation by targeting nuclear receptor TLX signaling. Proc.Natl. Acad. Sci. U. S. A. 107:1876–1881.

58. Zhao, N., Y. Guo, M. Zhang, L. Lin, and Z. Zheng. 2010. Akt-mTORsignaling is involved in Notch-1-mediated glioma cell survival and prolifer-ation. Oncol. Rep. 23:1443–1447.

59. Zheng, H., et al. 2008. p53 and Pten control neural and glioma stem/pro-genitor cell renewal and differentiation. Nature 455:1129–1133.

60. Zhu, H., et al. 2009. Oncogenic EGFR signaling cooperates with loss oftumor suppressor gene functions in gliomagenesis. Proc. Natl. Acad. Sci.U. S. A. 106:2712–2716.

3592 MEI ET AL. MOL. CELL. BIOL.

on February 13, 2018 by guest

http://mcb.asm

.org/D

ownloaded from