nuclear factor of activated t-cell activity is associated ... · molecular and cellular...

11
Molecular and Cellular Pathobiology Nuclear Factor of Activated T-cell Activity Is Associated with Metastatic Capacity in Colon Cancer Manish K. Tripathi 1 , Natasha G. Deane 1,2 , Jing Zhu 3 , Hanbing An 1 , Shinji Mima 1 , Xiaojing Wang 3 , Sekhar Padmanabhan 1 , Zhiao Shi 4 , Naresh Prodduturi 3 , Kristen K. Ciombor 5 , Xi Chen 6 , M. Kay Washington 7 , Bing Zhang 2,3,8 , and R. Daniel Beauchamp 1,2,8,9 Abstract Metastatic recurrence is the leading cause of cancer-related deaths in patients with colorectal carcinoma. To capture the molecular underpinnings for metastasis and tumor progression, we performed integrative network analysis on 11 independent human colorectal cancer gene expression datasets and applied expression data from an immunocompetent mouse model of metastasis as an additional lter for this biologic process. In silico analysis of one metastasis-related coexpression module predicted nuclear factor of activated T-cell (NFAT) transcription factors as potential regulators for the module. Cells selected for invasiveness and metastatic capability expressed higher levels of NFATc1 as compared with poorly metastatic and less invasive parental cells. We found that inhibition of NFATc1 in human and mouse colon cancer cells resulted in decreased invasiveness in culture and downregulation of metastasis-related network genes. Overexpression of NFATc1 signicantly increased the metastatic potential of colon cancer cells, whereas inhibition of NFATc1 reduced metastasis growth in an immunocompetent mouse model. Finally, we found that an 8-gene signature comprising genes upregulated by NFATc1 signicantly correlated with worse clinical outcomes in stage II and III colorectal cancer patients. Thus, NFATc1 regulates colon cancer cell behavior and its transcriptional targets constitute a novel, biologically anchored gene expression signature for the identication of colon cancers with high risk of metastatic recurrence. Cancer Res; 74(23); 694757. Ó2014 AACR. Introduction Colorectal carcinoma is a leading cause of cancer-related deaths in the United States (1, 2). Conventional staging is insufcient to adequately predict metastasis or recurrence in colorectal cancer, particularly for patients with stage II or stage III disease (3). Over the past several years, gene expression proling has been used to predict patient outcomes indepen- dently of conventional staging; however, the biologic mechan- isms driving poor clinical outcomes remain incompletely understood. Furthermore, because published prognostic colo- rectal cancer gene expression signatures are nonoverlapping, inference of regulatory molecular targets driving clinical out- comes poses a signicant challenge (4, 5). Nevertheless, it is likely that the combined data underlying these studies provide enough biologic information for inference of potential tran- scriptional regulators of colorectal cancer progression and metastasis. In the present study, we reasoned that the gene expression signatures of metastasis in mouse models and poor prognosis in humans reect a functional consequence of the deregulation of coregulated gene networks. Accordingly, we integrated eleven human microarray datasets, representing data from 1,295 colorectal cancer tumor specimens, to produce a set of coexpressed gene modules. These coexpression modules were ltered using gene expression data from an immunocompetent mouse model of metastatic colon cancer to identify a metas- tasis-associated coexpression module. We further predicted the nuclear factor of activated T cell (NFAT) family of tran- scription factors as a primary regulator of 21 genes in the module. NFAT is a cell signaling molecule involved in complex adaptive systems particular to vertebrate biology related to the Rel family of cell signal proteins (6, 7). NFATs are regulated by receptor-mediated calcium/calmodulin signaling driving nuclear transport where they control transcriptional events governing functions as diverse as cell proliferation, survival 1 Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee. 2 Vanderbilt Ingram Cancer Center, Vanderbilt University Med- ical Center, Nashville, Tennessee. 3 Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee. 4 Advanced Computing Center for Research & Education, Vanderbilt University, Nash- ville, Tennessee. 5 Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio. 6 Department of Biostatistics, Vanderbilt University Med- ical Center, Nashville, Tennessee. 7 Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee. 8 Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee. 9 Department of Cell and Development Biology, Vanderbilt University Med- ical Center, Nashville, Tennessee. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Authors: Robert D. Beauchamp, Department of Surgery, Vanderbilt University Medical Center, 1161 21st Avenue South, MCN D4316, Nashville, TN 37232-2730. Phone: 615-322-2363; Fax: 615-343- 5365; E-mail: [email protected]; and Bing Zhang, E-mail: [email protected] doi: 10.1158/0008-5472.CAN-14-1592 Ó2014 American Association for Cancer Research. Cancer Research www.aacrjournals.org 6947

Upload: others

Post on 05-Jul-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Nuclear Factor of Activated T-cell Activity Is Associated ... · Molecular and Cellular Pathobiology Nuclear Factor of Activated T-cell Activity Is Associated with Metastatic Capacity

Molecular and Cellular Pathobiology

Nuclear Factor of Activated T-cell Activity Is Associated withMetastatic Capacity in Colon Cancer

Manish K. Tripathi1, Natasha G. Deane1,2, Jing Zhu3, Hanbing An1, Shinji Mima1, Xiaojing Wang3,Sekhar Padmanabhan1, Zhiao Shi4, Naresh Prodduturi3, Kristen K. Ciombor5, Xi Chen6, M. Kay Washington7,Bing Zhang2,3,8, and R. Daniel Beauchamp1,2,8,9

AbstractMetastatic recurrence is the leading cause of cancer-related deaths in patients with colorectal carcinoma. To

capture the molecular underpinnings for metastasis and tumor progression, we performed integrative networkanalysis on 11 independent human colorectal cancer gene expression datasets and applied expression data froman immunocompetentmousemodel ofmetastasis as an additional filter for this biologic process. In silico analysisof one metastasis-related coexpression module predicted nuclear factor of activated T-cell (NFAT) transcriptionfactors as potential regulators for the module. Cells selected for invasiveness andmetastatic capability expressedhigher levels of NFATc1 as compared with poorly metastatic and less invasive parental cells. We found thatinhibition of NFATc1 in human and mouse colon cancer cells resulted in decreased invasiveness in culture anddownregulation of metastasis-related network genes. Overexpression of NFATc1 significantly increased themetastatic potential of colon cancer cells, whereas inhibition of NFATc1 reduced metastasis growth in animmunocompetent mouse model. Finally, we found that an 8-gene signature comprising genes upregulated byNFATc1 significantly correlated with worse clinical outcomes in stage II and III colorectal cancer patients. Thus,NFATc1 regulates colon cancer cell behavior and its transcriptional targets constitute a novel, biologicallyanchored gene expression signature for the identification of colon cancers with high risk ofmetastatic recurrence.Cancer Res; 74(23); 6947–57. �2014 AACR.

IntroductionColorectal carcinoma is a leading cause of cancer-related

deaths in the United States (1, 2). Conventional staging isinsufficient to adequately predict metastasis or recurrence incolorectal cancer, particularly for patientswith stage II or stageIII disease (3). Over the past several years, gene expressionprofiling has been used to predict patient outcomes indepen-dently of conventional staging; however, the biologic mechan-

isms driving poor clinical outcomes remain incompletelyunderstood. Furthermore, because published prognostic colo-rectal cancer gene expression signatures are nonoverlapping,inference of regulatory molecular targets driving clinical out-comes poses a significant challenge (4, 5). Nevertheless, it islikely that the combined data underlying these studies provideenough biologic information for inference of potential tran-scriptional regulators of colorectal cancer progression andmetastasis.

In the present study, we reasoned that the gene expressionsignatures of metastasis in mouse models and poor prognosisin humans reflect a functional consequence of the deregulationof coregulated gene networks. Accordingly, we integratedeleven human microarray datasets, representing data from1,295 colorectal cancer tumor specimens, to produce a set ofcoexpressed gene modules. These coexpression modules werefiltered using gene expression data froman immunocompetentmouse model of metastatic colon cancer to identify a metas-tasis-associated coexpression module. We further predictedthe nuclear factor of activated T cell (NFAT) family of tran-scription factors as a primary regulator of 21 genes in themodule.

NFAT is a cell signaling molecule involved in complexadaptive systems particular to vertebrate biology related tothe Rel family of cell signal proteins (6, 7). NFATs are regulatedby receptor-mediated calcium/calmodulin signaling drivingnuclear transport where they control transcriptional eventsgoverning functions as diverse as cell proliferation, survival

1Department of Surgery, Vanderbilt University Medical Center, Nashville,Tennessee. 2Vanderbilt Ingram Cancer Center, Vanderbilt University Med-ical Center, Nashville, Tennessee. 3Department of Biomedical Informatics,Vanderbilt University Medical Center, Nashville, Tennessee. 4AdvancedComputing Center for Research & Education, Vanderbilt University, Nash-ville, Tennessee. 5Division of Medical Oncology, Department of InternalMedicine, The Ohio State University Comprehensive Cancer Center,Columbus, Ohio. 6Department of Biostatistics, Vanderbilt University Med-ical Center, Nashville, Tennessee. 7Department of Pathology, VanderbiltUniversity Medical Center, Nashville, Tennessee. 8Department of CancerBiology, Vanderbilt University Medical Center, Nashville, Tennessee.9Department of Cell and Development Biology, Vanderbilt University Med-ical Center, Nashville, Tennessee.

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

Corresponding Authors: Robert D. Beauchamp, Department of Surgery,Vanderbilt University Medical Center, 1161 21st Avenue South, MCND4316, Nashville, TN 37232-2730. Phone: 615-322-2363; Fax: 615-343-5365; E-mail: [email protected]; and Bing Zhang, E-mail:[email protected]

doi: 10.1158/0008-5472.CAN-14-1592

�2014 American Association for Cancer Research.

CancerResearch

www.aacrjournals.org 6947

Page 2: Nuclear Factor of Activated T-cell Activity Is Associated ... · Molecular and Cellular Pathobiology Nuclear Factor of Activated T-cell Activity Is Associated with Metastatic Capacity

and differentiation (8, 9), epithelial-to-mesenchymal transition(EMT; refs. 6, 10, 11), and stem cell quiescence (12). NFATs alsomodulate inflammatory cell function, angiogenesis, cell migra-tion and invasion (11, 13), and regulation of tumor stromal cellfunction in both a cell-type–specific and context-dependentmanner (14).

Here, we used a systems biology approach to identify NFATas a potential regulator of colon cancer progression andmetastasis. A schematic of our overall workflow appears inSupplementary Fig. S1. We experimentally determined thatexpression of NFATc1, specifically, promotes increased coloncancer cell invasion and metastasis, and that expression of theNFATc1-driven transcriptional program correlates with poorprognosis in stage II and III colorectal cancer patients.

Materials and MethodsHuman and mouse gene expression data

Human gene expression datasets were downloaded from theGene Expression Omnibus (GEO) database (http://www.ncbi.nlm.nih.gov/geo/) and the ArrayExpress Archive (http://www.ebi.ac.uk/microarray-as/ae/; Supplementary Table S1). Eachgene expression datasetwas processed separately to generate agene by sample expression matrix with normalized, log-trans-formed, and standardized expression values (see Supplemen-tary Methods). The mouse gene expression dataset was down-loaded from the GEO GSE19073 (15) and the differentialexpression analysis was described in detail in SupplementaryMethods.

Integrated network construction, coexpression moduleidentification, and enrichment analysis

For each human CRC dataset selected for integration,Pearsons correlation coefficients and the log likelihood ratio(LLR) for functional relevance were calculated for all genepairs. Next, an integrated LLR score was calculated for eachgene pair, and a score cutoff was selected to achieve a balancebetween consistency with existing knowledge and networkcoverage. An integrated gene coexpression network was thenconstructed in which each node is a gene while two nodes areconnected by an edge if corresponding LLR score is above theselected cutoff (for additional detail, see Supplementary Meth-ods). For the identification of coexpressionmodules, we used amodified version of the Iterative Clique Enumeration (ICE)algorithm (16). GO biologic process enrichment analysis of thecoexpression modules and transcription factor target enrich-ment analysis were performed using WebGestalt (17). Theranked list of genes according to their differential expressionlevels in the mouse dataset was used to identify metastasis-related modules using the GseaPreranked analysis available inthe GSEA software downloaded from http://www.broadinsti-tute.org/gsea/. Default parameters were used for the analysis.

Survival analysisSurvival analysis was performed in R using the survival

package. Specifically, survival curves were estimated using theKaplan–Meier method, and survival comparisons amonggroups were made by the log-rank test. Two datasets were

used for the survival analysis. The dataset GSE17536 was fromthe Moffitt Cancer Center (MCC; Tampa, FL) and has beenpreviously published. The dataset GSE38832 was from theVanderbilt University Medical Center (VUMC; Nashville, TN),which included 54 samples from the previously publisheddataset GSE17537 and 68 newly analyzed samples. The COM-BAT approach based on empirical Bayes frameworks wasapplied to remove batch effects across the two batches ofmicroarray experiments (18).

Human tissue samplesHuman tissues used for microarray analysis were collected

following written informed consent and clinically annotatedaccording to established protocols and approved by the appro-priate Institutional ReviewBoards at theMoffitt Cancer Centerand Vanderbilt University as previously described (GSE17536and GSE17537; ref. 15).

RNA preparation and analysisTotal RNA from cells or tissues was isolated using QIAGEN

kits (QIAGEN) and DNase-I treated, quantified by Nanodrop-1000 (Thermo Scientific) and assessed for quality on an AgilentBioanalyzer as previously described (15). Chromosome immu-noprecipitation (ChIP) studies were conducted using mouseanti-NFATc1–specific antibody from Santa Cruz Biotechnol-ogy and a kit from Millipore, according to the manufacturer'sinstructions. qRT-PCR (qPCR) was performed as describedelsewhere (19), gene-specific primers are listed in Supplemen-tary Tables S2 and S3.

Cell cultureThe MC-38 mouse adenocarcinoma cell line and its deriva-

tives were provided by Dr. Robert Coffey (Department ofInternal Medicine, Vanderbilt University Medical Center,Nashville, TN) are described elsewhere (15). HCT116 and HT29colon cancer cell lines were obtained from ATCC. All cell lineswere maintained at low passage as monolayers in RPMI-1640medium (Gibco Life Technologies), supplemented with 10%FBS (Atlanta Biologicals), 500 U/mL penicillin G, 500 mg/mLstreptomycin (Gibco Life Technologies Inc.), and L-glutamine(Gibco Life Technologies Inc.). FK506 (Sigma) was used at 20ng/mL. Integrity of human cell lines used in this study wastested by RNAseq analysis in May 2013. Cytoplasmic andnuclear extracts were prepared using Nuclear Extract kit(Active Motif), according to manufacturer's instructions.

Immunoblot analysesCells were harvested in RIPA lysis buffer (50 mmol/L Tris,

pH7.5, 150 mmol/L NaCl, 1% NP-40, 0.5% Na-deoxy Cholate,0.1% SDS) containing a cocktail of protease inhibitors (RocheDiagnostics), with a brief sonication. Samples were mixed withLDS buffer containing DTT (Invitrogen), and fractionated on4% to 12% NuPAGE gels in MOPS-SDS buffer (Invitrogen).Antibodies against NFATc1-c4 and PARP1/2 were obtainedfrom Santa Cruz Biotechnology, b-actin from Sigma Chemical,and a-tubulin from Abcam Scientific. Ramos cell (Burkittlymphoma, B lymphocytes) lysate (Santa Cruz Biotechnology)was used as positive control.

Tripathi et al.

Cancer Res; 74(23) December 1, 2014 Cancer Research6948

Page 3: Nuclear Factor of Activated T-cell Activity Is Associated ... · Molecular and Cellular Pathobiology Nuclear Factor of Activated T-cell Activity Is Associated with Metastatic Capacity

Invasion assaysInvasion assays were conducted using both Boyden cham-

bers as described elsewhere (15) as well as the xCELLigencesystem from Roche Diagnostics (Supplementary Methods).

Overexpression and inhibition of NFATc1RNAi studies were performed as previously described using

NFATc1-specific ON-target plus SMART pool siRNA or ON-target plus Non-targeting Pool (Thermo Scientific). Specific si-RNA sequences are given in Supplementary Table S4.For preparing stable overexpressing cells, mouse wild-

type NFATc1 (Addgene plasmid 11101; ref. 20) was clonedinto vector pGP-Lenti3 (GenBank Accession no. JX861384)between unique XbaI and BamHI sites. Puromycin and GFPexpressions were used as selection markers for creation ofstable cell lines.

For gene knockdown experiments, NFATc1-specific GIPZlentiviral shRNAmir (V3LMM_418820; Thermo Scientific)was selected for experimental use. Briefly, MC38 cells wereelectroporated using NEON (Invitrogen). Puromycin andGFP expressions were used as selection markers for creationof stable cell lines. For HT29 studies, cells were transientlycotransfected with pEF6-NFATc1 and pMaxGFP vectors,flow sorted to enrich for highly expressing GFP-positivecells, and confirmed NFATc1 overexpression by Westernblot analysis.

Animal studiesAll animal studies were approved by the Vanderbilt Insti-

tutional Animal Care and Use Committee and performed inaccordance with the standards of the Association of Assess-ment and Accreditation of Laboratory Care. Procedures were

E–2 updown

10–10 updown

Transcription factor mRNA

mRNA differential expression

2

Translational elongation (n = 105)

Developmental process (n = 187)

Cell cycle (n = 107)

Unknown function (n = 72)

Immune system process (n = 85)

B

FDR=0.0008

0 2 4 6 8

0.0

0.2

0.4

0.6

0.8

1.0

Functional similarity

Cum

ulat

ive

dist

ribut

ion

C

Non

-m

etas

tatic

Met

asta

tic

Core(63 genes)

FDR < 0.001

D

A

Figure 1. Identification of NFAT as a potential regulator of metastasis. A, cumulative distribution function curves of semantic similarities for gene pairs incoexpression networks based on LLR cutoffs of 0.5 (cyan), 1 (pink), and 1.25 (green), compared with gene pairs in the stand alone coexpression networkderived from GSE17536 with a Pearson correlation coefficient cutoff of 0.74 (dotted pink). Cumulative distribution function curves for all gene pairs andinteracting protein pairs are included as negative (black) and positive (red) references, respectively. B, the largest five coexpressionmodules, each depicted ina distinct color scheme and annotated with associated biological processes are shown. Red nodes indicate genes shared between modules. C, GSEAanalysis indicates enrichment of mouse model genes in the module representative of developmental processes (FDR < 0.001). D, expression pattern for allgenes in the developmental processmodule appearing in themouse data, with the 63 leading edge genes identifiedbyGSEAdenoted as core genes. E, Fisherexact test indicates significant enrichment of NFAT targets (21 genes, FDR ¼ 0.0008) among the 63 core genes.

NFATc1 Regulates Colon Cancer Metastasis

www.aacrjournals.org Cancer Res; 74(23) December 1, 2014 6949

Page 4: Nuclear Factor of Activated T-cell Activity Is Associated ... · Molecular and Cellular Pathobiology Nuclear Factor of Activated T-cell Activity Is Associated with Metastatic Capacity

performed using both parental and stable transfected cell linesas previously described (15).

Statistical analyses for experimental proceduresStatistical analyses for all experimental procedures were

conducted in GraphPad Prism 5 (GraphPad Software). P < 0.05was considered statistically significant.

ResultsIntegrated human colorectal cancer coexpressionnetwork

We first identified 14 publicly available human colorectalcancer gene expression datasets (Supplementary Table S1)and eliminated three sets, for which the association betweenpairwise gene coexpression and functional relevance was notclear (Supplementary Results and Supplementary Fig. S2).We also found that the 11 remaining datasets containcomplementary functional information (SupplementaryResults, Supplementary Fig. S3) and were therefore suitablefor integration and development of a comprehensive coex-pression network.

Following Ramani and colleagues (21), we constructed threecoexpression networks based on three LLR cutoffs and eval-uated the functional relevance of the networks using randomgene pairs and interacting protein pairs (SupplementaryMeth-ods) as negative and positive references, respectively. As shownin Fig. 1A, all three networks (cyan, pink, and green curves)clearly outperformed the negative reference (black), and ahigher LLR cutoff corresponded to higher functional relevanceof gene pairs in the coexpression network. The functionalrelevance of the coexpression network derived from an LLRcutoff of 1.25 (green) approached that for the curated proteininteraction network (red), suggesting a high biologic validity ofthe coexpression network. To capture novel functional rela-tionships offered by the coexpression network analysis thatmay not be supported by existing knowledge, we used a slightlyrelaxed LLR cutoff of 1 and constructed the integrated colo-rectal cancer coexpression network with 2,285 genes and13,083 edges (Supplementary File S1). A coexpression networkderived from the GSE17536 dataset with an LLR cutoff of 0.74showed a similar level of functional relevance as that of theintegrated network (Fig. 1A, dashed pink curve). However, it

Figure 2. Inhibition of NFATc1 inMC38Met cells. Effect of control RNAi (siScr) or NFATc1-specific RNAi on specific NFATmRNA species: NFATc1 (A), NFATc2(B), NFATc3 (C). D, the effect of NFATc1 siRNA versus scrambled control si-Scr onNFAT family protein levels inMC38Met cells, and steady state levels in bothMC38Par and MC38Met cells with Ramos cell lysate positive control in the right hand lane. E, relative rates of invasion for cells shown in A–D . ���, P < 0.0005;����, P < 0.00005.

Tripathi et al.

Cancer Res; 74(23) December 1, 2014 Cancer Research6950

Page 5: Nuclear Factor of Activated T-cell Activity Is Associated ... · Molecular and Cellular Pathobiology Nuclear Factor of Activated T-cell Activity Is Associated with Metastatic Capacity

included only 1,226 genes and 7,516 edges. Therefore, dataintegration helped achieve a significantly better network cov-erage with 86% more genes and 74% more edges. The inte-grated network represents one of the most comprehensivecoexpression networks for a specific cancer type and can serveas a resource for other investigators for the study of cancer-associated gene function and identification of candidate can-cer driver gene programs.

Transcriptional programs encoded in the networkThe integrated network had a high clustering coefficient of

0.58, indicating a highly modular network structure (22). Usinga modified version of the ICE algorithm (16), we identified 441coexpressionmodules (Supplementary Fig. S4 and Supplemen-tary File S2) from the network, among which 38 included 10 ormore genes.Figure 1B depicts the five largest modules with 187, 107, 105,

85, and 72 genes, respectively. Four of the top five modulesshowed significant enrichment (FDR <0.01, hypergeometrictest) in distinct biologic processes including developmentalprocess, cell cycle, translational elongation, and immune sys-temprocess, respectively. The fifthmodule was not enriched inany of the defined GO biologic processes (labeled as unknownfunction). Interestingly, among the 72 genes in this module,only 27 (37%) had aGObiologic process annotation, in contrast

with an average of 85% annotated genes in the other fourmodules. It is possible that this module may represent atranscriptional program underlying an understudied biologicfunction.

Metastasis-related transcriptional programs andcandidate effectors

We ranked all human genes based on the differentialexpression level of their mouse orthologs in the poorly met-astatic parentalMC38 cell line (designated asMC38Par) and thehighly metastatic MC38 cell-derived colon cancer cell line(MC38Met; ref. 15). For each coexpression module, we usedthe GSEA to test whether genes in the module were signifi-cantly enriched in the top or bottom of the ranked list,indicating coordinate up- or downregulation of the modulein the metastatic cell line.

The module consisting of 187 genes functionally linked todevelopmental process (Fig. 1B) was enriched with genesupregulated in the metastatic cells (FDR < 0.001), whereasthe module consisting of 107 genes functionally linked to cellcycle was enriched with genes that are downregulated in themetastatic cells (FDR ¼ 0.01). This pattern is consistent withreduced cell proliferation and increased motility in cellsundergoing EMT, a developmental process implicated incancer metastasis (23). Owing to its significant association

Figure 3. NFATc1 expression is associated with cell invasion in human colon cancer cells. A, Western blot analysis showing NFATc1 levels in vector-transfected and NFATc1-transfected HT29 colon cancer cells. B, representative live cell measures after a 24-hour growth period using pools of HT29 cellsshown in A. ns, not significant. C, relative rates of invasion through Matrigel at 24 hours (�, P < 0.05) of cells depicted in A. D, Western blot analysis showingprotein expression of NFATc1-c3 family members in HCT116 cells treated with either control RNAi (si-Scr) or NFATc1-specific RNAi (si-NFATc1). E,representativemeasuresofHCT116 cells shown inD . F, relativemeasures of live cell growth ofHCT116 control (Scr) andNFATc1 knockdown (siNFATc1) after24 hours in culture. ns, not significant. G, relative rate of HCT116 cell invasion through Matrigel shown in D–F (representative experiment, �� P < 0.005).

NFATc1 Regulates Colon Cancer Metastasis

www.aacrjournals.org Cancer Res; 74(23) December 1, 2014 6951

Page 6: Nuclear Factor of Activated T-cell Activity Is Associated ... · Molecular and Cellular Pathobiology Nuclear Factor of Activated T-cell Activity Is Associated with Metastatic Capacity

to the metastatic cell gene expression profile, we focused onthe 187-gene module for further study. We identified 63 coregenes in this module that accounted for the enrichmentsignal in the metastatic cells (Fig. 1C; Supplementary FileS3). As shown in Fig. 1D, mouse orthologs of the 63 coregenes showed clear upregulation in MC38Met compared withMC38Par, whereas those of non-core genes were either notdifferentially expressed or downregulated in MC38Met. Thisresult indicates that a substantial portion, but not all, of thecoexpression relationship was conserved between humantumors and the mouse model. The 63 core genes are pos-itively correlated in human and mouse, and thus, serve ascandidate effectors of a transcriptional program associatedwith a metastatic phenotype.

Identification of NFAT as a potential regulator of themetastasis-associated transcriptional program

We performed enrichment analysis for the 63 core genesagainst the gene sets of transcription factor targets andfound that one third of the genes in the module wereputative targets of the NFAT family transcription factors,representing significant enrichment (FDR ¼ 0.0008, Fisherexact test, Fig. 1E and Supplementary File S4). As a set, these

genes are functionally enriched for skeletal and muscularsystem development, connective tissue development, and forcell movement, growth, and proliferation (SupplementaryTable S5).

We next assessed the mRNA expression of NFAT familymembers and 21 putative target genes in 22 stage II and IIIfresh-frozen primary human colorectal cancer tumor speci-mens. We found that mRNA for 19 out of the 21 putativeNFAT target genes were significantly upregulated in thetumor specimens as compared with normal adjacent tissue(Supplementary Fig. S5). We next assessed the expression ofindividual NFAT members and the 21 target genes usingmicroarray data from the isogenic mouse cell lines and fromprimary tumor data. Among the four NFAT members exam-ined, NFATc1 mRNA was the most differentially expressed(FDR ¼ 0.0001) between MC38Met and MC38Par cells, withalmost 3-fold (P < 3E-06, moderated t test) increase inmetastatic cells (Supplementary Table S6). Moreover, twoNFATc1 probe sets (21105_s_at and 210162_s_at) correlatedbest with the major cluster of the 21 metastasis-relatedNFAT transcriptional target genes in three independentpatient cohorts (Supplementary Fig. S6B–S6D). These datasuggested that NFATc1, specifically, is associated with a

Figure 4. Evidence for a dominant set of NFATc1 target genes. A, expression of eight putative target mRNA species significantly associated with NFATc1expression inMC38Met cells followingFK506 treatment (relative toDMSOcontrol,n¼4, significance isdeterminedbyone-sample t test against equivalenceorratio¼ 1 as shown by vertical line). B, expression of eightmRNA species significantly associatedwithNFATc1 expression inHCT116 cells following treatmentwithNFATc1-specificRNAi (relative to siSCR, n¼ 4, significance is determined by one-sample t test against equivalence or ratio¼ 1 as shownby vertical line).C, quantification of NFATc1 pulldown on putative target gene-specific promoter binding elements relative to IgG control (ASPNmRNA species undetectable,not shown). Not significant, P > 0.05; �, P < 0.05; ��, P < 0.005; ���, P < 0.0005; ����, P < 0.00005.

Tripathi et al.

Cancer Res; 74(23) December 1, 2014 Cancer Research6952

Page 7: Nuclear Factor of Activated T-cell Activity Is Associated ... · Molecular and Cellular Pathobiology Nuclear Factor of Activated T-cell Activity Is Associated with Metastatic Capacity

metastasis-associated transcriptional program in humancolorectal cancer.

NFATc1 expression is associated with invasive behaviorof colon cancer cellsWe found that pharmacologic inhibition of NFATs using

FK506 reduced both cell invasiveness and expression of metas-tasis-associated target genes (Supplementary Fig. S7). We nextwanted to assess the role of NFATc1 expression, specifically, inregulating invasiveness.We found that experimental inhibitionof NFATc1 expression in MC38Met cells by RNAi resulted in aspecific decrease in NFATc1 mRNA and protein expression.In Fig. 2, fourmembers of theNFAT family are evaluated for off-target effects of the RNAi at themRNA and protein levels. First,the effect of siNFATc1 on NFATc1-specific mRNA levels isevaluated in MC38Met cells relative to siSCR control andparental lines (Fig. 2A). Next, the effect of the RNAis areevaluated on NFATc2 (Fig. 2B) and NFATc3 (Fig. 2C)-specificmRNA expression, demonstrating no off-target effects relativeto parental siSCR controls. In Fig. 2D, the specificity of theNFATc1-specific knockdown can be appreciated at the proteinlevel, as well, with no off-target effects shown for NFAT family

members c2-c4. Importantly, treatment of MC38Met withNFATc1-specific RNAi caused a significant reduction in cellinvasion ofMC38Met cells (Fig. 2E). Thus, the siNFATc1-specificRNAi (a pool of four distinct RNAiswas used in this experiment,sequences are given in Supplementary Table S4) has no detect-able off-target effects on NFAT family member-specific mRNAspecies or proteins, demonstrating the specificity of theobserved effects on cell invasion.

To determine the relevance of NFATc1 expression in humancancer cells, we manipulated expression levels in HT29 (low toundetectable NFATc1 expression) and in HCT116 cells (highendogenous NFATc1 expression). We found that transientoverexpression of NFATc1 in HT29 cells (Fig. 3A) had no effecton cell viability or proliferation (Fig. 3B) but significantlyincreased cell invasion (Fig. 3C). In contrast, when RNAi wasused to specifically inhibit NFATc1 expression in HCT116 cells(Fig. 3D and E), we found that targeted RNAi treatment did notsignificantly affect cell proliferation (Fig. 3F) and was associ-ated with a significant decrease in the rate of invasion throughMatrigel as compared with the siSCR control (Fig. 3G).

NFATc1 activity is primarily regulated at the posttransla-tional level by phosphorylation, so variations in NFATc1

Sur

viva

l pro

babi

lity

Vanderbilt and Moffitt combinedNFATc1 signature (8 genes); OS

Survival time (months)

Sur

viva

l pro

babi

lity

Vanderbilt and Moffitt combinedNFATc1 signature (8 genes); DFS

Survival time (months)

Sur

viva

l pro

babi

lity

Vanderbilt and Moffitt combinedNFATc1 signature (8 genes); DSS

Survival time (months)

DC E

A B

IIIII II

Moffitt Cancer Center: Heat map

II IIIFAP

COL3A1

ASPN

MRC2

ANGPTL2

MGP

PTRF

TWIST1

Vanderbilt Medical Center: Heat map

DSSDFS

Stage

OS

II IIIFAP

ASPN

MRC2

ANGPTL2

TWIST1

PTRF

MGP

COL3A1

DSSDFS

Stage

OS

−3 updown 3

Below median expression group Below median expression group Below median expression groupAbove median expression group Above median expression group Above median expression group

P = 3.02e–05 P = 6.5e–06 P = 0.000106n = 188 n = 188 n = 185

HR = 3.56 (1.89–6.74) HR = 7.89 (2.73–22.8) HR = 3.76 (1.83–7.7)

0.0

0.2

0

.4

0.6

0.8

1.

0

0.0

0.2

0

.4

0.6

0.8

1.

0

0.0

0.2

0

.4

0.6

0.8

1.

0

0 20 40 60 80 100 120 0 20 40 60 80 100 120 0 20 40 60 80 100 120

Figure 5. An NFAT-driven transcriptional program is correlated with poor outcomes in stage II and III colorectal cancer patients. A and B, NFATc1 signaturegene expression patterns in stages II and III colorectal cancer specimens in the Vanderbilt Medical Center (GSE 38832; A) and the Moffitt Cancer Center(GSE17536; B) datasets. The top three rows indicate whether or not a disease-specific death event or a recurrence event was recorded in follow-up (black, noevent; red, event; white, not available), respectively. The fourth row indicates the cancer stage (green, stage II; dark blue, stage III). C–E, OS, DSS, and DFSanalyses based on combined Vanderbilt and Moffitt datasets.

NFATc1 Regulates Colon Cancer Metastasis

www.aacrjournals.org Cancer Res; 74(23) December 1, 2014 6953

Page 8: Nuclear Factor of Activated T-cell Activity Is Associated ... · Molecular and Cellular Pathobiology Nuclear Factor of Activated T-cell Activity Is Associated with Metastatic Capacity

expression levels across human colon cancer specimens, par-ticularly in the epithelial compartment, are low (data notshown). However, the range of expression of the putativetargets of NFATc1 activity identified with our filtered integrat-ed data network is diverse and so may be used as surrogatebiomarkers for NFATc1 activity in human specimens. Com-bining the results of experiments conducted in MC38 cells, wefound that eight out of 21 genes from the metastatic moduleare consistently altered by NFAT activity and NFATc1 expres-sion inMC38Met cells (Fig. 4A and B, Supplementary Fig. S7 andSupplementary Table S5). These eight genes include Angio-poeitin (ANGPTL2), Asporin (ASPN), Collagen 3A1 (COL3A1),Matrix Gla Protein (MGP), Mannose receptor (MRC2), Fibro-blast activating protein (FAP), Polymerase I and TransferRelease Factor (PTRF), and TWIST1. ChIP assays confirmedenrichment of NFATc1 on promoters on seven out of eight ofthese genes in metastatic MC38 cells (Fig. 4C, SupplementaryFile S5). This 8-gene set is functionally enriched in cell move-ment (ANGPTL2, COL3A1, FAP, MGP, MRC2, TWIST1) andcancer (TWIST1, COL3A1, ANGPTL2, ASPN, FAP, MGP, MRC2,PTRF). Importantly, we also observed significant overlapbetween these eight genes and previously published coloncancer-specific prognostic signatures (15, 24–27), indicatingthat our integrative approach largely supports and builds upona significant group of published studies in this area by pro-viding a biologically based underpinning for the signature

(Supplementary Table S7). Although developmental pathwaysand activation of cell movement processes have been stronglylinked with cancer progression in other experimental models(15, 25–29), these findings are the first to link them with cellautonomousNFATc1 expression and invasive activity in tumorcells.

To test whether these eight genes can be used to identifycolorectal cancers with poor prognosis, we examined theirexpression levels in two clinically annotated cohorts ofstage II and III colorectal cancer tumors. As shownin Fig. 5A and B, increased expression of these genes isassociated with increased number of recurrence and deathevents for stage II and III patients. To statistically evaluatethe correlation of the eight genes with colorectal canceroutcomes, we performed a survival analysis based on acombined VUMC and MCC dataset. On the basis of theaverage of standardized gene expression levels of the eightgenes, patients were classified into two groups (abovemedian expression and below median expression). Wefound that the group of patients with above-median expres-sion had significantly shorter overall survival (OS: P ¼ 3.2e-05, HR ¼ 3.56; Fig. 5C), worse disease-specific survival (DSS;P ¼ 6.5E-06, HR ¼ 7.89, Fig. 5D), and shorter disease-freesurvival (DFS; P ¼ 1.1E-04, HR ¼ 3.76, Fig. 5E) as comparedwith the below-median group. These data show thatincreased expression of the NFATc1-driven transcriptional

Figure 6. Overexpression of NFATc1 in MC38Par cells increases tumor incidence and liver metastases. A, Western blot analysis showing NFAT proteins inMC38Par cells transfectedwith either empty vector (VEC)orNFATc1.b-actinwasusedasa loading control andRamosextract aspositivecontrol. B, analysis ofNFATc1 mRNA in MC38Par cells shown in A. C, rates of trans-endothelial invasion for MC38Par cells shown in A and B. Individual replicate wells from arepresentative experimentare plottedwith themeanand theSEM (bars andwhiskers).D, representativemice fromsplenicmetastasismodel (n¼12-14/group)using MC38Par cells shown in A–D. E, bioluminescence of mice injected with MC38Par cells shown in A–D at day 14 postinjection. F, incidence of livermetastases for mice injected at day 14. G, liver weight to body weight ratio in mice injected with either MC38Par shown in A–F cells at day 14 postinjection.�, P < 0.02; ���, P ¼ 0.0004; ����, P < 0.0001.

Tripathi et al.

Cancer Res; 74(23) December 1, 2014 Cancer Research6954

Page 9: Nuclear Factor of Activated T-cell Activity Is Associated ... · Molecular and Cellular Pathobiology Nuclear Factor of Activated T-cell Activity Is Associated with Metastatic Capacity

program correlates with worse outcomes for stage II and IIIpatients and suggest a novel role for NFATc1 in promotingtumor progression.

NFATc1 expression and themetastatic potential of MC38colon cancer cells in vivoTo determine whether the NFATc1-associated changes in

cell migration and invasion observed in cell culture are alsoassociated with changes in metastatic potential in vivo, wemanipulated NFATc1 expression in MC38 cells and testedtheir metastatic potential in isogenic and immunocompe-tent mice as described previously (15). For these studies, weperformed stable transfection of MC38Parcells with consti-tutively expressed NFATc1 (MC38ParþNFATc1; Fig. 6A and B)and showed that these cells exhibited a higher rate of trans-endothelial invasion as compared with the MC38ParþVec

control cells (Fig. 6C). For the in vivo experiments,mice injected intrasplenically with MC38ParþNFATc1 cellsexhibited a greater metastatic liver tumor incidence andoverall tumor burden as compared with those injected withMC38ParþVec control cells. Strikingly, liver metastasis fromMC38ParþNFATc1 cells were mostly diffuse and replaced mostof the liver parenchyma in contrast with very little meta-static colonization in mice injected with MC38ParþVec con-trol cells (n ¼ 12-14 mice per cell line, Fig. 6D–G and

Supplementary Fig. S8C and S8D). Moreover, target geneexpression was predictably upregulated in these cells, con-sistent with a regulatory role for this network in metastasisin vivo (Supplementary Fig. S8E). Thus, overexpression ofNFATc1 and the target gene network in MC38Par cellsincreased their metastatic potential in vivo.

In complementary studies, stable NFATc1-specific shRNA-mir expressing MC38Met cells (MC38MetþshNFATc1) or controlscrambled shRNAmir (MC38MetþshCtrl) cells were generated(Fig. 7A and B). We found that MC38MetþshNFATc1 cells exhib-ited a lower rate of invasion as compared with theMC38MetþshCtrl cells (Fig. 7C). Intrasplenic injection of theMC38MetþshNFATc1 cells resulted in reduced metastatic livertumor burden as measured by bioluminescence (Fig. 7D and E;P < 0.04), lower tumor incidence (Fig. 7F; P < 0.03), and reducedliver weight to body weight ratio (Fig. 7G; P < 0.0004) ascompared with mice injected with MC38MetþshCtrl cells (Sup-plementary Fig. S8A–S8C). Moreover, target gene expressionwas predictably downregulated in these cells and consistentwith a role for this NFATc1 network in regulating metastasisin vivo. Supplementary Table S8 summarizes the statistics ofthe in vivo experiments in table format.

Thus, the above-described in vivo studies confirm a rolefor tumor cell expression of NFATc1 in regulating themetastatic behavior of colon cancer cells in an

Figure 7. Knockdown of NFATc1 in MC38Met cells decreases tumor incidence and liver metastases. A, Western blot analysis showing NFAT proteins inMC38MetþshCtrl and MC38MetþshNFATc1 cell lines. b-actin was used as a loading control and Ramos cell extract as positive control. B, analysis of NFATc1specific mRNA in cells shown in A. C, rates of trans-endothelial invasion for cells are shown in A and B. Individual replicate wells from a representativeexperiment are plotted with the mean and the SEM (bars and whiskers). D, representative (n ¼ 14-15 mice/group) bioluminescence image correspondingday 21. E, summary- of bioluminescence signal quantified at day 21. F, summary incidence of liver metastases. G, liver metastases measured by liverweight to body weight ratio at 21 days postinjection. �, P < 0.03; ���, P < 0.001; ����, P < 0.0001.

NFATc1 Regulates Colon Cancer Metastasis

www.aacrjournals.org Cancer Res; 74(23) December 1, 2014 6955

Page 10: Nuclear Factor of Activated T-cell Activity Is Associated ... · Molecular and Cellular Pathobiology Nuclear Factor of Activated T-cell Activity Is Associated with Metastatic Capacity

immunocompetent mouse model, strengthening the evi-dence that such a mechanism can also play a role in humancolorectal cancer. However, our results neither rule out acooperative role of other NFAT members in disease pro-gression, as has been shown by others in pancreatic cancer(30–32), leukemia (33, 34), breast cancer (35, 36), and inmelanoma (37), nor they rule out cooperative effects of othersignaling pathways in contributing to these effects. Furtherstudies are needed to determine the precise regulation ofgenes from this module and to determine whether or notnon-cell autonomous signals may be required for full expres-sion of the prognostic genes.

In summary, we combinedmultiple publicly availablemicro-array datasets to identify a novel cell autonomous role forNFATc1 in regulating cell invasion and metastasis in coloncancer and to further link that activity to expression of eighttarget genes whose expression is associated with clinical out-comes in patient samples. A unique strength of this study is thequality evaluation of publicly available human microarraydatasets and systematic integration of the data into a coex-pression network for module analysis. Although the coexpres-sion modules were identified in an unsupervised manner,filtering the modules using expression data from a mousemodel allowed us to identify a group of coregulated geneswhose expression is associated with poor outcomes in humancolorectal cancer. Moreover, NFATc1, the regulator of a groupof coexpressed genes associated with poor colorectal cancerpatient outcomes, was demonstrated to promotemetastasis inmice. Thus, the integrative approach addresses the sample sizelimitations of earlier prognostic gene signature studies and theuse of the immunocompetent mouse model represents impor-tant aspects of a disease-appropriate microenvironment in themetastatic process.

Disclosure of Potential Conflicts of InterestThe authors disclose no potential conflicts of interest.

Authors' ContributionsConception and design: N. Deane, H. An, S. Mima, K. Ciombor, B. Zhang,R.D. BeauchampDevelopment of methodology: M.K. Tripathi, N. Deane, H. An, K. Ciombor,B. ZhangAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): M.K. Tripathi, N. Deane, H. An, C. Padmanabhan,K. Ciombor, N. Prodduturi, M.K. WashingtonAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis):M.K. Tripathi, N. Deane, J. Zhu, H. An, X.Wang, Z. Shi,X. Chen, M.K. Washington, B. Zhang, R.D. BeauchampWriting, review, and/or revision of themanuscript:M.K. Tripathi, N. Deane,J. Zhu, H. An, K. Ciombor, M.K. Washington, B. Zhang, R.D. BeauchampAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): M.K. Tripathi, N. Deane, H. An,K. Ciombor, B. ZhangStudy supervision: N. Deane, B. Zhang, R.D. Beauchamp

AcknowledgmentsThe authors thank Tanner J. Freeman for critical input and Connie Weaver

and Keeli Lewis for technical expertise.

Grant SupportThis work was supported by grants GM088822 (B. Zhang, R.D. Beau-

champ), CA095103 (R.D. Beauchamp), CA158472 (X. Chen and R.D. Beau-champ), K12 CA9060625 (X. Chen), the Vanderbilt-Ingram Cancer CenterSupport Grant (CA0684845), the Clinical and Translational Science Award toVanderbilt (UL1RR024975), Vanderbilt Digestive Disease Research Center(DK058404), and the Vanderbilt Cooperative Human Tissue NetworkU01CA094664 and NIH.

The costs of publication of this article were defrayed in part 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 May 29, 2014; revised August 12, 2014; accepted September 19, 2014;published OnlineFirst October 15, 2014.

References1. Siegel R, Naishadham D, Jemal A. Cancer statistics, 2012. CA Cancer

J Clin 2012;62:10–29.2. ACS. Cancer facts and figures. Atlanta Georgia: American Cancer

Society; 2012.3. Niedzwiecki D, Bertagnolli MM, Warren RS, Compton CC, Kemeny

NE, Benson AB III, et al. Documenting the natural history ofpatients with resected stage II adenocarcinoma of the colon afterrandom assignment to adjuvant treatment with edrecolomab orobservation: results from CALGB 9581. J Clin Oncol 2011;29:3146–52.

4. Kopetz S, Abbruzzese JL. Barriers to integrating gene profiling forstage II colon cancer. Clin Cancer Res 2009;15:7451–2.

5. Shi M, Beauchamp RD, Zhang B. A network-based gene expressionsignature informs prognosis and treatment for colorectal cancerpatients. PLoS ONE 2012;7:e41292.

6. Crabtree GR, Olson EN. NFAT signaling: choreographing the sociallives of cells. Cell 2002;109::S67–79.

7. Hogan PG, Chen L, Nardone J, Rao A. Transcriptional regulation bycalcium, calcineurin, and NFAT. Genes Dev 2003;17:2205–32.

8. Macian F. NFAT proteins: key regulators of T-cell development andfunction. Nat Rev Immunol 2005;5:472–84.

9. Wu H, Peisley A, Graef IA, Crabtree GR. NFAT signaling and theinvention of vertebrates. Trends Cell Biol 2007;17:251–60.

10. RangerAM,OukkaM,Rengarajan J,Glimcher LH. Inhibitory functionoftwo NFAT family members in lymphoid homeostasis and Th2 devel-opment. Immunity 1998;9:627–35.

11. Oikawa T, Nakamura A, Onishi N, Yamada T, Matsuo K, Saya H.Acquired expression of NFATc1 downregulates E-cadherin and pro-motes cancer cell invasion. Cancer Res 2013;73:5100–9.

12. Horsley V, Aliprantis AO, Polak L, Glimcher LH, Fuchs E. NFATc1balances quiescence and proliferation of skin stem cells. Cell 2008;132:299–310.

13. FougereM, GaudineauB, Barbier J, Guaddachi F, Feugeas JP, AuboeufD, et al. NFAT3 transcription factor inhibits breast cancer cell motilityby targeting the Lipocalin 2 gene. Oncogene 2010;29:2292–301.

14. Singh G, Singh SK, Konig A, Reutlinger K, Nye MD, Adhikary T, et al.Sequential activation of NFAT and c-Myc transcription factors med-iates the TGF-beta switch from a suppressor to a promoter of cancercell proliferation. J Biol Chem 2010;285:27241–50.

15. Smith JJ, Deane NG, Wu F, Merchant NB, Zhang B, Jiang A, et al.Experimentally derived metastasis gene expression profile predictsrecurrence and death in patients with colon cancer. Gastroenterology2010;138:958–68.

16. Shi Z, Derow CK, Zhang B. Co-expression module analysis revealsbiological processes, genomicgain, and regulatorymechanismsasso-ciated with breast cancer progression. BMC Syst Biol 2010;4:74.

17. Zhang B, Kirov S, Snoddy J. WebGestalt: an integrated system forexploring gene sets in various biological contexts. Nucleic Acids Res2005;33:W741–8.

18. Johnson WE, Li C, Rabinovic A. Adjusting batch effects in microarrayexpression data using empirical Bayes methods. Biostatistics2007;8:118–27.

Cancer Res; 74(23) December 1, 2014 Cancer Research6956

Tripathi et al.

Page 11: Nuclear Factor of Activated T-cell Activity Is Associated ... · Molecular and Cellular Pathobiology Nuclear Factor of Activated T-cell Activity Is Associated with Metastatic Capacity

19. Freeman TJ, Smith JJ, Chen X,WashingtonMK, Roland JT,MeansAL,et al. Smad4-mediated signaling inhibits intestinal neoplasia by inhi-biting expression of beta-catenin. Gastroenterology 2012;142:562–71e562.

20. Monticelli S, Rao A. NFAT1 and NFAT2 are positive regulators of IL-4gene transcription. Eur J Immunol 2002;32:2971–8.

21. Ramani AK, Li Z, Hart GT, CarlsonMW,Boutz DR,Marcotte EM. Amapof human protein interactions derived from co-expression of humanmRNAs and their orthologs. Mol Syst Biol 2008;4:180.

22. Barabasi AL, Oltvai ZN. Network biology: understanding the cell'sfunctional organization. Nat Rev Genet 2004;5:101–13.

23. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transi-tion. J Clin Invest 2009;119:1420–8.

24. Zhu J, Wang J, Shi Z, Franklin JL, Deane NG, Coffey RJ, et al.Deciphering genomic alterations in colorectal cancer through tran-scriptional subtype-based network analysis. PLoS ONE 2013;8:e79282.

25. Anastassiou D, Rumjantseva V, ChengW, Huang J, Canoll PD, Yama-shiro DJ, et al. Human cancer cells express Slug-based epithelial-mesenchymal transition gene expression signature obtained in vivo.BMC Cancer 2011;11:529.

26. LobodaA,NebozhynMV,Watters JW,BuserCA, ShawPM,HuangPS,et al. EMT is the dominant program in human colon cancer. BMCMedGenomics 2011;4:9.

27. Oh SC, Park YY, Park ES, Lim JY, Kim SM, Kim SB, et al. Prognosticgene expression signature associated with two molecularly distinctsubtypes of colorectal cancer. Gut 2012;61:1291–8.

28. Grade M, Hormann P, Becker S, Hummon AB, Wangsa D, Varma S,et al. Gene expression profiling reveals a massive, aneuploidy-depen-dent transcriptional deregulation and distinct differences betweenlymph node-negative and lymph node-positive colon carcinomas.Cancer Res 2007;67:41–56.

29. Padua D, Zhang XH, Wang Q, Nadal C, Gerald WL, Gomis RR, et al.TGFbeta primes breast tumors for lung metastasis seeding throughangiopoietin-like 4. Cell 2008;133:66–77.

30. Baumgart S, Glesel E, Singh G, Chen NM, Reutlinger K, Zhang J, et al.Restricted heterochromatin formation links NFATc2 repressor activitywith growth promotion in pancreatic cancer. Gastroenterology 2012;142:388–98 e381–7.

31. Koenig A, Linhart T, SchlengemannK, Reutlinger K,Wegele J, Adler G,et al. NFAT-induced histone acetylation relay switch promotes c-Myc-dependent growth in pancreatic cancer cells. Gastroenterology2010;138:1189–99 e1181–2.

32. Buchholz M, Schatz A, Wagner M, Michl P, Linhart T, Adler G, et al.Overexpression of c-myc in pancreatic cancer caused by ectopicactivation of NFATc1 and the Ca2þ/calcineurin signaling pathway.EMBO J 2006;25:3714–24.

33. GludSZ, SorensenAB,AndrulisM,WangB,KondoE, JessenR, et al. Atumor-suppressor function for NFATc3 in T-cell lymphomagenesis bymurine leukemia virus. Blood 2005;106:3546–52.

34. Medyouf H, Alcalde H, Berthier C, Guillemin MC, dos Santos NR,Janin A, et al. Targeting calcineurin activation as a therapeuticstrategy for T-cell acute lymphoblastic leukemia. Nat Med 2007;13:736–41.

35. Yoeli-LernerM,ChinYR,HansenCK, TokerA. Akt/protein kinase bandglycogen synthase kinase-3beta signaling pathway regulates cellmigration through the NFAT1 transcription factor. Mol Cancer Res2009;7:425–32.

36. Yoeli-Lerner M, Yiu GK, Rabinovitz I, Erhardt P, Jauliac S, Toker A. Aktblocks breast cancer cell motility and invasion through the transcrip-tion factor NFAT. Mol Cell 2005;20:539–550.

37. Flockhart RJ, Armstrong JL, ReynoldsNJ, Lovat PE. NFAT signalling isa novel target of oncogenicBRAF inmetastaticmelanoma.Br JCancer2009;101:1448–55.

www.aacrjournals.org Cancer Res; 74(23) December 1, 2014 6957

NFATc1 Regulates Colon Cancer Metastasis