mir-146a exerts differential effects on melanoma growth...

16
Oncogenes and Tumor Suppressors miR-146a Exerts Differential Effects on Melanoma Growth and Metastatization Monica Raimo 1,2 , Francesca Orso 1,2,3 , Elena Grassi 1,2 , Daniela Cimino 1,2,3 , Elisa Penna 1,2 , Cristiano De Pitt a 4 , Michael B. Stadler 5 , Luca Primo 6 , Enzo Calautti 1,2 , Pietro Quaglino 7 , Paolo Provero 1,2 , and Daniela Taverna 1,2,3 Abstract Malignant melanoma is the most aggressive form of skin cancer; therefore, it is crucial to disclose its underlying molecular mechanisms. MicroRNAs (miRNAs) are small endogenous non- coding RNAs able to posttranscriptionally downregulate the expression of direct target genes. Using a melanoma progression model, miR-146a was identied as a key double-acting player in melanoma malignancy. In fact, miR-146a is able to enhance tumor growth, while it suppresses dissemination. It was deter- mined that miR-146a coordinated melanoma cell growth by its direct targets lunatic fringe (LFNG) and NUMB, which operate on the NOTCH/PTEN/Akt pathway; while inhibition of metastasis formation was linked to decreased expression of ITGAV and ROCK1. Relevantly, miR-146a expression correlated with melanoma recurrence and was enriched in both patient-derived melanoma and cutaneous metastasis specimens, while its direct targets were depleted. However, miR-146a levels drop in circu- lating tumor cells (CTCs), suggesting the necessity for miR-146a expression to uctuate during tumor progression in order to favor tumor growth and allow dissemination. This study reconciles the contradictory biologic functions of miR-146a in melanoma pro- gression and unravels distinct molecular mechanisms that need to be considered for therapeutic interventions. Implications: miR-146a controls melanoma progression in a dual way, promoting growth and inhibiting dissemination; however, it is poorly expressed in CTCs, resulting in overall tumor spreading and distant-site colonization. Mol Cancer Res; 14(6); 54862. Ó2016 AACR. Introduction Malignant melanoma is the most aggressive form of skin cancer and the sixth most common cancer in the United States; it accounts for only 4% of cases but for as many as 74% of all skin cancer deaths (1). Pathogenesis of melanoma is characterized by mutations of MAPK pathway components, N-RAS, B-RAF, PTEN and p53, while metastatic transition is accompanied by gain of functions of transcription factors like NF-kB and STATs and loss of TFAP2 family members (2, 3). Given the complexity of networks and pathways involved, disclosing the molecular mechanisms underlying melanoma formation and progression is crucial to provide novel therapeutic strategies (4, 5). MicroRNAs (miRNAs) are small endogenous noncoding RNAs able to posttranscription- ally regulate the expression of target genes via sequence-specic interactions with the 3 0 untranslated regions (UTR) of cognate mRNAs (6). Given that an individual miRNA is able to inhibit multiple distinct mRNAs, they act as fundamental regulators of tumor establishment and progression in different types of cancer, including melanoma (5). miRNAs can act either as oncogenes or as tumor suppressor genes. Among others, miR-7, miR-15b, miR- 182, miR-19, miR-21, miR-192, miR-221/222, and miR-214 are upregulated during melanoma growth and invasion, while let-7b, miR-145, the miR-200 family, and miR-206 are downmodulated (4, 79). In this study, we evidenced that miR-146a exerts a dual role in melanoma cells, favoring tumor cell growth, while inhibiting cell invasion. However, miR-146a expression is strictly modulated across the various steps of melanoma progression; it is high in both primary and distant-site tumors, where it promotes tumor cell growth, but it drops in circulating tumor cells (CTCs), there- fore favoring the escaping from the primary tumor mass. Different players operate in the various moments of tumor progression: the targeting of lunatic fringe (LFNG) and NUMB and the subsequent induction of the NOTCH1/PTEN/Akt pathway triggers tumor growth, while the inhibition of ITGAV and ROCK1 is crucial to block cell invasion. Here, we present the molecular mechanisms coordinated by miR-146a during melanoma malignancy that need to be carefully considered for any therapeutic intervention. Materials and Methods Vectors, primers, antibodies, reagents, RNA, protein, and human melanoma samples analysis Detailed information about the vectors, primers, and anti- bodies, RNA extraction, RNA and protein analysis methods 1 Molecular Biotechnology Center (MBC), University of Torino,Torino, Italy. 2 Department of Molecular Biotechnology and Health Sciences, University of Torino,Torino, Italy. 3 Center for Molecular Systems Biol- ogy, University of Torino,Torino, Italy. 4 Department of Biology, Uni- versity of Padova, Padova, Italy. 5 Friederich Miescher Institute and Swiss Institute of Bioinformatics, Basel, Switzerland. 6 Department of Oncology, University of Torino and Candiolo Cancer Institute-FPO IRCCS, Candiolo (TO), Italy. 7 Section of Dermatology, Department of Medical Sciences, University of Torino,Torino, Italy. Note: Supplementary data for this article are available at Molecular Cancer Research Online (http://mcr.aacrjournals.org/). Corresponding Author: Daniela Taverna, MBC and Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza, 52, 10126 Torino, Italy. Phone: 39-011-670-6497; Fax: 39-011-670-6432; E-mail: [email protected] doi: 10.1158/1541-7786.MCR-15-0425-T Ó2016 American Association for Cancer Research. Molecular Cancer Research Mol Cancer Res; 14(6) June 2016 548 on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Upload: nguyenliem

Post on 16-Feb-2019

236 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

Oncogenes and Tumor Suppressors

miR-146a Exerts Differential Effects on MelanomaGrowth and MetastatizationMonica Raimo1,2, Francesca Orso1,2,3, Elena Grassi1,2, Daniela Cimino1,2,3,Elisa Penna1,2, Cristiano De Pitt�a4, Michael B. Stadler5, Luca Primo6, Enzo Calautti1,2,Pietro Quaglino7, Paolo Provero1,2, and Daniela Taverna1,2,3

Abstract

Malignant melanoma is the most aggressive form of skincancer; therefore, it is crucial to disclose its underlying molecularmechanisms. MicroRNAs (miRNAs) are small endogenous non-coding RNAs able to posttranscriptionally downregulate theexpression of direct target genes. Using a melanoma progressionmodel, miR-146a was identified as a key double-acting player inmelanoma malignancy. In fact, miR-146a is able to enhancetumor growth, while it suppresses dissemination. It was deter-mined that miR-146a coordinated melanoma cell growth by itsdirect targets lunatic fringe (LFNG) and NUMB, which operate onthe NOTCH/PTEN/Akt pathway; while inhibition of metastasisformation was linked to decreased expression of ITGAVand ROCK1. Relevantly, miR-146a expression correlated withmelanoma recurrence and was enriched in both patient-derived

melanoma and cutaneous metastasis specimens, while its directtargets were depleted. However, miR-146a levels drop in circu-lating tumor cells (CTCs), suggesting the necessity for miR-146aexpression to fluctuate during tumor progression in order to favortumor growth and allow dissemination. This study reconciles thecontradictory biologic functions of miR-146a in melanoma pro-gression andunravels distinctmolecularmechanisms that need tobe considered for therapeutic interventions.

Implications: miR-146a controls melanoma progression in adual way, promoting growth and inhibiting dissemination;however, it is poorly expressed in CTCs, resulting in overalltumor spreading and distant-site colonization. Mol Cancer Res;14(6); 548–62. �2016 AACR.

IntroductionMalignantmelanoma is themost aggressive formof skin cancer

and the sixth most common cancer in the United States; itaccounts for only 4% of cases but for as many as 74% of all skincancer deaths (1). Pathogenesis of melanoma is characterized bymutations of MAPK pathway components, N-RAS, B-RAF, PTENand p53, while metastatic transition is accompanied by gain offunctions of transcription factors likeNF-kB and STATs and loss ofTFAP2 family members (2, 3). Given the complexity of networksand pathways involved, disclosing the molecular mechanismsunderlying melanoma formation and progression is crucial toprovide novel therapeutic strategies (4, 5). MicroRNAs (miRNAs)are small endogenous noncoding RNAs able to posttranscription-

ally regulate the expression of target genes via sequence-specificinteractions with the 30 untranslated regions (UTR) of cognatemRNAs (6). Given that an individual miRNA is able to inhibitmultiple distinct mRNAs, they act as fundamental regulators oftumor establishment and progression in different types of cancer,including melanoma (5). miRNAs can act either as oncogenes oras tumor suppressor genes. Among others, miR-7, miR-15b, miR-182, miR-19, miR-21, miR-192, miR-221/222, and miR-214 areupregulated duringmelanoma growth and invasion, while let-7b,miR-145, the miR-200 family, and miR-206 are downmodulated(4, 7–9).

In this study, we evidenced that miR-146a exerts a dual role inmelanoma cells, favoring tumor cell growth, while inhibiting cellinvasion. However, miR-146a expression is strictly modulatedacross the various steps of melanoma progression; it is high inboth primary and distant-site tumors, where it promotes tumorcell growth, but it drops in circulating tumor cells (CTCs), there-fore favoring the escaping from the primary tumormass.Differentplayers operate in the variousmoments of tumor progression: thetargeting of lunatic fringe (LFNG) andNUMB and the subsequentinduction of the NOTCH1/PTEN/Akt pathway triggers tumorgrowth, while the inhibition of ITGAV and ROCK1 is crucial toblock cell invasion. Here, we present the molecular mechanismscoordinated by miR-146a during melanoma malignancy thatneed to be carefully considered for any therapeutic intervention.

Materials and MethodsVectors, primers, antibodies, reagents, RNA, protein, andhuman melanoma samples analysis

Detailed information about the vectors, primers, and anti-bodies, RNA extraction, RNA and protein analysis methods

1Molecular Biotechnology Center (MBC), University of Torino, Torino,Italy. 2Department of Molecular Biotechnology and Health Sciences,University of Torino, Torino, Italy. 3Center for Molecular Systems Biol-ogy, University of Torino, Torino, Italy. 4Department of Biology, Uni-versity of Padova, Padova, Italy. 5Friederich Miescher Institute andSwiss Institute of Bioinformatics, Basel, Switzerland. 6Department ofOncology, University of Torino and Candiolo Cancer Institute-FPOIRCCS, Candiolo (TO), Italy. 7Section of Dermatology, Department ofMedical Sciences, University of Torino, Torino, Italy.

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

Corresponding Author: Daniela Taverna, MBC and Department of MolecularBiotechnology and Health Sciences, University of Torino, Via Nizza, 52, 10126Torino, Italy. Phone: 39-011-670-6497; Fax: 39-011-670-6432; E-mail:[email protected]

doi: 10.1158/1541-7786.MCR-15-0425-T

�2016 American Association for Cancer Research.

MolecularCancerResearch

Mol Cancer Res; 14(6) June 2016548

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 2: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

used in this study is available in the Supplementary Materialsand Methods section. miRNA precursors and inhibitorsand assays for quantitative real-time PCR (qRT-PCR) detectionwere purchased from Applied Biosystems. Luciferase assaysand immunohistochemistry were performed using theDual-Luciferase Reporter System (Promega) or the Anti-MouseHRP-DAB Cell and Tissue Staining Kit, R&D Systems kits,respectively. Melanoma samples were collected from theDepartment of Medical Sciences, University of Torino, Torino,Italy.

In vitro growth, adhesion, and migration assaysFor proliferation and anchorage-independent growth assays,

cells were plated in 96-well plates or in 6-cm bacterial dishes(mixed with agar), and growth was followed up to 5 or 21 days,respectively. For adhesion assays, cells were seeded onto 0.5mg/mL Vitronectin V8379 (Sigma-Aldrich) precoated 96-wellplates, and adhesion was assessed 1 hour later. To measuremigration, cells were seeded in the upper chamber of Trans-wells with or without Matrigel (Becton Dickinson) and migra-tion to the lower side was assessed 18 hours later. For trans-endothelial migration assays, Transwell inserts were coveredwith a human umbilical vein endothelial cell (HUVEC)-GFPcells monolayer.

In vivo tumor and metastasis and extravasation assaysFor all, cells were subcutaneously injected in the flank or into

the tail vein of mice; for long-term experiments, tumor and/orlung colonies growth was evaluated from 4 to 6 weeks later, whilefor extravasation assays lung seeding was assessed 48 hours later,as described in Penna, E. and Taverna, D. (2014). In vivo Extrav-asation Assay. Bio-protocol 4(4): e1051. http://www.bio-protocol.org/e1051.

Circulating tumor cells isolationTo isolate circulating tumor cells (CTCs), blood was col-

lected from heart-punctured mice and put in culture plateswith melanoma cells medium for 3 days; subsequently,attached cells were washed and cultured in fresh mediumfor at least 4 days. Puromycin was added to the medium toselect human melanoma cells. Cells were then washed thor-oughly to remove nonadherent cells, fixed with 2.5% glu-taraldehyde and stained with 0.1% crystal violet or total RNAwas isolated.

Ingenuity Pathway Analysis (IPA) systemThe Ingenuity Pathway knowledge Base (http://www.inge-

nuity.com) is currently the world's largest database of knowl-edge on biological networks, with annotations organized byexperts. We exploited this database to look for Canonical Path-ways and Gene Ontology functions characterizing miR-146a puta-tive target genes.

Statistical analysesData are presented as mean � standard error of the mean and

two-tailed Student t test or one- or two-tailed Mann–Whitneytest were used for comparison, with �, P < 0.05; ��, P < 0.01;���, P < 0.001 considered to be statistically significant. n.s. indi-cates a not statistically significant P value.

ResultsmiR-146a expression is upregulated during melanomaprogression and enhances in vitro and in vivo cell and tumorgrowth

With the goal of identifying microRNAs involved in malig-nant melanoma progression, we performed miRNA profilingon a human melanoma cellular model consisting of a poorlyaggressive B-RAF V600E-mutated parental cell line (A375P) andits four variants MA-1, MC-1, MA-2, and MC-2 derived fromone (MA-1 and MC-1) or two (MA-2 and MC-2) subsequentrounds of injections in mice and showing progressivelyincreased metastatic potential (10). Ninety-eight significantlydownmodulated or upmodulated miRNAs have been identifiedwhen invasive (MA-1, MC-1, MA-2, and MC-2) versus nonin-vasive (A375P) cells were used (Supplementary Fig. S1 andSupplementary Table S1). Among the upregulated miRNAs, wefocused on miR-146a and, with the intent to validate micro-array results, we analyzed its expression in our melanomamodel and in the unrelated B-RAF V600E-mutated 1300-Meland SK-Mel-28 or B-RAF-wild-type WK-Mel melanoma celllines by qRT-PCR analysis. Interestingly, only B-RAF–mutatedcell lines showed high miR-146a expression (Fig. 1A). Rele-vantly, when patient-derived melanoma samples were evalu-ated, miR-146a expression changed accordingly, being low inin situ melanomas, while increasing in invasive samples as wellas in cutaneous metastases (qRT-PCR, Fig. 1B). Here, it isrelevant to refer to ref. 11 for melanoma classification, whichconsiders thickness (mm), ulceration, and mitotic status asmain parameters to distinguish more or less aggressive mela-nomas. Moreover, it is important to point out that the selectionof the tumor areas was performed on hematoxylin and eosin(H&E) archival slides originally used for histopathologic diag-nosis. The pathologist marked the areas of tumor enrichmenton the slide from histologic specimens and recorded the per-centage and absolute number of cancer cells. In this way, asimilar number of tumor cells were evaluated for all kinds oftumors. Furthermore, when we explored the GSE62372 dataset(12), composed of 92 primary melanomas, we observed thatmiR-146a expression significantly correlated with tumor thick-ness and recurrence (Supplementary Fig. S2A–S2B).

Based on all the evidence, we aimed to study miR-146ainvolvement in cell and tumor growth; therefore, we silencedmiR-146a in MA-2 cells by stable lentiviral infections with miR-146a–specific antisense sponges, compared with empty vectors(pLentiGFP-sponge or -EV), or in MC-1 cells by transienttransfection of miR-146a antisense inhibitors compared withnegative controls (anti-146a or anti-Cntrl). Sponge efficacy wasevaluated, as shown in Supplementary Fig. S3. Conversely, weinduced miR-146a expression in MA-2 and 1300-Mel cells bystable lentiviral transduction of miR-146a (pLemiR-146a), orupon transient transfections of pre-miR-146a (pre-146a), com-pared with empty controls (pLemiR-EV, pre-Cntrl). The efficacyof miR-146a modulations was tested by qRT-PCR (Supplemen-tary Fig. S4A–S4E). Interestingly, miR-146a stable or transientdownmodulation (pLentiGFP-sponge and anti-146a) in MA-2and MC-1 cells strongly impaired cell proliferation up to 5 daysas well as cell anchorage-independent growth in soft agar,in vitro (Fig. 1C and D, a–b and Supplementary Fig. S5A, top,and Supplementary Fig. S5B, a–b). Conversely, miR-146a over-expression (pLemiR-146a and pre-146a) in MA-2 and 1300-Mel cells reverted these effects, as it increased both growth rate

Dual Role of miR-146a in Tumor Progression

www.aacrjournals.org Mol Cancer Res; 14(6) June 2016 549

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 3: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

Figure 1.miR-146a expression is upregulated during melanoma progression and enhances in vitro and in vivo cell and tumor growth. A, miR-146a expression in A375P,MA-1, MC-1, MA-2, MC-2, 1300-Mel, SK-Mel-28, and WK-Mel cells was assessed by qRT-PCR. Results are shown as fold changes (mean � SEM) relative toA375P cells, normalized on U44 RNA levels. (Continued on the following page.)

Raimo et al.

Mol Cancer Res; 14(6) June 2016 Molecular Cancer Research550

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 4: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

and colony-forming ability (Fig. 1C and D, c–d and Supple-mentary Fig. S5A, bottom). Relevantly, we observed that miR-146a affects cell proliferation by enhancing growth rather thaninhibiting cell death; in fact, it resulted irrelevant in cellapoptosis and anoikis studies (Supplementary Fig. S5C). Moreimportantly, miR-146a inhibition strongly impaired primarytumor growth, as assessed by subcutaneous injections of miR-146a–modulated MA-2 cells into the flank of NSG mice;compared with control cells, miR-146a–downmodulated(pLentiGFP-sponge) tumors resulted significantly smaller, 5weeks after injection. Instead, miR-146a–overexpressing (pLe-miR-146a) cells generated bigger tumor masses than emptycontrols (Fig. 1E). Moreover, proliferating cell nuclear antigen(PCNA) staining of primary tumors and distant lung colonies(5 weeks after subcutaneous injections of MA-2 cells) revealedthat miR-146a–downmodulated cells were less able to prolif-erate in vivo, both in primary and in disseminated masses,compared with controls (pLentiGFP-sponge vs. -EV; Fig. 1F,a–b and c–d). In conclusion, all these results reveal the crucialinvolvement of miR-146a in primary melanoma and distantmetastasis growth as a proproliferative player.

miR-146a inhibits cell extravasation and metastasis formationin vivo and is poorly expressed in CTCs

Due to the fact that miR-146a is enriched during melanomaprogression, we investigated its possible involvement in meta-static traits, such as cell motility, migration, and distant-sitecolonization. miR-146a stable (pLentiGFP-sponge) or transient(anti-146a) downmodulation in MA-2 and MC-1 cells led to a30% to 50% increase in migration and invasion compared withcontrols, as evaluated by in vitro Transwell assays in the presenceor absence of Matrigel. Instead, transient (pre-146a) or stable(pLemiR-146a) overexpression in 1300-Mel, MA-2, SK-Mel-28,and WK-Mel cells reduced cell migration and invasion of about20% to 50% compared with controls (Fig. 2A and B and Fig.S6A–S6C). miR-146a level modulation was always assessed, asin Supplementary Fig. S4. Considering that malignant cells haveto detach from the primary tumor mass, enter and survive in theblood vessels and then extravasate, we evaluated miR-146ainvolvement in an in vitro transendothelial migration assay. Forthis purpose, we seeded CMRA-labeled (red) miR-146a–over-expressing or control MA-2 or WK-Mel cells in the upperchambers of fibronectin-coated Transwells, covered by a con-fluent GFP-transduced HUVEC monolayer, and evaluated theendothelium remodeling and melanoma cells migration (Fig.2C; Supplementary Fig. S6D). In the absence of melanoma cells(a) the HUVECs monolayer remained intact. Notably, in thepresence of miR-146a–overexpressing (pre-146a) MA-2 or

WK-Mel cells, the endothelial monolayer was more compact,compared with controls (pre-Cntrl, b–c) and accordingly,miR-146a–overexpressing cells showed a 50% to 60% reductionin their ability to migrate through the endothelial layer (d–e).miR-146a's ability to inhibit cell extravasation was reproducedin vivo, where miR-146a–overexpressing cells (pre-146a) extrav-asated poorly from the lung vessels to the tissue parenchyma 48hours after tail-vein injections in CD1 nude mice, comparedwith control cells (pre-Cntrl; Fig. 2D, e–f). Equal lodging of thelungs was assessed 2 hours after injections (Fig. 2D, c–d).Intravascular localization of cells at 2 hours, and subsequentextravasation at 48 hours was proved by immunofluorescenceanalysis (Fig. 2D, a–b and Supplementary Fig. S6E). miR-146a–dependent impairment of cell extravasation resulted crucial inthe inhibition of metastasis formation. In fact, as shown in Fig.2E, a–b, miR-146a–overexpressing cells (pWPT-146a) generateda significant lower number of lung metastases compared withcontrols (pWPT-EV), 5 weeks after injections into the tail veinof immunocompromised mice. Efficient miR-146a overex-pression was verified (Supplementary Fig. S4I). Furthermore,miR-146a was found to affect cell dissemination from primarytumors: miR-146a–downmodulated (pLentiGFP-sponge) MA-2cells injected into the flank of NSG mice were more proficient inthe dissemination from tumors to lungs, compared with con-trols (pLentiGFP-EV; Fig. 2E, c–d; Supplementary Fig. S7). Onthe contrary, lung colonization was impaired in miR-146a–overexpressing cells (pLemiR-146a vs. -EV), as shown in Fig.2E, e–f. Taken together, all our experiments demonstrate thatmiR-146a favors tumor andmetastasis growth but it impairs celldissemination. Aiming to understand the dual and apparentlycontradictory role of miR-146a, we investigated its expression inCTCs. For this purpose, we collected CTCs from immunosup-pressed mice 5 weeks after subcutaneous injections of MA-2 cellsand evaluated miR-146a expression compared with primarytumors and lung metastases by qRT-PCR. Human CTCs viabilitywas proven to be comparable with control cells, as assessed bythe measurements of the survival markers BAX and BCL2(qRT-PCR), while murine white blood cells contamination wasexcluded by the analysis of PTPRC1, a specific bone marrowmarker (qRT-PCR; Supplementary Fig. S8A and S8B). As in Fig.2F, miR-146a levels resulted very low in CTCs while high both intumors and in disseminated lung colonies. Concomitantly, twoother miRNAs involved in melanoma metastatization, miR-21and miR-214, were unchanged (Supplementary Fig. S8C), indi-cating that miR-146a downmodulation is a specific trait ofescaping tumor cells. In fact, we hypothesize that primarytumors are heterogeneous and contain cells with high orlow levels of miR-146a at different percentages. While cells

(Continued.) B, miR-146a expression in in situ melanomas (n ¼ 18), invasive melanomas (n ¼ 34), and cutaneous metastases (n ¼ 25) was evaluated byqRT-PCR. Results are shown as box-and-whisker plots of the fold changes relative to in situmelanomas, normalized on U44 RNA levels. C andD, in vitro proliferation(C) and anchorage-independent growth (D) of miR-146a–downmodulated (pLentiGFP-sponge vs. pLentiGFP-EV) or miR-146a–overexpressing (pre-146a vs.pre-Cntrl, or pLemiR-146a vs. pLemiR-EV) MA-2 cells was evaluated starting from 12 hours after transfection up to 5 days or 21 days after transfection, respectively.Results are shownasmean�SEMof the optical density (OD) or of the areaoccupiedby colonies or aspictures (a–d); three independent experimentswere performed(in sestuplicate or triplicate, respectively) and results are shown as representative ones. Bar, 4 mm. E, primary tumor growth was analyzed 5 weeks aftersubcutaneous injections of NSGmice of MA-2 cells stably downmodulated for miR-146a (pLentiGFP-sponge vs. -EV), or overexpressing miR-146a (pLemiR-146a vs.-EV); tumor weight was evaluated in 5 mice per group. Tumor growth upon miR-146a overexpression was evaluated in two independent experiments; one isshown. F, Primary tumors (a, b) or lung metastases (c, d) generated in NSG mice 5 weeks after subcutaneous injection of miR-146a–downmodulated MA-2 cells(pLentiGFP-sponge) or of control cells (pLentiGFP-EV) were stained in IHC for the proliferating cell nuclear antigen (PCNA); nuclei were counterstained withhematoxylin (blue). Reciprocal intensity of the precipitated chromogen was measured in a.u., as described (46). Bar, 100 mm (a, b) or 50 mm (c, d). Forstatistical analyses, the two-tailed Student t test (A, C–F), or the two-tailed Mann–Whitney test (B) were used, considering � , P < 0.05;

��, P < 0.01;

���, P < 0.001; ns:

no significant difference; a.u. ¼ arbitrary units.

Dual Role of miR-146a in Tumor Progression

www.aacrjournals.org Mol Cancer Res; 14(6) June 2016 551

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 5: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

Raimo et al.

Mol Cancer Res; 14(6) June 2016 Molecular Cancer Research552

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 6: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

expressing high levels of miR-146a have an advantage in growth,miR-146a–depleted ones show increased migration/invasionand extravasation and are responsible for distant metastasisformation. Supporting our hypothesis is the fact that weobserved the formation of a significantly higher number ofcolonies when CTCs were derived from mice bearing miR-146a–dowmodulated tumors (pLentiGFP-sponge), comparedwith controls (pLentiGFP-EV), as shown in Fig. 2G.

miR-146a affects different tumor progression-related genes andpathways

Because miRNAs biologic functions are exerted throughnegative regulation of cognate target genes, we sought formiR-146a targets potentially involved in melanoma establish-ment and spreading by using TargetScan 6.1 (13) to predict itstargets. In this way, we identified 223 conserved predicted genescontaining one or more putative binding sites for miR-146a(Supplementary Table S2), later on used to identify miR-146a–altered pathways and functions by employing the IPA software(www.ingenuity.com). Notably, the most statistically signifi-cant Canonical Pathway that emerged was Notch Signaling,where 3 out of 38 genes in the pathway were putative miR-146atargets (NUMB, LFNG, and NOTCH2; P ¼ 6.77e�03; Supple-mentary Fig. S9A). NUMB is an adaptor protein that triggersNOTCH degradation (14), while lunatic fringe (LFNG) is abeta-1,3-N-acetylglucosaminyltransferase that leads to elonga-tion of O-linked fucose residues on NOTCH extracellulardomain, causing inhibition of signaling by the Serrate/JAGGEDclass of ligands (15). Instead, NOTCH2 is highly expressed inhuman melanomas and favors tumor formation and invasion(16), making its targeting by miR-146a unlikely. On the otherhand, searching for putative targets involved in tumor invasion,IPA identified 22 candidate genes (Supplementary Fig. S9B);among them, NUMB and integrin alpha-V (ITGAV) are keyplayers in tumor cell invasion (17). Interestingly, ROCK1 istargeted by miR-146a in prostate cancer cells, thereby inhibit-ing cell proliferation and invasion (18), suggesting that thisgene could be a fundamental driver of melanoma cell spread-

ing. With all that in mind, we focused on the role of LFNG,NUMB, ITGAV, and ROCK1.

miR-146a stable (pLentiGFP-sponge) or transient (anti-146a) inhibition in MA-2 and 1300-Mel cells (efficient down-modulation was verified in Supplementary Fig. S4) led toincreased LFNG, ROCK1, ITGAV, and NUMB protein expressionin a range going fromþ15% toþ300% compared with controls;conversely, transient miR-146a overexpression (pre-146a)reduced protein levels of LFNG, ROCK1, ITGAV, and NUMBby about 15% to 75%, 48 hours after transfection comparedwith controls, as assessed by Western blot (WB) analysis in Fig.3A; Supplementary Fig. S10A. mRNA levels of these genes werealso decreased by about 50% to 70% compared with controls, asassessed by qRT-PCR, 48 hours after transient miR-146a over-expression in MA-2 cells (pre-146a compared with pre-Cntrl, Fig. 3B), while miR-146a downmodulation in MA-2 cellsled to opposite effects (Supplementary Fig. S10B). Notably,analysis of 352 melanoma metastatic samples in The CancerGenome Atlas (TCGA) dataset (http://cancergenome.nih.gov/)showed a significant negative correlation between miR-146aand LFNG, ROCK1, ITGAV, and NUMB expression (Supple-mentary Fig. S11), suggesting the relevance of miR-146a and itstargets in melanoma dissemination. To assess if LFNG, ITGAV,ROCK1, and NUMB fluctuations in miR-146a–modulated cellsdirectly depend on the binding between miR-146a seed and thecomplementary sequence present on the 30-UTRs of these genes,their 30-UTR sequences were cloned into reporter vectorsand luciferase assays performed in MA-2 cells transfected withmiR-146a precursors or negative controls (pre-146a or pre-Cntrl). As shown in Fig. 3C, miR-146a significantly impairedluciferase expression in the presence of LFNG, ITGAV, ROCK1,and NUMB 30-UTRs. Being completely novel target genes,miR-146a specificity on repression of LFNG and ITGAV wasfurther validated by inserting three point mutations into their30-UTR miR-146a complementary sequences (LFNG-mut,ITGAV-mut). Binding site mutations completely abrogatedmiR-146a–dependent inhibition of luciferase expression.Strong evidence of the relevance of LFNG, ROCK1, and NUMB

Figure 2.miR-146a inhibits cell extravasation and metastasis formation in vivo and is poorly expressed in CTCs. A and B, Transwell migration or Matrigel invasion wasassayed upon stable downmodulation ofmiR-146a inMA-2 cells, comparedwith controls (pLentiGFP-sponge or -EV), or 24 hours after transfection of 1300-Mel cellswith miR-146a precursor or its negative control (pre-146a vs. pre-Cntrl). Results are shown as mean � SEM of the area covered by migrated or invaded cells.Three independent experiments were performed in triplicates and representative results are shown. C, transendothelial migration was mimicked in vitro byanalyzing MA-2 CMRA-labeled (red) cells migration through a fibronectin-coated Transwell membrane covered by a confluent monolayer of HUVEC-GFP. Top,HUVEC-GFP monolayer, upper side of the Transwell at time 0 hour (a) or 18 hours after seeding MA-2 cells transfected with pre-146a or negative control(pre-146a vs. pre-Cntrl; b, c). Bottom, transmigratedMA-2 (d, e) cells, transfected as indicated, lower side of the Transwell. Bar, 30mm. Two independent experimentswere performed in triplicates and representative results are shown. D, in vivo extravasation of miR-146a–overexpressing cells previously labeled with CMRA(red; pre-146a), compared with controls (pre-Cntrl) was evaluated 2 hours (c, d) or 48 hours (e, f) following tail-vein injections in CD1 nude mice. Representativepictures of the whole lungs are shown; bar, 500 mm. Results are presented as mean � SEM of the number of extravasated cells at 48 hours for 5 mice pergroup, evaluated aswhole lungs. Representative fields (a, b) ofmurine lung sections at 2 hours (a) or 48 hours (b) following tail-vein injections of CMRA-labeled (red)melanoma cells in CD1 nude mice, stained for CD31 to highlight blood vessels (green) and counterstained with DAPI (blue). Arrowheads indicate humanmelanoma red or orange (when overlapping to green vessels) cells within or outside the vessels. Bar, 10 mm. Two independent experiments were performed, andrepresentative results are shown. E, metastasis formation was evaluated in the lungs of CB.17 scid mice 5 weeks after tail-vein injections of MA-2 cells stablytransducedwithmiR-146a overexpression lentiviral vectors or their controls (pWPT-146a vs. -EV, a and b), or subcutaneous injections of miR-146a–downmodulated(pLentiGFP-sponge vs. -EV, c and d) or overexpressing (pLemiR-146a vs. -EV, e and f) MA-2 cells. Representative images of the lungs are shown. Thenumber of lung colonies is shown as mean � SEM for 5 mice per group. Bar, 1 mm. One (a–b) or two (c–d, e–f) independent experiments were performed andrepresentative results are shown. F, miR-146a expression was evaluated by qRT-PCR in primary tumors, CTCs, and lung colonies generated 5 weeks uponsubcutaneous injection of MA-2 cells into NSGmice. Results are shown as fold changes (mean� SEM) relative toMA-2–cultured cells, normalized on U44RNA levels.n ¼ 5/6/8 mice per group were evaluated. G, CTCs in the blood of mice injected subcutaneously with miR-146a downmodulated MA-2 cells versus controls(pLentiGFP-sponge vs. -EV) were evaluated by counting the number of colonies obtained from blood-derived cells in culture for 7 days. Cells were stained withcrystal violet; n ¼ 4 or 5 mice per group were evaluated. For statistical analyses, the two-tailed Student t test (A–E, G) or the two-tailed Mann–Whitney test (F)were used, considering � , P < 0.05; �� , P < 0.01; ��� , P < 0.001; ns: no significant difference. SQ, subcutaneous.

Dual Role of miR-146a in Tumor Progression

www.aacrjournals.org Mol Cancer Res; 14(6) June 2016 553

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 7: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

in melanoma progression came from qRT-PCR analysis of theirexpression in patient-derived samples: all the genes showed lessexpression in invasive human melanomas and in cutaneous

metastases than in in situ tumors (Fig. 3D). Instead, ITGAV wasnot found statistically significant here (Fig. 3D), maybe becausenot enough samples were analyzed.

Figure 3.miR-146a affects different tumorprogression–related genes andpathways. A, WB analysis of LFNG,ROCK1, ITGAV, and NUMB protein levelswas performed in MA-2 cells stablytransduced with miR-146a-downmodulation or control lentiviralvectors (pLentiGFP-sponge vs. -EV) or48 hours after miR-146a transientoverexpression (pre-146a vs. pre-Cntrl).Protein modulations were calculatedrelative to the indicated controls,normalized on the hsp90 or alpha-tubulin loading control and expressed aspercentages. B, LFNG, ROCK1, ITGAV,and NUMB mRNA levels were measuredby qRT-PCR in MA-2 cells 48 hours aftertransfectionwithmiR-146a precursors orcontrols (pre-146a or pre-Cntrl). Resultsare shown as fold changes (mean �SEM) relative to controls, normalized toGAPDHmRNA levels. Three experimentswith independent protein and RNApreparations were performed for A andB, and representative ones are shown. C,luciferase assays were performed inMA-2 cells cotransfected with miR-146aprecursors or negative controls (pre-146a or pre-Cntrl) and reporterconstructs containing the wild-type30-UTR of LFNG or ITGAV or ROCK1 orNUMB, including miR-146a bindingsequences at positions 623, 3168, 1144and 1512, respectively, or themutated 30-UTR of LFNG or of ITGAV containingthree point mutations in the miR-146abinding sites. Alternatively, a syntheticsequence composed of three perfectmiR-146a binding sequences (Sensor)was used as a positive control. Resultsare shown as mean � SEM of Fireflyluciferase activity relative to controls,normalized on Renilla luciferase activity.Three independent experiments wereperformed, and representative ones areshown. D, LFNG, ROCK1, ITGAV, andNUMB expression was evaluated byqRT-PCR in human in situ melanomas(n ¼ 22, 8, or 12), invasive melanomas(n ¼ 36, 7, 22, or 20) and cutaneousmetastases (n ¼ 23, 10, 7, or 18,respectively). Results are shown as box-and-whisker plots of fold changes (mean� SEM) relative to in situ melanomas,normalized on U44 RNA levels. Forstatistical analyses, the two-tailedStudent t test (B–C), or the two-tailedMann–Whitney test (D) were used,considering � , P < 0.05; �� , P < 0.01;��� , P < 0.001; ns: no significantdifference.

Raimo et al.

Mol Cancer Res; 14(6) June 2016 Molecular Cancer Research554

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 8: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

miR-146a controls cell and tumor growth, but not cellmovement, through LFNG and NUMB

To characterize LFNG and NUMB involvement in miR-146a–driven phenotypes, MA-2, 1300-Mel and WK-Mel cells weretransiently transfected either with a control or a LFNG or/and

NUMB specific siRNAs (siRNA-Cntrl, siRNA-LFNG, or siRNA-NUMB or siRNA-LFNG þ siRNA-NUMB), leading to efficientsilencing of LFNG and NUMB expression, as verified by qRT-PCRand WB analysis (Supplementary Fig. S12A–S1C and S13A).Downmodulation of NUMB and mainly of LFNG increased

Figure 4.miR-146a controls cell and tumor growth,but not cell movement, through LFNG andNUMB.A, invitroproliferationofMA-2cellswas analyzed starting at 12 hours afterLFNG, NUMB, or LFNG plus NUMBtransient downmodulation, comparedwith controls (siRNA-LFNG, siRNA-NUMB,or siRNA-LFNGþ siRNA-NUMBvs. siRNA-Cntrl). Results are shown as mean � SEMof the optical density (OD); threeindependent experiments wereperformed in sestuplicates, and results areshown as representative ones. B,anchorage-independent growth of MA-2cells was measured 21 days aftertransfection with LFNG or control siRNAs(siRNA-LFNG, panel b, vs. siRNA-Cntrl, a).Results are shown as mean � SEM of thearea covered by colonies. Threeindependent experiments wereperformed in triplicate and representativeresults are shown. Bar, 4 mm. C, primarytumor growthwas analyzed 4 weeks afterinjection in the flank of NSG mice of MA-2cells stably downmodulated for LFNGand/or NUMB, compared with controls(pLKO-shLFNG and pLKO-shNUMB vs.pLKO-EV; pLKO-shLFNG þ shNUMB vs.pLKO-EVþEV); tumor weight wasevaluated in n ¼ 3 or 5 mice per group.Two independent experiments wereperformed and a representative one isshown. D, Transwell migration wasevaluated 24 hours upon transient LFNGplus NUMBdownmodulation inMA-2 cells,compared with controls (siRNA-LFNG þsiRNA-NUMB vs. siRNA-Cntrl). Results areshown asmean� SEMof the area coveredby migrated cells. Three independentexperiments were performed in triplicateand representative results are shown. E,Transendothelial migration was mimickedin vitroby analyzing themigrationofMA-2CMRA-labeled (red) cells through afibronectin-coated Transwell membranecovered by a confluent monolayer ofHUVECs-GFP. Top, HUVECs-GFPmonolayer, upper side of the Transwell attime 0 hour (a) or 18 hours followingseeding of MA-2 cells (b, c) cotransfectedwith LFNG plus NUMB siRNAs or theirnegative controls (siRNA-LFNGþNUMBvs. siRNA-Cntrl). Bottom, transmigratedMA-2 (d, e) cells, transfected as indicated,lower side of the Transwell. Bar, 30 mm.Three independent experiments wereperformed in triplicate, and representativeresults are shown.The two-tailedStudent ttest was used for statistical analysesconsidering � , P < 0.05; ��, P < 0.01;��� , P < 0.001; ns: no significant difference.

Dual Role of miR-146a in Tumor Progression

www.aacrjournals.org Mol Cancer Res; 14(6) June 2016 555

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 9: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

Raimo et al.

Mol Cancer Res; 14(6) June 2016 Molecular Cancer Research556

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 10: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

in vitro cell proliferation up to 4 days in culture, if compared withcontrols, resembling miR-146a overexpression (Fig. 4A; Supple-mentary Fig. S14A, black triangles or black squares vs. blackcircles). Concomitant downmodulation of NUMB increased cellgrowth at ahigher extent, suggesting anadditive role for LFNGandNUMB in the regulation of cell proliferation (Fig. 4A; Supple-mentary Fig. S14A, white squares and dashed line). Furthermore,as depicted in Fig. 4B, LFNG transient downmodulation (siRNA-LFNG, b) in MA-2 cells increased colony formation in soft-agarassays, compared with controls (siRNA-Cntrl, a). Most relevantly,LFNG plus NUMB involvement in in vivo tumor growth wasassessed upon subcutaneous injections in the flank of immuno-compromised mice of MA-2 cells stably downmodulated forLFNG þ NUMB (pLKO-shLFNG þ shNUMB) or LFNG only(pLKO-shLFNG) orNUMBonly (pLKO-shNUMB) and comparedwith controls (pLKO-EVþEVor pLKO-EV), as verifiedbyqRT-PCRand WB in Supplementary Fig. S12D and S13B: 4 weeks afterinjections, LFNG and NUMB-downmodulated cells, in combina-tion or as single modulations, generated significantly biggertumors compared with all controls (Fig. 4C), phenocopyingmiR-146a overexpression. Notably, even if the effect of LFNGsilencing wasmore pronounced compared with NUMB silencing,an additive effect in tumor growth was not observed for LFNG þNUMB-downmodulated cells, suggesting that, in vivo, LFNG actsas the main player.

Because NUMB was depicted by IPA and by the literature as acell invasion–related molecule, we investigated if miR-146a-dependent targeting of NUMB plus LFNG could inhibit melano-ma progression. However, in vitro Transwell and transendothelialmigration of MA-2 cells through a HUVEC monolayer wascompletely unaffected by LFNG þ NUMB co-downmodulation(siRNA-LFNGþ siRNA-NUMB), compared with controls (siRNA-Cntrl), as shown in Fig. 4D and E. Furthermore, NUMB does notaffect in vivo cell extravasation, as evaluated 48hours after tail-veininjection into immunocompromised mice of NUMB-downmo-dulated cells (siRNA-NUMB, compared with siRNA-Cntrl), orlung metastasis formation, 6 weeks after tail-vein injection intomice of NUMB-overexpressing MA-2 cells (pLenti-NUMB) com-

pared with controls (pLenti-EV; Supplementary Fig. S14B and C).NUMB cDNA efficient overexpression was verified in Supplemen-tary Fig. S13D. Collectively, we evidenced that two NOTCHpathway genes, LFNGandNUMB, are pivotal inmiR-146a–drivenenhancement of cell growth and primary tumor formation, con-sidering LFNG as the main player. However, these two genes areirrelevant for cell invasion and metastatization.

miR-146a–dependent activation of NOTCH1 via LFNG þNUMB induces cell growth through the Akt/PTEN pathway

Because LFNGþNUMBmodulation in melanoma cells affectstumor growth, we investigated whether miR-146a can activateNOTCH signaling in melanoma cells through the double actionof these two players. Reporter assays performed using aNOTCH1-dependent promoter located upstream of a luciferase cassetteconfirmed that miR-146a overexpression (pre-146a) in MA-2cells enhances luciferase expression of about 60%, comparedwith control (pre-Cntrl), as shown in Fig. 5A, light gray bars.LFNG or NUMB dowmodulation (siRNA-LFNG or siRNA-NUMB) increased luciferase expression to the same extent, com-pared with control (siRNA-Cntrl). Interestingly, LFNG plusNUMB codownmodulation (siRNA-LFNG þ siRNA-NUMB)strongly induced NOTCH activity, to a level comparable withmiR-146a alone, revealing again an additive effect of the doublegene targeting (Fig. 5A, dark gray bars). NOTCH1 constitutivelyactive cleaved form (pCNA-hNIC, overexpression verified inSupplementary Fig. S13E) was used as a positive control (pat-terned bars). Furthermore, NOTCH1 transcriptional targets hairyenhancer of Split (HES)-1 and -5, and Cyclin D1 (CCND1) werefound strongly induced inMA-2 cells uponmiR-146a overexpres-sion (pre-146a vs. pre-Cntrl), or significantly reduced by miR-146a inhibition (anti-146a vs. anti-Cntrl); accordingly, theirexpression also increased upon LFNG or NUMB stable dowmo-dulation (pLKO-shLFNG or pLKO-shNUMB, compared withpLKO-EV), but the effect of LFNG inhibition was stronger, asassessed by qRT-PCR (Fig. 5B; Supplementary Fig. S15A).NOTCH1 and Cyclin D1 protein modulations were assessed byWB in Supplementary Fig. S15B. Based on this evidence, we asked

Figure 5.miR-146a–dependent activation of NOTCH1 via LFNGþNUMB induces cell growth through the Akt/PTEN pathway. A, luciferase assayswere performed inMA-2 cellscotransfected with a vector containing a NOTCH1-dependent promoter cloned upstream of the luciferase coding sequence, together with miR-146aprecursors or negative controls (pre-146 or pre-Cntrl), LFNG, and/or NUMB siRNAs or controls (siRNA-LFNG, siRNA-NUMB, siRNA-LFNG þ siRNA-NUMB, andsiRNA-Cntrl), or Notch intracellular domain–overexpressing or empty vectors (pCNA-hNIC or -EV). Results are shown as mean � SEM of Firefly luciferaseactivity relative to controls, normalized on Renilla luciferase activity. Three independent experiments were performed and representative results are shown. B, HES1and Cyclin D1 (CCND1) mRNA levels were measured by qRT-PCR in MA-2 cells 48 hours after transfection with miR-146a precursors or inhibitors or theirnegative controls (pre- and anti-146a or control), or in MA-2 cells stably transduced with LFNG or NUMB shRNAs, compared with empty vectors (pLKO-shLFNGand pLKO-shNUMB vs. pLKO-EV). Results are shown as fold changes (mean � SEM) relative to controls, normalized on GAPDH mRNA levels. Three experimentswith independent RNA preparations were performed and representative results are shown. C, in vitro proliferation assays were performed in MA-2 cellsstarting from 12 hours after transfection with NOTCH1 or HES1 siRNAs, compared with controls (siRNA-NOTCH1, siRNA-HES1 vs. siRNA-Cntrl). Results are shown asmean � SEM of the optical density (OD); three independent experiments were performed in sestuplicate and representative results are shown. D, Aktphosphorylation status at Thr308 or Ser473 was assessed in MA-2, WK-Mel, and 1300-Mel cells 48 hours after transfection with miR-146a precursors (pre-146aor pre-Cntrl), or in MA-2 cells stably transduced with miR-146a–overexpressing or control lentivirus vectors (pLemir-146a or -EV). Phosphorylated proteinmodulations were calculated relative to controls, normalized on the total amount of proteins and on hsp90 or alpha-tubulin loading controls and expressed aspercentages. E and F, PTEN protein and mRNA levels were assessed by WB (E) and qRT-PCR (F) in MA-2 and 1300-Mel cells 48 hours after transfection withmiR-146a precursors or inhibitors or their negative controls (pre- and anti-146a vs. control), upon stablemiR-146a overexpression or empty controls (pLemir-146a vs.-EV) or 48 hours after LFNG or NUMB transient downmodulation (siRNA-LFNG or siRNA-NUMB vs. siRNA-Cntrl). Protein modulations in E were calculatedrelative to controls, normalized on hsp90 or alpha-tubulin loading controls and expressed as percentages; results in F are shown as fold changes (mean � SEM)relative to controls, normalized on GAPDH mRNA levels. Three experiments with independent protein and RNA preparations were performed andrepresentative ones are shown. G, in vitro proliferation was assessed in MA-2 cells from 12 hours after transfection withmiR-146a precursors or controls (pre-146a vs.pre-Cntrl), combined with a daily treatment with the Calbiochem Akt inhibitor VIII 612847-09-3 (Akti) or controls (dimethyl sulfoxide, DMSO). Results areshown asmean� SEMof the optical density (OD); three independent experimentswere performed in triplicate, and representative results are shown. The two-tailedStudent t test was used for statistical analyses considering � , P < 0.05; �� , P < 0.01; ��� , P < 0.001; ns: no significant difference.

www.aacrjournals.org Mol Cancer Res; 14(6) June 2016 557

Dual Role of miR-146a in Tumor Progression

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 11: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

Figure 6.miR-146a–dependent downregulation of ITGAV and ROCK1 affects cell motility, but not cell growth. A, in vitro proliferation of ROCK1-downmodulated(siRNA-ROCK1) or control (siRNA-Cntrl) MA-2 cellswas evaluated starting from 12 hours after transfection. Results are shown asmean� SEMof the area occupied bycolonies, or of the optical density (OD); three independent experiments were performed, and representative results are shown. B, Transwell migration orMatrigel invasion was assayed 24 hours after transfection of MA-2 cells with ROCK1 siRNAs or their negative controls (siRNA-ROCK1 vs. siRNA-Cntrl). Results areshown as mean � SEM of the area covered by migrated or invaded cells. Three independent experiments were performed in triplicate, and representativeresults are shown. C, transendothelial migration was mimicked in vitro by analyzing the migration of CMRA-labeled (red) MA-2 cells through a fibronectin-coated Transwell membrane covered by a confluent monolayer of HUVEC-GFP. (Continued on the following page.)

Mol Cancer Res; 14(6) June 2016 Molecular Cancer Research558

Raimo et al.

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 12: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

whethermiR-146a–dependent activationofNOTCH1 is crucial inpromoting melanoma cell growth. In vitro proliferation wasevaluated for MA-2 cells transiently downmodulated forNOTCH1 or HES1 (siRNA-NOTCH1 and siRNA-HES1), and,interestingly, inhibition of these two genes reduced cell growth,if compared with controls (siRNA-Cntrl) up to 5 days in culture,opposing miR-146a–driven phenotype (Fig. 5C). EfficientNOTCH1 and HES1 downmodulation after siRNA transfectionwas evaluated by qRT-PCR (Supplementary Fig. S12E-F). In vitrotransendothelial migration of NOTCH1-downmodulated cells(siRNA-NOTCH1 vs. siRNA-Cntrl) was also performed, andresults confirmed that the alteration of this pathway is not relatedto cell migration (Supplementary Fig. S15C).

Given the important role of the NOTCH1/HES1 axis in miR-146a–dependent control of cell growth, we investigated themolecular pathways responsible for these phenotypes. Interest-ingly, Palomero and colleagues demonstrated that, in aNOTCH1-induced T-cell acute lymphoblastic leukemia (T-ALL) model,HES1 activation leads to reduced PTEN transcription and expres-sion, resulting in increased phosphatidylinositol-4,5-bispho-sphate 3-kinase (PI3K)/Akt pathway activation and cell prolifer-ation (19). In the same way, we found that miR-146a expressionin melanoma cells affects Akt phosphorylation, as shown by WBanalysis in Fig. 5D: 24hours after transfection,miR-146a transientoverexpression in MA-2 cells (pre-146a) increased Akt phosphor-ylation (50% and 30%, respectively) on Thr308 and Ser473,compared with controls (pre-Cntrl), while stable expression ofmiR-146a inMA-2 cells (pLemiR-146a) induced a65% increase ofThr308 phosphorylation, compared with controls (pLemiR-EV).The same effect was described upon miR-146a transient over-expression in WK-Mel and 1300-Mel cells. Consistently, miR-146a transient (pre-146a) or stable (pLemiR-146a) overexpres-sion inMA-2, 1300-Mel, andWK-Mel cells reduced PTEN proteinand mRNA levels (at least 20%), compared with controls (pre-Cntrl or pLemiR-EV, respectively), while miR-146a transientdownmodulation led to a 40% increase in protein expression,as assessed byWBandqRT-PCR (Fig. 5E and F and SupplementaryFig. S15D). Furthermore, LFNG and NUMB transient downmo-dulation inMA-2 cells (siRNA-LFNG or siRNA-NUMB, vs. siRNA-Cntrl) reduced PTENmRNA amount by about 50% (Fig. 5F) andeither HES1 or NOTCH1 transient downmodulation (siRNA-HES1 and siRNA-NOTCH1, vs. siRNA-Cntrl) increased PTENprotein expression (11% and 16%, respectively), compared withcontrols (siRNA-Cntrl), as shown by WB analysis in Supplemen-tary Fig. S15E. All the evidence indicates that miR-146a–depen-dent inhibition of LFNG and NUMB and the subsequent activa-tion of NOTCH1 and HES1 are responsible for PTEN down-modulation and Akt increased phosphorylation. To understandif this signaling is important for cell growth, miR-146a–over-expressing MA-2, 1300-Mel, and WK-Mel cells were treated withlow concentrations (0.5 mmol/L) of an Akt inhibitor (612847-09-3), in order to restore the Akt phosphorylation status of control

cells (efficacy was verified in Supplementary Fig. S13F), andanalyzed for in vitro proliferation. As shown in Fig. 5G andSupplementary Fig. S15F, the proliferative advantage of miR-146a–overexpressing MA-2, 1300-Mel, and WK-Mel cells (pre-146a þ Akti, black rhombi) was completely inhibited up to 5days in culture, compared with nontreated cells (pre-146a þDMSO, white triangles). Importantly, this effect specifically occursinmiR-146a–overexpressing cells, because proliferation of controlcells was poorly affected (pre-Cntrl þ Akti, white squares anddashed line, andpre-CntrlþDMSO,black triangles). This evidenceindicates thatmiR-146a–dependent increased growth is duemain-ly to Akt pathway activation, making the inhibition of the specificmiR-146a/Akt pathway potentially relevant for therapy.Moreover,in line with the above presented results, Akt inhibition (up to 5mmol/L) in miR-146a-overexpressing cells (pre-146a þ Akti) didnot affect either cell migration or invasion in vitro (SupplementaryFig. S15G), further demonstrating that miR-146a effect on cellmovement does not involve NOTCH1 pathway modulation.

miR-146a-dependent downregulation of ITGAV and ROCK1affects cell motility, but not cell growth

Because integrins are the main coordinators of tumor dissem-ination, and ITGAV as well as the integrin downstream player,ROCK1, are modulated by miR-146a (Fig. 3A and B; Supplemen-tary Fig. S10), we evaluated the role of ROCK1 and ITGAV in celladhesion/motility. We first downmodulated ROCK1 with a spe-cific siRNA (whose efficacy was evaluated in Supplementary Figs.S12G and S13C) and observed no effect on in vitro cell prolifer-ation, up to 4 days in culture (siRNA-ROCK, compared withsiRNA-Cntrl), as shown in Fig. 6A. However, we observed a25% to 50% reduction in migration and invasion through Matri-gel for MA-2, 1300-Mel, andWK-Mel cells, as evaluated by in vitroTranswell assays (siRNA-ROCK1 vs. siRNA-Cntrl, Fig. 6B; andSupplementary Fig. S16A). Moreover, a less pronounced remo-deling (b and c) and an 80% inhibition of transendothelialmigration (d and e) through a HUVEC monolayer was found(siRNA-ROCK1 vs. siRNA-Cntrl; Fig. 6C; Supplementary Fig.S16B). This in vitro evidence was further supported by an in vivoextravasation experiment, where ROCK1-downmodulated cells(siRNA-ROCK1) showed a significantly reduced ability to colo-nize the lung parenchyma, compared with controls (siRNA-Cntrl), 48 hours after tail-vein injections into immunocompro-misedmice (Fig. 6D, c–d). Equal lung lodgingwas verified 2hoursafter injection (a–b). Furthermore, we analyzed the role of ITGAVin the miR-146a cascade involved in tumor dissemination byperforming an in vitro adhesion assay on vitronectin, the glyco-protein specific for the avb3 integrin heterodimer binding. Rel-evantly, we found that miR-146a–overexpressing MA-2, 1300-Mel, and WK-Mel cells (pre-146a) showed significant reduction(�30%) of vitronectin adhesion, compared with controls (pre-Cntrl), 1 hour after plating (Fig. 6E; Supplementary Fig. S16C),suggesting the importance of ITGAV downmodulation in

(Continued.) Top, HUVEC-GFP monolayer, upper side of the Transwell at time 0 hour (a) or 18 hours following seeding of MA-2 cells (b, c) transfected with ROCK1siRNAsor negative controls (siRNA-ROCK1 vs. siRNA-Cntrl). Bottom, transmigratedMA-2 (d, e) cells, transfected as indicated, lower side of the Transwell. Bar, 30mm.Three independent experiments were performed in triplicate, and representative results are shown. D, in vivo extravasation of ROCK1-downmodulated cellspreviously labeled with CMRA (red; pre-146a), compared with controls (pre-Cntrl) was evaluated 2 hours (a, b) or 48 hours (c, d) following tail-vein injectioninNSGmice. Representative pictures of the lungs are shown (a–d); bar, 500mm.Results are presented asmean� SEMof the number of extravasated cells at 48 hoursfor 5 mice per group. One experiment was performed. E, MA-2 cells transiently transfected with miR-146a (pre-146a) or controls (pre-Cntrl) were plated on avitronectin layer and adhesion was assessed 1 hour later as mean� SEM of the OD. Three independent experiments were performed and representative results areshown. The two-tailed Student t test was used for statistical analyses considering � , P < 0.05; �� , P < 0.01; ��� , P < 0.001; ns: no significant difference.

Dual Role of miR-146a in Tumor Progression

www.aacrjournals.org Mol Cancer Res; 14(6) June 2016 559

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 13: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

miR-146a–driven impairment of cell invasion and metastatiza-tion. Taken together, these experiments suggest that the miR-146a–dependent ITGAV and ROCK1 modulations are relevantevents for melanoma dissemination.

In summary, we showed thatmiR-146a exerts a complex role inmelanoma: its expression varies through the different steps ofprogression, where it promotes growth but it impairs cell dissem-ination by targeting different and unrelated genes and pathways.Disclosing this dualism is crucial to identify new, specific, mel-anoma therapeutic treatments (Fig. 7).

DiscussionIn this work, we present a new mechanism by which a small

noncoding RNA, miR-146a, regulates melanoma establishmentand dissemination by two parallel and opposite interventions.We proved that while high levels of miR-146a have been foundin malignant melanomas and are able to promote tumorgrowth, at the same time, miR-146a impairs tumor cell dissem-ination. miR-146a opposite activities are obtained by the target-ing of different genes. miR-146a direct targeting of LFNG andNUMB leads to NOTCH1 signaling activation, which coincideswith increased levels of HES1, HES5, Cyclin D1 and p-Akt,decreased PTEN expression and tumor growth gain. Instead,silencing of ROCK1 and ITGAV by miR-146a favors tumor celldissemination and distant metastasis formation. The dual andcontradictory functions of miR-146a are acquired by miR-146aexpression fluctuations during melanoma progression. In fact,while we found that the tumor mass, as a whole, is enriched inmiR-146a, the evaluation of its amount in CTCs showed poormiR-146a expression. These findings suggest the presence of aheterogeneous primary mass which mostly contains cells withhigh miR-146a levels that have an advantage in proliferation.However, most probably, cells that poorly express miR-146acoexist, and these are the cells that can easily escape anddisseminate. This hypothesis is also supported by the fact thatthe number of CTCs is higher in mice carrying xenotransplants

with low miR-146a levels compared with controls. In summary,here we propose a dual function for miR-146a.

miR-146a is a well-known regulator of many biologic process-es, including cancer. miR-146a locus is characterized by a C/Gsingle nucleotide polymorphism (SNP; rs2910164) located in thestem region of the precursor passenger strand. TheG allele is oftenassociated with papillary thyroid carcinomas (20), cervical andprostate cancers as well asmelanomas (21–23). Conversely, the Callele is more often associated with breast, ovarian, and gastrictumors (24–26). Apart from this polymorphism, miR-146a isgenerally overexpressed in cervical (27), gastric (25), pancreaticand breast (28) cancers as well as in oral squamous cell carcino-mas (29). However, it has been found poorly expressed in someprostate, pancreatic, and gastric cancers (18, 28, 30) and itsenforced expression in breast cancer cells inhibits their metastaticpotential (31).

Based on the above-listed findings, considering the relevanceof miR-146a deregulation in tumor progression, we investigatedthe intervention of miR-146a in melanoma, an unfortunatelystill fatal neoplasia. We found that miR-146a is highly expressedin invasive human melanomas as well as in cutaneous metas-tases, in line with other studies (32), and that its expressioncorrelates with tumor thickness and recurrence. Using humanmelanoma cells, we evidenced the protumorigenic role of miR-146a, leading to increased cell proliferation, anchorage-inde-pendent growth and primary tumor formation in mice. Inter-estingly, cell proliferation is favored also in distant-site metas-tases. Concomitantly, we found that miR-146a high expressionimpairs migration and invasion as well as metastatic dissemi-nation of melanoma cells and that it affects tumor cells ability tocross the vessels. In line, as anticipated above, we evidenced lowmiR-146a expression in CTCs, compared with tumor or meta-static masses. These results, together with the evidence that miR-146a does not affect melanoma cell survival and anoikis, let us tohypothesize that miR-146 levels need to be low when melano-ma cells detach from the primary tumor mass and disseminate,in order to favor cell spreading. miR-146a expression is thensomehow restored in distant-site colonies to favor metastaticgrowth. Putting all these data together, we believe that cellpopulations with different miR-146a expression are needed toobtain melanoma dissemination.

Taking advantage of bioinformatics predictions and functionalanalyses, we identified different relevant candidate target genesthat could account formiR-146a–driven phenotypes. Looking fortumor growth-related pathways, we observed that the NOTCHpathway was significantly enriched, including three putativetargets genes, LFNG, NUMB, and NOTCH2. miR-146a–depen-dent inhibition of NUMB and subsequent activation of theNOTCH pathway has been characterized before (23). LFNG isaO-fucose-b1,3-N-acetylglucosaminyltransferase able to glycosy-late epidermal growth factor (EGF)-like repeats on the extracel-lular domain of NOTCH, thereby enhancing DELTA-mediatedand inhibiting JAGGED-mediated NOTCH activation (33, 34).JAGGED1-dependent NOTCH activation is crucial for colon andprostate cancer cells proliferation and migration (35, 36). Zhangand colleagues demonstrated that loss of LFNG alters NOTCHsignaling in the prostate, leading to intraepithelial neoplasia (37)and Xu and colleagues described that the mammary-specificdeletion of Lfng induces basal-like breast cancer facilitating JAG-GED/NOTCH signaling (38). Relevantly, we observed that LFNGand NUMB expression is significantly lower in human invasive

Figure 7.Summary of the results presented in this work.

Raimo et al.

Mol Cancer Res; 14(6) June 2016 Molecular Cancer Research560

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 14: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

melanomas and in cutaneous metastases, compared with in situ,showing an opposite expression than miR-146a. Accordingly,LFNG andNUMB targeting is crucial to promote cell proliferationand primary tumor growth, leading to increased NOTCH1 andHES1 expression with a strong impact onmelanoma cells growth,as well as Cyclin D1 induction, which may play a role as well,considering that it drives melanoma formation (39). miR-146a–dependent activation of NOTCH1/HES1 leads to reduced PTENexpression and increased Akt phosphorylation, which drives cellgrowth. Activation of this pathway may also affect other featuresof tumor cells, like glucose intake, cell survival and protectionfrom apoptosis (40), making the spectrum of miR-146a–depen-dent effects even broader.

If cell growth is driven through the activation of the NOTCHpathway, we demonstrated that cell movement and metastasisformation are independently regulated by miR-146a. NeitherLFNG nor NUMB did affect melanoma cell migration and inva-sion in vitro, while, interestingly, two key players of the integrinsignaling, ITGAV and ROCK1, are affected by miR-146a. ROCK1mediates "amoeboid," rounded cell migration, while ITGAV isinvolved in "mesenchymal-like" invasion, characterized by cellpolarization and membrane protruding, two invasion strategiesused by melanoma cells during dissemination (41). ROCK1 ispoorly expressed in invasive human melanomas and cutaneousmetastases, whereas miR-146a is enriched. ROCK1 downmodu-lation inhibits in vitro invasion and in vivo extravasation (48hours) of melanoma cells, resembling miR-146a–dependenteffect, making it a crucial gene driving cell movement. RegardingITGAV/miR-146a function, forced miR-146a expression in mel-anoma cells impairs their ability to adhere on vitronectin, themajor integrin avb3 receptor (42). Taken together, our resultsindicate that miR-146a affects melanoma cell migration in mul-tiple ways, impairing the two major strategies for invasiveness.Considering that miR146a is poorly expressed in invading mel-anoma cells in the blood circulation, it is likely that subsequentactivation of the invasion molecules ITGAV and ROCK1 maydrive efficient cell spreading.

In conclusion, we identified and characterized the multiple,antithetic roles of miR-146a in melanoma progression; in fact,while miR-146a favors tumor growth, it impairs cell metastatiza-tion and we propose that melanoma cells overcome this dualismby strongly expressing miR-146a in tumors and metastases andreducing it during invasion, resulting in ultimate efficient growthand spreading. Considering that we and others described thatB-RAF V600E-bearing (but not B-RAF wild-type) aggressive

melanoma cell lines show high miR-146a expression, and thatmiR-146a is activated in melanoma cell lines via B-RAF–depen-dent activation of MYC (23), it is possible that this pathwaymediates miR-146a expression regulation in human tumors.Other regulators have also been described, such as NF-kB (43),V-Ets oncogene homolog 1 (ETS1; ref. 44), as well as epigenetics(45) and feedback (43) mechanisms. Autoregulation could alsooccur during melanoma progression; this would make the sce-nario even more complex if one considers miR-146a for thera-peutic interventions in melanoma.

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

Authors' ContributionsConception and design: M. Raimo, F. Orso, E. Calautti, D. TavernaDevelopment of methodology: M. Raimo, E. Penna, M.B. Stadler, L. PrimoAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): F. Orso, D. Cimino, L. Primo, P. QuaglinoAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): M. Raimo, E. Grassi, C. De Pitt�a, M.B. Stadler,E. Calautti, P. ProveroWriting, review, and/or revision of the manuscript: M. Raimo, F. Orso,M.B. Stadler, P. Quaglino, D. TavernaAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): D. Cimino, E. PennaStudy supervision: D. Taverna

AcknowledgmentsThe authors are grateful to Lei Xu and Richard Hynes for the A375P cells and

the metastatic variants, to Paola Circosta and Alessandro Cignetti for providingseveral melanoma cell lines, to Paolo Dotto and PP Di Fiore for NOTCH andNUMB vectors, to Alessandra Solero, Rosalia Curto and Angela Rita Elia forhelpingwith the experiments set up, to Carlo Tomasini for pathological analysisof human melanoma specimens, and to Flavio Cristofani for his help with theimmunocompromised mice.

Grant SupportThis work was supported by grants from Compagnia di San Paolo, Torino,

2008.1054 (D. Taverna), AIRC IG2010-10104DT, IG2013-14201DT, Fonda-zione CRT-2014.1085 (D. Taverna), Telethon TCP 06001 (E. Calautti), andFIRB giovani 2008 (RBFR08F2FS-002, F. Orso). E. Penna was a FIRC fellow(2012–2014).

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 October 20, 2015; revised February 8, 2016; accepted February 26,2016; published OnlineFirst March 10, 2016.

References1. Mueller DW, Bosserhoff AK. Role of miRNAs in the progression of malig-

nant melanoma. Br J Cancer 2009;101:551–6.2. Swick JM, Maize Sr JC. Molecular biology of melanoma. J Am Acad

Dermatol 2012;67:1049–54.3. Bar-Eli M.Role of AP-2 in tumor growth and metastasis of human mela-

noma. Cancer Metastasis Rev 1999;18:377–85.4. Aftab MN, Dinger ME, Perera RJ. The role of microRNAs and long non-

coding RNAs in the pathology, diagnosis, and management of melanoma.Arch Biochem Biophys 2014;563:60–70.

5. Luo C, Weber CEM, Osen W, Bosserhoff A-K, Eichm€uller SB. The role ofmicroRNAs in melanoma. Eur J Cell Biol 2014;93:11–22.

6. Bartel DP. MicroRNAs: Target recognition and regulatory functions. Cell2009;136:215–33.

7. Bennett PE, Bemis L, Norris DA, Shellman YG. miR in melanoma devel-opment: miRNAs and acquired hallmarks of cancer in melanoma. PhysiolGenomics 2013;45:1049–59.

8. Penna E, Orso F, Cimino D, Tenaglia E, Lembo A, Quaglino E, et al.microRNA-214 contributes to melanoma tumour progression throughsuppression of TFAP2C. EMBO J 2011;30:1990–2007.

9. Penna E, Orso F, Cimino D, Vercellino I, Grassi E, Quaglino E, et al.miR-214 coordinates melanoma progression by upregulating ALCAMthrough TFAP2 and miR-148b downmodulation. Cancer Res 2013;73:4098–111.

10. Xu L, Shen SS, Hoshida Y, Subramanian A, Ross K, Brunet J-P, et al. Geneexpression changes in an animal melanoma model correlate with aggres-siveness of human melanoma metastases. Mol Cancer Res 2008;6:760–9.

Dual Role of miR-146a in Tumor Progression

www.aacrjournals.org Mol Cancer Res; 14(6) June 2016 561

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 15: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

11. Balch CM, Gershenwald JE, Soong S-j, Thompson JF, Atkins MB, Byrd DR,et al. Final version of 2009 AJCC melanoma staging and classification.J Clin Oncol 2009;27:6199–206.

12. Hanniford D, Segura MF, Zhong J, Philips E, Jirau-Serrano X, Darvishian F,et al. Identification of metastasis-suppressive microRNAs in primary mel-anoma. J Natl Cancer Inst 2015;107: dju494, doi:10.1093/jnci/dju494.

13. Friedman RC, Farh KK-H, Burge CB, Bartel DP. Most mammalian mRNAsare conserved targets of microRNAs. Genome Res 2009;19:92–105.

14. Pece S, Confalonieri S, R. Romano P, Di Fiore PP. NUMB-ing down cancerby more than just a NOTCH. Biochim Biophys Acta 2011;1815:26–43.

15. Haltiwanger RS, Stanley P. Modulation of receptor signaling by glycosyl-ation: fringe is an O-fucose-b1,3-N-acetylglucosaminyltransferase. Bio-chim Biophys Acta 2002;1573:328–35.

16. Asnaghi L, Ebrahimi KB, Schreck KC, Bar EE, Coonfield ML, Bell WR, et al.Notch signaling promotes growth and invasion in uveal melanoma. ClinCancer Res 2012;18:654–65.

17. Seftor RE, Seftor EA, Gehlsen KR, Stetler-Stevenson WG, Brown PD,Ruoslahti E, et al. Role of the alpha v beta 3 integrin in human melanomacell invasion. Proc Natl Acad Sci U S A 1992;89:1557–61.

18. Lin S-L, Chiang A, Chang D, Ying S-Y. Loss of mir-146a function inhormone-refractory prostate cancer. RNA 2008;14:417–24.

19. Palomero T, Dominguez M, Ferrando AA. The role of the PTEN/AKTPathway in NOTCH1-induced leukemia. Cell cycle (Georgetown, Tex)2008;7:965–70.

20. Jazdzewski K, Murray EL, Franssila K, Jarzab B, Schoenberg DR, de laChapelle A. Common SNP in pre-miR-146a decreases mature miR expres-sion and predisposes to papillary thyroid carcinoma. Proc Natl Acad Sci2008;105:7269–74.

21. Xu B, Feng N-H, Li P-C, Tao J, Wu D, Zhang Z-D, et al. A functionalpolymorphism inPre-miR-146a gene is associatedwith prostate cancer riskand mature miR-146a expression in vivo. Prostate 2010;70:467–72.

22. Yue C,WangM,Ding B,WangW, Fu S, ZhouD, et al. Polymorphism of thepre-miR-146a is associated with risk of cervical cancer in a Chinesepopulation. Gynecol Oncol 2011;122:33–7.

23. Forloni M, Dogra SK, Dong Y, Conte D, Ou J, Zhu LJ, et al. miR-146apromotes the initiation and progression of melanoma by activating Notchsignaling. eLife 2014;3.

24. Lian H,Wang L, Zhang J. Increased risk of breast cancer associated with CCGenotype of Has-miR-146a Rs2910164 polymorphism in Europeans.PLoS ONE 2012;7:e31615.

25. Kogo R, Mimori K, Tanaka F, Komune S, Mori M. Clinical significance ofmiR-146a in gastric cancer cases. Clin Cancer Res 2011;17:4277–84.

26. PastrelloC, Polesel J, Della Puppa L, Viel A,Maestro R. Association betweenhsa-mir-146a genotype and tumor age-of-onset in BRCA1/BRCA2-negativefamilial breast and ovarian cancer patients. Carcinogenesis 2010;31:2124–6.

27. Wang X, Tang S, Le S-Y, Lu R, Rader JS, Meyers C, et al. Aberrant expressionof oncogenic and tumor-suppressive MicroRNAs in cervical cancer isrequired for cancer cell growth. PLoS ONE 2008;3:e2557.

28. Volinia S, Calin GA, Liu C-G, Ambs S, Cimmino A, Petrocca F, et al. AmicroRNAexpression signature of human solid tumors defines cancer genetargets. Proc Natl Acad Sci U S A 2006;103:2257–61.

29. Hung P-S, Liu C-J, Chou C-S, Kao S-Y, Yang C-C, Chang K-W, et al.miR-146a enhances the oncogenicity of oral carcinoma by concomitant

targeting of the IRAK1, TRAF6 and NUMB Genes. PLoS ONE 2013;8:e79926.

30. Labbaye C, Testa U. The emerging role of MIR-146A in the control ofhematopoiesis, immune function and cancer. J Hematol Oncol 2012;5:1–10.

31. Bhaumik D, Scott GK, Schokrpur S, Patil CK, Campisi J, Benz CC. Expres-sion of microRNA-146 suppresses NF-[kappa]B activity with reduction ofmetastatic potential in breast cancer cells. Oncogene 2008;27:5643–7.

32. PhilippidouD, Schmitt M,Moser D,Margue C, Nazarov PV,Muller A, et al.Signatures of MicroRNAs and Selected MicroRNA Target Genes in HumanMelanoma. Cancer Research 2010;70:4163–73.

33. MoloneyDJ, Panin VM, Johnston SH,Chen J, Shao L,WilsonR, et al. Fringeis a glycosyltransferase that modifies Notch. Nature 2000;406:369–75.

34. Panin VM, Shao L, Lei L, Moloney DJ, Irvine KD, Haltiwanger RS. Notchligands are substrates for ProteinO-Fucosyltransferase-1 and Fringe. J BiolChem 2002;277:29945–52.

35. Wang Z, Li Y, Banerjee S, Kong D, Ahmad A, Nogueira V, et al. Down-regulation of Notch-1 and Jagged-1 inhibits prostate cancer cell growth,migration and invasion, and induces apoptosis via inactivation of Akt,mTOR, and NF-kB signaling pathways. J Cell Biochem 2010;109:726–36.

36. KimM-H, KimH-B, Yoon SP, Lim S-C, ChaMJ, Jeon YJ, et al. Colon cancerprogression is driven by APEX1-mediated upregulation of Jagged. J ClinInvest 2013;123:3211–30.

37. Zhang S, Chung W-c, Wu G, Egan SE, Xu K. Tumor-Suppressive activity oflunatic fringe in prostate throughdifferentialmodulationof notch receptoractivation. Neoplasia 2014;16:158–67.

38. Xu K, Usary J, Kousis Philaretos C, Prat A, Wang D-Y, Adams Jessica R, et al.Lunatic fringe deficiency cooperates with the met/caveolin gene ampliconto induce basal-like breast cancer. Cancer Cell 2012;21:626–41.

39. Sauter ER, Yeo U-C, von Stemm A, Zhu W, Litwin S, Tichansky DS, et al.Cyclin D1 is a candidate oncogene in cutaneous melanoma. Cancer Res2002;62:3200–6.

40. Palomero T, Ferrando A. Oncogenic NOTCH1 Control of MYC and PI3K:Challenges and Opportunities for Anti-NOTCH1 Therapy in T-Cell AcuteLymphoblastic Leukemias and Lymphomas. Clin Cancer Res 2008;14:5314–7.

41. Sanz-Moreno V. Tumour Invasion: A New Twist on Rac-Driven Mesen-chymal Migration. Cur Biol 2012;22:R449–R51.

42. Cheresh DA, Spiro RC. Biosynthetic and functional properties of an Arg-Gly-Asp-directed receptor involved in human melanoma cell attachmentto vitronectin, fibrinogen, and von Willebrand factor. J Biol Chem1987;262:17703–11.

43. Taganov KD, Boldin MP, Chang K-J, Baltimore D. NF-kB-dependentinduction of microRNA miR-146, an inhibitor targeted to signaling pro-teins of innate immune responses. Proc Natl Acad Sci U S A 2006;103:12481–6.

44. Wei L, Zhou Q, Hou S, Bai L, Liu Y, Qi J, et al. MicroRNA-146a and Ets-1Gene Polymorphisms Are Associated with Pediatric Uveitis. PLoS ONE2014;9:e91199.

45. Wang X, Gao H, Ren L, Gu J, Zhang Y, Zhang Y. Demethylation of themiR-146a promoter by 5-Aza-20-deoxycytidine correlates with delayed progres-sion of castration-resistant prostate cancer. BMC Cancer 2014;14:1–11.

46. Nguyen D. Quantifying chromogen intensity in immunohistochemistryvia reciprocal intensity. 2013.

Mol Cancer Res; 14(6) June 2016 Molecular Cancer Research562

Raimo et al.

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Page 16: miR-146a Exerts Differential Effects on Melanoma Growth ...mcr.aacrjournals.org/content/molcanres/14/6/548.full.pdf · miR-146a Exerts Differential Effects on Melanoma Growth and

2016;14:548-562. Mol Cancer Res   Monica Raimo, Francesca Orso, Elena Grassi, et al.   MetastatizationmiR-146a Exerts Differential Effects on Melanoma Growth and

  Updated version

  http://mcr.aacrjournals.org/content/14/6/548

Access the most recent version of this article at:

   

   

  Cited articles

  http://mcr.aacrjournals.org/content/14/6/548.full#ref-list-1

This article cites 43 articles, 17 of which you can access for free 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/14/6/548To request permission to re-use all or part of this article, use this link

on February 15, 2019. © 2016 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from