signature in esophageal cancer Author Manuscript NIH...

19
Periostin, a cell adhesion molecule, facilitates invasion in the tumor microenvironment and annotates a novel tumor invasive signature in esophageal cancer Carmen Z. Michaylira 1,2,4,* , Gabrielle S. Wong 1,2,4,* , Charles G. Miller 1,2,4 , Christie M. Gutierrez 1,2,4 , Hiroshi Nakagawa 1,2,4 , Rachel Hammond 4,5 , Andres J. Klein-Szanto 6 , Ju- Seog Lee 7 , Sang Bae Kim 7 , Meenhard Herlyn 9 , J. Alan Diehl 4,8 , Phyllis Gimotty 4,5 , and Anil K. Rustgi 1,2,3,4 1 Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA, 19104, USA 2 Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA 3 Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA 4 Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA 5 Division of Biostatistics, Center for Clinical Epidemiology and Biostatistics, University of Pennsylvania, Philadelphia, PA, 19104, USA 6 Division of Basic Science, Tumor Cell Biology, Fox Chase Cancer Center, Philadelphia, PA, 19104, USA 7 Department of Systems Biology, MD Anderson Cancer Center, Houston, TX, 77030, USA 8 Wistar Institute, Philadelphia, PA, 19104, USA 9 Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA Abstract Human squamous cell cancers are the most common epithelially derived malignancies. One example is esophageal squamous cell carcinoma (ESCC), which is associated with a high mortality rate (1) that is related to a propensity for invasion and metastasis (2). Here we report that periostin, a highly expressed cell adhesion molecule, is a key component of a novel tumor invasive signature obtained from an organotypic culture model of engineered ESCC. This tumor invasive signature classifies with human ESCC microarrays, underscoring its utility in human cancer. Genetic modulation of periostin promotes tumor cell migration and invasion as revealed in gain of and loss of function experiments. Inhibition of EGFR signaling and restoration of wild-type p53 function were each found to attenuate periostin, suggesting interdependence of two common genetic alterations with periostin function. Collectively, our studies reveal periostin as an important mediator of ESCC tumor invasion and they indicate that organotypic (3D) culture can offer an important tool to discover novel biologic effectors in cancer. Corresponding Author: Anil K. Rustgi, MD, T. Grier Miller Professor of Medicine & Genetics, Chief of Gastroenterology, 600 CRB, University of Pennsylvania, 415 Curie Blvd., Philadelphia, PA 19104, 215-898-0154, FAX: 215-573-5412, [email protected]. * Authorship note: Carmen Z. Michaylira and Gabrielle S. Wong have contributed equally to this work. Disclosure of Potential Conflicts of Interest No potential conflicts of interest were disclosed. NIH Public Access Author Manuscript Cancer Res. Author manuscript; available in PMC 2012 February 7. Published in final edited form as: Cancer Res. 2010 July 1; 70(13): 5281–5292. doi:10.1158/0008-5472.CAN-10-0704. NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript

Transcript of signature in esophageal cancer Author Manuscript NIH...

Page 1: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

Periostin, a cell adhesion molecule, facilitates invasion in thetumor microenvironment and annotates a novel tumor invasivesignature in esophageal cancer

Carmen Z. Michaylira1,2,4,*, Gabrielle S. Wong1,2,4,*, Charles G. Miller1,2,4, Christie M.Gutierrez1,2,4, Hiroshi Nakagawa1,2,4, Rachel Hammond4,5, Andres J. Klein-Szanto6, Ju-Seog Lee7, Sang Bae Kim7, Meenhard Herlyn9, J. Alan Diehl4,8, Phyllis Gimotty4,5, and AnilK. Rustgi1,2,3,4

1Division of Gastroenterology, University of Pennsylvania, Philadelphia, PA, 19104, USA2Department of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA3Department of Genetics, University of Pennsylvania, Philadelphia, PA, 19104, USA4Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, 19104, USA5Division of Biostatistics, Center for Clinical Epidemiology and Biostatistics, University ofPennsylvania, Philadelphia, PA, 19104, USA6Division of Basic Science, Tumor Cell Biology, Fox Chase Cancer Center, Philadelphia, PA,19104, USA7Department of Systems Biology, MD Anderson Cancer Center, Houston, TX, 77030, USA8Wistar Institute, Philadelphia, PA, 19104, USA9Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA, 19104, USA

AbstractHuman squamous cell cancers are the most common epithelially derived malignancies. Oneexample is esophageal squamous cell carcinoma (ESCC), which is associated with a highmortality rate (1) that is related to a propensity for invasion and metastasis (2). Here we report thatperiostin, a highly expressed cell adhesion molecule, is a key component of a novel tumor invasivesignature obtained from an organotypic culture model of engineered ESCC. This tumor invasivesignature classifies with human ESCC microarrays, underscoring its utility in human cancer.Genetic modulation of periostin promotes tumor cell migration and invasion as revealed in gain ofand loss of function experiments. Inhibition of EGFR signaling and restoration of wild-type p53function were each found to attenuate periostin, suggesting interdependence of two commongenetic alterations with periostin function. Collectively, our studies reveal periostin as animportant mediator of ESCC tumor invasion and they indicate that organotypic (3D) culture canoffer an important tool to discover novel biologic effectors in cancer.

Corresponding Author: Anil K. Rustgi, MD, T. Grier Miller Professor of Medicine & Genetics, Chief of Gastroenterology, 600 CRB,University of Pennsylvania, 415 Curie Blvd., Philadelphia, PA 19104, 215-898-0154, FAX: 215-573-5412,[email protected].*Authorship note: Carmen Z. Michaylira and Gabrielle S. Wong have contributed equally to this work.Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

NIH Public AccessAuthor ManuscriptCancer Res. Author manuscript; available in PMC 2012 February 7.

Published in final edited form as:Cancer Res. 2010 July 1; 70(13): 5281–5292. doi:10.1158/0008-5472.CAN-10-0704.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Administrator
Highlight
Administrator
Highlight
Administrator
Highlight
美国细胞修复系统医学中心
Comment on Text
肿瘤治疗,决战「血流动力学」 www.CytoThesis.US
Administrator
Underline
Administrator
Highlight
Page 2: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

Keywordstumor microenvironment; periostin; EGFR; p53

IntroductionDevelopment of ESCC involves a multistep, progressive process starting with increasedesophageal epithelial cell proliferation leading to basal cell hyperplasia, dysplasia,carcinoma in situ and finally, advanced invasive carcinoma (3). Several genetic alterations,such as amplification of the epidermal growth factor receptor (EGFR), dysregulation ofcyclin D1 and somatic mutations in the DNA binding domain of the tumor suppressor p53,are involved in initiation and progression of ESCC (4). While EGFR overexpression andp53 inactivation are key genetic alterations associated with ESCC, how these geneticalterations contribute to ESCC progression remains to be elucidated. Previously, we hadaddressed this by modeling EGFR overexpression and p53 missense mutation (R175H) inprimary human esophageal epithelial cells (EPC2), which have been immortalized byhTERT overexpression (designated as EPC2-hTERT-EGFR-p53R175H cells). EPC2-hTERT-EGFR-p53R175H cells were grown in 3D organotypic culture, resulting in invasion of thesecells into the underlying extracellular matrix (ECM) compared to EPC2-hTERT-EGFR orEPC2-hTERT-p53R175H cells, which did not invade (5). Combined expression of thesegenes also resulted in anchorage-independent growth and in tumor formation in xenograftmodels, which was not observed in control cells overexpressing either EGFR or mutant p53alone (5).

Recent experimental results have provided mounting evidence that altered expression of celladhesion molecules can contribute directly to tumor progression by modulating cellsignaling. Therefore, we sought to identify genes involved in cell adhesion that weredifferentially expressed in invading tumor cells and could shed new insights into processesaffecting tumor progression.

Deriving a novel invasive tumor signature from a gene expression profile analysis ofinvading EPC2-hTERT-EGFR-p53R175H cells in 3D culture and human ESCC tumormicroarrays, we have identified periostin to be the most significantly upregulated genetriggering tumor cell invasion. Periostin (POSTN) is a secreted 90 kDa protein that wasidentified originally as a bone adhesion molecule responsible for differentially recruiting andattaching osteoblasts to the periosteum (6). Periostin is a member of the fasciclin (FAS)family and has a N-terminal signal peptide sequence, a cysteine-rich domain, four internalhomologous repeats and a hydrophilic C-terminal domain (6). These four internal repeatdomains share structural homology with Fasciclin 1, an insect neuronal adhesion protein (6,7) and big-h3, a TGF-β1 inducible gene (7, 8). The high degree of structural and sequencesimilarity with these cell adhesion molecules suggest that periostin has a role in celladhesion and migration. Indeed, not only is its expression induced in human ESCC andmouse tumor xenografts, but more importantly, genetic modulation of periostin also affectstumor invasion in a direct fashion. The novel identification of periostin as an essentialmediator of tumor invasion in the microenvironment has implications upon tumor detectionand therapy.

Materials and MethodsCell culture

Primary esophageal cells (EPC2) were established from surgical specimens of normalesophagus as described previously (9) and immortalized by overexpression of the catalytic

Michaylira et al. Page 2

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Administrator
Underline
Administrator
Highlight
Page 3: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

subunit of telomerase (hTERT) (EPC2-hTERT cells) (10). All cells were maintained inkeratinocyte-SFM medium (KSFM) (Invitrogen) supplemented with 40 mg/ml BPE, 1.0 ng/ml EGF, 100 U/ml penicillin, and 100 mg/ml streptomycin (Full KSFM). Cells were grownat 37 °C in 5% CO2.

Retroviral vectors and cell line generationThe PFB-neo or pBABE-zeo retroviral vectors were used to overexpress human EGFR inEPC2-hTERT cells. Additionally, p53R175H was subcloned into the pBABE-puro or thepBABE-zeo retroviral vectors. Periostin cDNA was used (Open Biosystems) and subclonedinto the pFBneo retroviral vector. Two periostin short hairpin RNAs (shRNA#1 5′-CCCATGGAGAGCCAATTAT-3′and shRNA#2 5′-CTCTGACATCATGACAACAAAT-3′) were used (Open Biosystems). Plasmids weretransfected into Phoenix-Ampho packaging cells (gift of G. Nolan) using the Lipofectamine2000 reagent (Invitrogen). Supernatants of the retroviruses encoding EGFR, mutant p53,periostin over-expression and shRNA vector constructs were collected 48 hrs. and 72 hrs.after transfection. Sub-confluent EPC-hTERT cells were infected with retrovirus supernatantin 4mg/ml polybrene (Sigma). 48 hours after infection, cells were selected in 300 μg/mlG418, 0.5 μg/ml puromycin, 2 mg/ml zeocin or 5 mg/ml blastocidin for 5 days.Overexpression of EGFR, mutant p53 and periostin, as well as periostin knock-down wasconfirmed by Western blot analysis.

Organotypic cultureKeratinocytes were grown in organotypic culture to recreate their microenvironment bysupplying extracellular matrix components including collagen and laminin and fetalesophageal fibroblasts, its detailed procedures were described previously (5). Cells grown inorganotypic culture were processed for histology by fixing in 10% formaldehyde andparaffin embedded for LCM studies by embedding in OCT medium followed by freezing inliquid nitrogen and then stored at −80 °C.

LCM and RNA isolationFrozen organotypic cultures were sectioned to 8 μm onto membrane mounted metal frameslides (MMI, Switzerland) using a Microm HM 505E cryostat (Richard Allen Scientific).Sections were immediately fixed, stained and dehydrated before laser microdissection.Microdissection was performed with a Nikon Eclipse TE 2000-5 microscope with a UVlaser (MMI, Switzerland). Microdissected cells were collected and 50 ul of extraction buffer(Arcturus) was added and incubated for 30 min at 42 °C, followed by centrifugation at 800 xg for 2 minutes. RNA was extracted using the Arcturus PicoPure RNA isolation kitfollowing the manufacturer’s instructions. The integrity and concentration of the RNA wasassessed using the Agilent Bioanalyzer 2100 (Agilent Technologies).

RNA amplification and microarray studiesThe RNA obtained from the LCM studies was amplified using the Affymetrix GeneChipExpression 3′-Amplification Two-Cycle cDNA Synthesis Kit (Affymetrix) followed by theAffymetrix GeneChip Hybridization Wash and Stain kit (Affymetrix). The resulting cRNA(200 ng) was used as template for random-primed cDNA synthesis and a second round of invitro transcription, which incorporated biotinylated CTP and UTP. The cRNA products werefragmented to 200 bp or less, heated at 99°C for 5 min, and hybridized for 16 h at 45°C toAffymetrix U133Plus 2.0 oligonucleotide microarrays (Affymetrix). Microarrays weresubsequently washed at low (6 x SSPE) and high (100 mM MES, 0.1 M NaCl) stringencyand stained with streptavidin-phycoerythrin. The fluorescence signal was amplified byaddition of biotinylated anti-streptavidin, and an additional aliquot of streptavidin-

Michaylira et al. Page 3

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Administrator
Highlight
Administrator
Highlight
Administrator
Highlight
Administrator
Highlight
Administrator
Highlight
Page 4: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

phycoerythrin stain. A confocal scanner was used to acquire the fluorescent signal afterexcitation (570 nm).

qPCRLCM was repeated to isolate invading and non-invading EPC-hTERT-EGFR-p53R175H cellsgrown in organotypic culture. Amplification and cDNA synthesis was performed using WT-Ovation RNA Amplification System (NuGen Technologies) according to manufacturer’sinstructions. Real-Time PCR was performed and analyzed using ABI PRISM 7000 sequencedetection system software (PE Applied Biosystems) and using Power SYBR Green PCRMaster Mix (PE Applied Biosystems) for β-actin according to the manufacturer’sinstructions. Taqman assays for periostin were run using Taqman Universal PCR MasterMix (PE Applied Biosystems) according to the manufacturer’s instructions.

Tumor specimensEsophageal tumor tissue specimens and adjacent normal tissue were surgically procuredfrom patients at the Okayama University Hospital (Drs. Shirakawa and Naomoto, Japan).All tumor specimens were pathologically diagnosed as esophageal squamous cellcarcinomas (Grade III) and obtained from informed-consent patients in accordance withInstitutional Review Board standards and guidelines.

Specimens were immediately snap-frozen for RNA and protein analyses. The human ESCCtissue microarray was subjected to immunohistochemistry analysis using a polyclonal anti-periostin antibody and scored for periostin expression as follows; Negative (0), Marginalconsidered negative (0.5), mild positive stain (1), moderate positive stain (2), and intensepositive stain (3). Scores >0.5 are considered positive. Each case on the tissue microarraycomprises of 2 cores and the mean scores of 2 cores were taken.

AntibodiesThe following antibodies were used for immunoblotting: EGFR (NeoMarkers, Ab-12), p53(Oncogene Research Products, Ab-6), phospho-EGFR (Cell Signaling, Tyr 1068), rabbitpolyclonal periostin (Abcam, ab 14041), p21 (Oncogene Research Products), WAF1 (ab-1),β-actin (Sigma) was used as a loading control. For immunohistochemistry: periostin(Abcam, ab 14041).

Western blottingTo confirm protein expression, western blot analysis was performed as described previously(5). To confirm secreted periostin expression, conditioned media was collected and proteinconcentration was determined by Bio-Rad protein assay (Bio-Rad) and Western blotanalysis was performed as described above.

Migration and invasion assaysMigration and invasion assays were performed as described previously (5). All experimentswere performed at least three times in triplicate.

Data ProcessingAffymetrix array cel files were processed in R using an algorithm that uses genes in the leastvariant set (LVS) to normalize the expression data (11). In the analysis of invasive and non-invasive cells, we then identified 6106 probe sets for subsequent statistical analysis that hada minimum expression level (expression greater than 100 in all samples) and that hadsignificant variability among the 11 cell lines (standard deviation was greater than 150). Thebackground correction, summarization and normalization of Affymetrix cel files were done

Michaylira et al. Page 4

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 5: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

in the same manner independently for each of the two tumor/normal studies. GeneExpression Omnibus (GEO) database accession number: GSE21293

Statistical analysisAll statistical analyses used natural log transformed expression data. Using the arrays ofcells from the organotypic culture model, a multivariate regression model was fit for eachgene that included two independent binary variables. The first variable indicated cells withboth EGFR overexpression and P53 mutation only and the second indicated cells withEGFR overexpression, P53 mutation and invasion. The third group (the reference group)included cells with a non-invading phenotype that had either EGFR overexpression or P53mutation. The p-values from the test that the regression model was significant (F-statistic,2df) were adjusted using the Benjamini-Hochberg technique; probe sets with a FalseDiscovery Rate (FDR) < 0.05 were selected for further investigation (12). Genes on this listwere compared to annotated Gene Ontology biologic processes and a functional annotationcluster analysis was done using the online DAVID tool (http://niaid.abcc.ncifcrf.gov/).Annotations were derived from the DAVID databases. Supervised hierarchical clusteringused in the heat maps was based on Euclidean distance. The statistical analyses were doneusing SAS/STAT software version 9.2 of the SAS System for PC.

ResultsIdentification of periostin from tumor invasion signature derived from gene expressionprofile analysis on invading EPC2-hTERT-EGFR-p53R175H cells and ESCC tumormicroarrays

The molecular pathways and networks underlying tumor invasion remain largely to beelucidated. In order to characterize a gene expression profile underlying the invasivephenotype of invading EPC2-hTERT-EGFR-p53R175H cells, we profiled mRNA obtainedfrom laser capture microdissected EPC-hTERT cells overexpressing EGFR and with a p53missense mutation (R175H) that was either invasive or non-invasive in the organotypicculture model, as well as non-invasive cells that either overexpressed EGFR alone or hadp53 (R175H) mutation alone, using Affymetrix U133-2 plus microarrays.

Analysis of the gene expression microarray data revealed a tumor invasive signaturecorresponding to a probe set list (PSL) of 939 probesets, where there was significantdifferential expression of these probe sets in invasive versus non-invasive cells(Supplementary Table 1). A supervised hierarchical cluster analysis demonstrated that thistumor invasive signature could distinguish significant differences between cell lines inorganotypic culture with and without an invasive phenotype as presented in a heat map(Figure 1A).

To determine whether this invasive signature derived from invading EPC2-hTERT-EGFR-p53R175H cells grown in organotypic culture can be extended to human ESCC, we analyzedgene expression profiles from five paired human ESCCs. We found that overall geneexpression patters of invasive cells grown in organotypic culture are highly similar to thosefrom ESCC tumors and this tumor invasive signature was able to discriminate esophagealtumors from adjacent normal tissue and classified all tumors and 80% of the normal samplescorrectly (Figure 1B). To validate this tumor invasive signature, we performed a secondaryanalysis on an independent cohort of five paired human ESCC tumors using this tumorinvasive signature and observed that it was capable of classifying 80% of ESCC tumors andall adjacent normal tissues correctly (Figure 1C). Using the t-test statistic to rank theprobesets in this tumor invasive signature, we identified periostin, a cell adhesion molecule,with the most significant difference in gene expression between invading transformed

Michaylira et al. Page 5

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 6: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

esophageal epithelial cells compared to non-invading normal or dysplastic epithelial cells (t-test for two independent groups, p<0.0001). Strikingly, periostin was also found to besignificantly upregulated in both independent cohorts of human ESCC tumors compared toadjacent normal tissue, the fold change in mean periostin expression was found to be 5.0 and5.7 in the respective cohorts.

To explore the biological processes associated with invasion of ESCC, we tested forenrichment of gene ontology (GO) terms within this tumor invasive signature. This tumorinvasive signature was found to be significantly enriched in nine biological processes (FDR,p<0.01) (data not shown, Supplementary Table S1 and S2). To investigate the cellularprocesses altered within the tumor microenvironment, we selected three biological processesfor further investigation and observed that probesets clustered in each biological process(cell/biological adhesion, development-related and cellular component organization andbiogenesis) were able to classify invading transformed esophageal epithelial cells from non-invading cells (Table 1 and Supplementary Figure S1A-C). Interestingly, periostin wasfound to be the highest upregulated gene (6.54 fold) across the three biological processes(Table 1), and was unique in this feature amongst all the genes in the datasets. In aggregate,these results reveal a tumor invasive signature derived from the organotypic culture modelwhich is applies faithfully to human ESCC and highlights periostin as an important generequired for facilitating tumor cell invasion.

Periostin is expressed preferentially in invading EPC2-hTERT-EGFR-p53R175H cells both invitro and in vivo

We next sought to validate whether the upregulation of periostin is concomitant withinvading transformed esophageal cells as well as establish the localization of periostinexpression in these cells. Consistent with the results of the microarray analysis, increasedperiostin mRNA expression in invading EPC2-hTERT-EGFR-p53R175H cells was confirmedby LCM isolation of invading and non-invading EPC2-hTERT-EGFR-p53R175H cells in theorganotypic culture model followed by RNA isolation, amplification and qPCR analysis(Figure 2A). Furthermore, periostin protein accumulation in invading EPC2-hTERT-EGFR-p53R175H cells grown in organotypic culture was observed by immunohistochemicalstaining (Figure 2B, Right panel b), but not in control EPC2-hTERT-EGFR cells (Figure 2B,Left panel a). Interestingly, periostin protein expression was also observed in the epithelial-stromal interface in tumors formed by EPC2-hTERT-EGFR-p53R175H cells in a xenografttumor model (Figure 2C, white arrows). These results establish preferential expression ofperiostin in invading EPC2-hTERT-EGFR-p53R175H cells both in vitro and in vivo.

Loss of periostin leads to decreased migration and invasion in EPC2-hTERT-EGFR-p53R175H cells

Previous efforts involving other epithelial cancers had indicated that periostin promotesinvasion and anchorage-independent growth in several epithelial cancer cell lines and intumors such as head and neck cell carcinoma, oral, breast and ovarian cancers (13-16).Elevated levels of periostin have been detected in sera of patients with breast, thymoma andnon-small cell lung cancer, suggesting periostin secretion during tumorigenesis (17-19). Wesought to investigate directly whether periostin had a functional role in facilitating tumorcell migration and invasion in ESCC. An RNA interference approach with shRNA was usedto induce stable knockdown of periostin expression in EPC2-hTERT-EGFR-p53R175H cells(Figure 3A). Knockdown of periostin in EPC2-hTERT-EGFR-p53R175H cells led tosignificant decrease in cell migration and invasion compared to control shscrambled cells(Figure 3B, C). Notably, reduced periostin expression in EPC2-hTERT-EGFR-p53R175H

cells also resulted in decreased invasion of these cells into the underlying matrix inorganotypic culture (Figure 3D).

Michaylira et al. Page 6

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 7: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

Periostin overexpression promotes increased migration and invasion in EPC2-hTERT-EGFR-p53R175H cells

In parallel studies, periostin was retrovirally overexpressed in two independent cell lines(EPC2-hTERT-EGFR-p53R175H-1 and EPC2-hTERT-EGFR-p53R175H-2) (Figure 4A) andwhile overexpression of periostin showed no effect on proliferation (data not shown), weobserved that these cells displayed increased migration and invasion (Figure 4B, C).Furthermore, when EPC2-hTERT-EGFR-p53R175H cells overexpressing periostin weregrown in organotypic culture, increased invasion into the underlying matrix was alsoobserved (Figure 4D). Collectively, these results demonstrate the direct functional role ofperiostin in promoting tumor cell migration and invasion, particularly within the context ofthe tumor microenvironment.

Upregulation of periostin expression in primary ESCC tumors and in a cancer tissuemicroarray

We next sought to determine if upregulation of periostin is also found in human ESCC byassessing periostin expression in primary ESCC (Grade III) tumors. Increased periostinmRNA expression was observed in ESCC tumors compared to their matched normalcontrols (Figure 5A). A marked increase in periostin protein levels was observed in allESCC tumors studied compared with their matched normal mucosa controls (Figure 5B andSupplementary Figure S2). To determine the localization of periostin in human ESCC,further immunohistochemical analysis of periostin expression was performed in a tissuemicroarray containing 73 ESCC tumors and adjacent normal tissue and revealed periostinexpression in invasive ESCC tumor cells (Figure 5C, Bottom panel c, arrowheads) andconsistent accumulation in tumor stroma (Figure 5C, Bottom panel c, arrows) compared tonormal esophageal tissue where periostin expression was detected primarily in and aroundblood vessels (Figure 5C, Left panel a, arrows). Periostin expression was also detected inhigh grade esophageal intraepithelial neoplasia (EIN), which was found in a restrictednumber of cases, particularly in the epithelial-stromal interface (Figure 5C, Right panel b,arrows). In addition, the stroma in all 73 ESCC cases were scored for periostinimmunohistochemical staining intensity and it was shown to have higher levels of periostinstaining compared to matched normal esophagus which showed weak to no periostinstaining. Specifically, the ESCC scored either 2 or 3, meaning moderate or intense staining,whereas the normal esophagus was 0 or 0.5 (Figure 5C, panel d), which would suggest thatinduction of periostin could also arise in the stroma during ESCC progression.

Induction of periostin is dependent upon EGFR signaling and p53 mutationIt is believed, but not established, that periostin might enhance tumor cell invasion andmetastasis through increased integrin signaling, augmenting cell survival through the PI3K/Akt pathway or fostering epithelial-to-mesenchymal transition (EMT)(16, 20, 21). Giventhat periostin was identified initially in invading EPC-hTERT-EGFR-p53R175H cells throughfunctional genomics and bioinformatic analysis, we hypothesized that periostin expression isdependent upon both activated EGFR signaling and p53 mutation. Notably, periostinexpression was upregulated in EPC2-hTERT-EGFR-p53R175H cells compared to controlcells overexpressing EGFR alone (EPC2-hTERT-EGFR) or mutant p53 alone (EPC2-hTERT-p53R175H) (Figure 6A). In addition, periostin promoter reporter assays in EPC2-hTERT-EGFR-p53R175H cells displayed the highest promoter activity compared to controlcells with either EGFR overexpression or p53 mutation alone (Supplementary Figure S3),suggesting that both genetic alterations may be required to activate periostin expression at atranscriptional level. This result was further corroborated when EPC2-hTERT-EGFR-p53R175H cells were stimulated with EGF and increased periostin protein expression wasobserved (Figure 6B). This induction of periostin upon EGFR stimulation led us to testwhether inhibition of EGFR signaling and/or restoration of wild-type p53 function could

Michaylira et al. Page 7

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 8: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

inhibit periostin by treating EPC2-hTERT-EGFR-p53R175H cells. We employed AG1478, aEGFR tyrosine kinase inhibitor, and 5-iminodaunorubicin, a small molecule compoundwhich restores wild-type p53 signaling by inducing apoptosis and cell cycle arrest, asillustrated by induction of p21(22). Periostin protein expression was noted to be decreasedwhen inhibited by AG1478 or 5-iminodaunorubicin or both (Figure 6B). Taken together,these data support the notion that periostin expression is modulated mechanistically byactivated EGFR signaling and p53 mutation.

DiscussionOverall, our results reveal a novel tumor invasive signature derived from invadingtransformed cells compared to control noninvading normal and dysplastic cells inorganotypic (3D) culture. This signature is one that annotates primary invasive humanesophageal squamous cell cancer as distinctive from adjacent normal human esophagealmucosa in two independent cohorts of tumors, underscoring the fidelity and utility of thistumor invasive signature. It is conceivable that this molecular signature might beinformative in the future for other squamous cell cancers arising in different tissues.

Gene ontology analysis (DAVID databases) of specific biological processes that arebelieved to be involved in the tumor microenvironment reveals the consistent and uniqueupregulation of periostin, suggesting its critical functional role. First, periostin was found tobe upregulated in invading transformed cells and in primary esophageal squamous cellcancers based upon immunohistochemical and Western blot analysis. Second, periostin’sdirect functional role is underscored by overexpression and knockdown experiments inwhich genetic manipulation of periostin dramatically influences the degree of tumorinvasion in organotypic culture. Third, EGFR signaling and p53 mutation, both canonicalgenetic alterations in ESCC, appear to converge upon periostin based upon luciferasereporter gene assays as well as inhibition of EGFR signaling and restoration of wild typep53 function. In aggregate, these novel results underscore the utility of the organotypicculture model for the discovery of direct biological effectors of tumor invasion into themicroenvironment, which has been largely elusive to date. Local tumor invasion in themesenchymal stromal compartment is important given that it temporally precedes tumordissemination in the lymphatic and blood vessels for tumor metastasis.

Various avenues of investigation have highlighted the important role of themicroenvironment in enhancing the initial dissemination of malignant tumor cells. Dynamicinteractions between the epithelium and mesenchymal stroma contribute to boosting theinvasive phenotype of tumor cells by activating a variety of genes facilitating cellproliferation, de-differentiation, migration and invasion (23, 24). Significant changes such asloss of cell-cell contacts, disruptions in cell adhesion junctions and altered cell-extracellularmatrix interactions within the tumor microenvironment converge to increase the ultimatemetastatic potential of tumor cells (25). Therefore, identification of gene expression patternchanges during initial stages of tumor progression within the tumor microenvironment iscrucial to understanding the causes of tumor invasion, and ultimately, tumor metastasis todistant organ sites. Thus, our results provide new platforms into the investigation of tumorinvasion.

Periostin has been shown also to have a role in bone, tooth and heart formation duringdevelopment (26, 27) and is only re-expressed and upregulated in adult tissue after vascular,skeletal or bone injuries (28, 29).Periostin is similarly overexpressed in human cancers (14),(30). Our results highlight that a tumor invasive signature defines genetically engineeredESCC that is reproduced in human ESCC. Our studies also suggest that induction ofperiostin, a secreted protein with a long half-life (Supplementary Figure S4 and data not

Michaylira et al. Page 8

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 9: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

shown), may alter the tumor microenvironment by accumulating in the stroma andfacilitating invasion through matrix remodeling. This may be achieved through regulation ofcollagen I fibrillogenesis (31), serving as a bridge between tenascin C and the extracellularmatrix, as well as through interaction with αVβ3 integrins. Furthermore, periostin maypromote tumor cell survival in the matrix of the microenvironment by activating the Akt/PI3K pathway. Lack of periostin may lead to suppression of Notch1 signaling (32),conversely, it is conceivable that overexpression of periostin could activate Notch1 signalingin cancers. Indeed, we have evidence of activated Notch signaling in invasive tumor cellsgrown in organotypic culture (data not shown). Future translational and clinicalinvestigations might seek to exploit periostin as an attractive therapeutic target, especially ina combinatorial fashion, for example, with inhibitors of receptor tryosine kinases.

Supplementary MaterialRefer to Web version on PubMed Central for supplementary material.

AcknowledgmentsThis work was supported by NIH/NCI P01-CA098101 (CZM, GSW, AKS, JAD, MH, HN, RH, PG, AKR), NIH/NRSA F32DK-075230 (CZM), NIH T32-CA115299 (GSW) and NIH/NIDDK Center for Molecular Studies inDigestive and Liver Diseases (P30-DK050306). We acknowledge the Morphology Core (S. Mitchell, D. Budo, G.Swain) and Molecular Biology/Gene Expression Core (G. Wu, S. Keilbaugh). We are grateful to other members ofthe Rustgi lab for helpful discussions.

References1. Parkin DM,BF, Ferlay J, et al. Global cancer statistics, 2002. CA Cancer J Clin. 2005:74–108.

[PubMed: 15761078]2. Nair KS, Naidoo R, Chetty R. Expression of cell adhesion molecules in oesophageal carcinoma and

its prognostic value. J Clin Pathol. 2005; 58:343–351. [PubMed: 15790695]3. Lehrbach DM, Nita ME, Cecconello I. Molecular aspects of esophageal squamous cell carcinoma

carcinogenesis. Arq Gastroenterol. 2003; 40:256–261. [PubMed: 15264049]4. Xu XC. Risk factors and gene expression in esophageal cancer. Methods Mol Biol. 2009; 471:335–

360. [PubMed: 19109788]5. Okawa T, Michaylira CZ, Kalabis J, et al. The functional interplay between EGFR overexpression,

hTERT activation, and p53 mutation in esophageal epithelial cells with activation of stromalfibroblasts induces tumor development, invasion, and differentiation. Genes Dev. 2007; 21:2788–2803. [PubMed: 17974918]

6. Takeshita S, Kikuno R, Tezuka K, Amann E. Osteoblast-specific factor 2: cloning of a putative boneadhesion protein with homology with the insect protein fasciclin I. Biochem J. 1993; 294(Pt 1):271–278. [PubMed: 8363580]

7. Horiuchi K, Amizuka N, Takeshita S, et al. Identification and characterization of a novel protein,periostin, with restricted expression to periosteum and periodontal ligament and increasedexpression by transforming growth factor beta. J Bone Miner Res. 1999; 14:1239–1249. [PubMed:10404027]

8. Skonier J, Neubauer M, Madisen L, Bennett K, Plowman GD, Purchio AF. cDNA cloning andsequence analysis of beta ig-h3, a novel gene induced in a human adenocarcinoma cell line aftertreatment with transforming growth factor-beta. DNA Cell Biol. 1992; 11:511–522. [PubMed:1388724]

9. Andl CD, Mizushima T, Nakagawa H, et al. Epidermal growth factor receptor mediates increasedcell proliferation, migration, and aggregation in esophageal keratinocytes in vitro and in vivo. J BiolChem. 2003; 278:1824–1830. [PubMed: 12435727]

10. Harada H, Nakagawa H, Oyama K, et al. Telomerase induces immortalization of humanesophageal keratinocytes without p16INK4a inactivation. Mol Cancer Res. 2003; 1:729–738.[PubMed: 12939398]

Michaylira et al. Page 9

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 10: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

11. Calza S,VD, Pawitan Y. Normalization of oligonucleotide arrays based on the least-variant set ofgenes. BMC Bioinformatics. 2008; 9

12. Benjamini Y HY. Controlling the False Discovery Rate: A Practical and Powerful Approach toMultiple Testing. JRRSB. 1995; 57:289–300.

13. Kudo Y, Ogawa I, Kitajima S, et al. Periostin promotes invasion and anchorage-independentgrowth in the metastatic process of head and neck cancer. Cancer Res. 2006; 66:6928–6935.[PubMed: 16849536]

14. Siriwardena BS, Kudo Y, Ogawa I, et al. Periostin is frequently overexpressed and enhancesinvasion and angiogenesis in oral cancer. Br J Cancer. 2006; 95:1396–1403. [PubMed: 17060937]

15. Puglisi F, Puppin C, Pegolo E, et al. Expression of periostin in human breast cancer. J Clin Pathol.2008; 61:494–498. [PubMed: 17938160]

16. Gillan L, Matei D, Fishman DA, Gerbin CS, Karlan BY, Chang DD. Periostin secreted byepithelial ovarian carcinoma is a ligand for alpha(V)beta(3) and alpha(V)beta(5) integrins andpromotes cell motility. Cancer Res. 2002; 62:5358–5364. [PubMed: 12235007]

17. Sasaki H, Yu CY, Dai M, et al. Elevated serum periostin levels in patients with bone metastasesfrom breast but not lung cancer. Breast Cancer Res Treat. 2003; 77:245–252. [PubMed: 12602924]

18. Sasaki H, Dai M, Auclair D, et al. Serum level of the periostin, a homologue of an insect celladhesion molecule, as a prognostic marker in nonsmall cell lung carcinomas. Cancer. 2001;92:843–848. [PubMed: 11550156]

19. Sasaki H, Dai M, Auclair D, et al. Serum level of the periostin, a homologue of an insect celladhesion molecule, in thymoma patients. Cancer Lett. 2001; 172:37–42. [PubMed: 11595127]

20. Bao S, Ouyang G, Bai X, et al. Periostin potently promotes metastatic growth of colon cancer byaugmenting cell survival via the Akt/PKB pathway. Cancer Cell. 2004; 5:329–339. [PubMed:15093540]

21. Yan W, Shao R. Transduction of a Mesenchyme-specific gene Periostin into 293T cells inducescell invasive activity through epithelial-mesenchymal transformation. J Bio Chem. 2006;28:19700–19708. [PubMed: 16702213]

22. Wang W, Kim SH, El-Deiry WS. Small-molecule modulators of p53 family signaling andantitumor effects in p53-deficient human colon tumor xenografts. Proc Natl Acad Sci U S A.2006; 103:11003–11008. [PubMed: 16835297]

23. Mueller MM, Fusenig NE. Friends or foes - bipolar effects of the tumour stroma in cancer. NatRev Cancer. 2004; 4:839–849. [PubMed: 15516957]

24. Joyce JA, Pollard JW. Microenvironmental regulation of metastasis. Nat Rev Cancer. 2009; 9:239–252. [PubMed: 19279573]

25. Cavallaro U, Christofori G. Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. NatRev Cancer. 2004; 4:118–132. [PubMed: 14964308]

26. Rios H, Koushik SV, Wang H, et al. Periostin null mice exhibit dwarfism, incisor enamel defects,and an early-onset periodontal disease-like phenotype. Mol Cell Biol. 2005; 25:11131–11144.[PubMed: 16314533]

27. Butcher JT, Norris RA, Hoffman S, Mjaatvedt CH, Markwald RR. Periostin promotesatrioventricular mesenchyme matrix invasion and remodeling mediated by integrin signalingthrough Rho/PI 3-kinase. Dev Biol. 2007; 302:256–266. [PubMed: 17070513]

28. Nakazawa T, Nakajima A, Seki N, et al. Gene expression of periostin in the early stage of fracturehealing detected by cDNA microarray analysis. J Orthop Res. 2004; 22:520–525. [PubMed:15099630]

29. Lindner V, Wang Q, Conley BA, Friesel RE, Vary CP. Vascular injury induces expression ofperiostin: implications for vascular cell differentiation and migration. Arterioscler Thromb VascBiol. 2005; 25:77–83. [PubMed: 15514205]

30. Baril P, Gangeswaran R, Mahon PC, et al. Periostin promotes invasiveness and resistance ofpancreatic cancer cells to hypoxia-induced cell death: role of the beta4 integrin and the PI3kpathway. Oncogene. 2007; 26:2082–2094. [PubMed: 17043657]

31. Norris RA, Damon B, Mironov V, et al. Periostin regulates collagen fibrillogenesis and thebiomechanical properties of connective tissues. J Cell Biochem. 2007; 101:695–711. [PubMed:17226767]

Michaylira et al. Page 10

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 11: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

32. Tkatchenko TV, Moreno-Rodriguez RA, Conway SJ, Molkentin JD, Markwald RR, TkatchenkoAV. Lack of periostin leads to suppression of Notch1 signaling and calcific aortic valve disease.Physiol Genomics. 2009; 39:160–168. [PubMed: 19723774]

Michaylira et al. Page 11

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 12: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

Figure 1. Invasive signature from EPC2-hTERT-EGFR-p53R175H cells classifies with humanESCC(A) Microarray analysis of LCM-extracted RNA from invading EPC2-hTERT-EGFR-p53R175H cells grown in organotypic culture (n=3) compared to non-invading EPC2-hTERT-EGFR-p53R175H (n=3) as well as non-invading EPC2-hTERT-EGFR-puro (n=3)and EPC2-hTERT-neop53R175H cells (n=2). Differentially expressed probesets weresubjected to supervised hierarchical clustering and expression is based on a log2 scale wherered represents upregulation and green represents downregulation. Heatmap denotes probesetlist (PSL) of 939 probesets representing unique tumor invasive signature characterizinginvasive phenotype. Tumor invasive signature comprises differentially expressed probesets(p<0.0001).(B and C) Heatmap representation of gene expressing profiles from two independent cohortseach comprising of five paired ESCC tumors classified using the tumor invasive signaturefrom (A). Due to microarray platform difference (U133 v2.0 and U133A) used in 2independent cohorts, only 648 probesets were shared in both platforms. All probesets are inthe same order as seen in (A).

Michaylira et al. Page 12

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 13: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

Figure 2. Periostin is expressed preferentially in invading EPC2-hTERT-EGFR-p53R175H cells(A) Fold change of periostin expression in invading versus non-invading EPC2-hTERT-EGFR-p53R175H cells isolated by LCM from organotypic culture and quantified by qPCR.(*) P-value < 0.05(B) Immunohistochemistry analysis of periostin expression in control EPC2-hTERT-EGFRcells (Left panel a) and invading EPC2-hTERT-EGFR-p53R175H cells (Right panel b) grownin organotypic culture (OTC). Rabbit polyclonal periostin antibody recognizes exogenousand secreted periostin.(C) Periostin expression by immunohistochemistry of tumor formed in vivo by EPC2-hTERT-EGFR-p53R175H cells. Periostin expression in tumor epithelial-stromal interfaceindicated (white arrows). (200x Magnification)

Michaylira et al. Page 13

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 14: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

Figure 3. Periostin knockdown in EPC2-hTERT-EGFR-p53R175H cells reduces migration andinvasion(A) Western blot confirming periostin (90 kDal) knock-down in EPC2-hTERT-EGFR-p53R175H cells using two independent shRNA contructs.(B) Representative fluorescent images obtained from bottom filter of a Boyden chambermigration assay show reduced migration in EPC2-hTERT-EGFR-p53R175H cells expressingshRNA to periostin versus control (scrambled) shRNA to periostin. Experiments wereperformed in triplicate. (*) P<0.04 for shPOSTN#1 vs shscrambled, (*) P<0.001 forshPOSTN#2 vs shscrambled.(C) Images and quantifications of the Matrigel invasion assay were acquired and processedas in (B). Experiments were performed in triplicate. (*) P < 0.02 for shPOSTN#1 vsshscrambled, (*) P<0.05 for shPOSTN#2(D) H & E staining of organotypic culture comparing shRNA to periostin versus scrambledshRNA showing decreased invasion of EPC2-hTERT-EGFR-p53R175H cells expressingshRNA to periostin. Bar graphs represent fold change in invasion +/− SEM, P=0.015 (200xmagnification)

Michaylira et al. Page 14

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 15: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

Figure 4. Periostin overexpression in EPC2-hTERT-EGFR-p53R175H cells promotes increasedmigration and invasion in vitro and in organotypic culture(A) Western blot confirming periostin (90 kDal) overexpression in two independent EPC2-hTERT-EGFR-p53R175H cell lines. pFB neo was used as an empty control vector.(B) Representative fluorescent images obtained from bottom filter of a Boyden chambermigration assay show enhanced migration in EPC2-hTERT-EGFR-p53R175H cells thatoverexpress periostin versus control EPC2-hTERT-EGFR-p53R175H-neo cells. Bar graphsrepresent fold changes +/− SEM (*) P <0.01 (Student t-test, EPC2-hTERT-EGFR-p53R175H

periostin overexpressing cells vs. control cells). Note that P < 0.05 is statistically significant.Experiments were done in triplicate(C) Images and quantifications of the Matrigel invasion assay were acquired and processedas in (B). (*) P<0.04(D) H & E staining of organotypic cultures comparing EPC2-hTERT-EGFR-p53R175H

periostin overexpressing cells to empty vector control cells reveal increased invasion ofEPC2-hTERT-EGFR-p53R175H periostin overexpressing cells. Bar graphs represent foldchange in invasion +/− SEM, (*) P< 0.03 (EPC2-hTERT-EGFR-p53R175H periostinoverexpressing cells vs empty control vector cells). (200x magnification)

Michaylira et al. Page 15

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 16: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

Figure 5. Periostin is overexpressed in primary ESCC tumors and is associated with ESCCtumor progression(A) Relative periostin mRNA expression measured by real-time PCR in 5 primary ESCCtumor specimens with paired adjacent non-invasive esophageal tissue. (*) P-value<0.05.(B) Western blot analysis of periostin expression in 5 primary ESCC tumor specimens withpaired adjacent non-invasive esophageal tissue. Expression of β-actin was used as aninternal loading control.(C) Immunohistological analysis of a tissue microarray comprising of 73 human ESCCcases with normal esophageal tissue. Representative photomicrographs of periostinexpression in normal esophageal tissue, high grade esophageal intraepithelial neoplasia(EIN) and ESCC. Periostin expression was observed around blood vessels (Left panel a,arrows). Periostin staining observed in EIN epithelial-stroma interface (Right panel b,arrows) and accumulation in tumor stroma (Bottom panel c, arrows) and tumor cells(Bottom panel c, arrowheads). (400x magnification). Stroma in tissue microarray was scoredfor periostin staining intensity and average score of 2 cores from each case were taken.ESCC cases scored higher levels of periostin expression (n=73, Mean= 1.44, s.d +/− = 0.97)compared to matched normal controls (n=69, Mean= 0.44, s.d +/− = 0.68) (panel d). Closedcircles represent outliers. Student’s t-test, ESCC cases vs matched normal esophaguscontrols. (**) p-value < 0.01

Michaylira et al. Page 16

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 17: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

Figure 6. Periostin expression is dependent upon EGFR signaling and mutant p53 activation(A) Western blot analysis of periostin (90kDa) in EPC2-hTERT-EGFR-p53R175H, EPC2-hTERT-puro-neo, EPC2-hTERT-EGFR-neo and EPC2-hTERT-neo-p53R175H cells.(B) Western blot analysis of periostin expression in EPC2-hTERT-EGFR-p53R175H celllysates after 24h treatment with EGF (10ng/μl), EGFR inhibitior AG1478 (1 μM) and 5-iminodaunorubicin (3 μM). Immunoblotting for total EGFR and phosphorylated EGFR toconfirm inhibition of EGFR as well as p21 to indicate restoration of wildtype p53 signaling.β-actin was used as a loading control. Densitometry ratios of periostin/β-actin werecalculated and recorded.

Michaylira et al. Page 17

Cancer Res. Author manuscript; available in PMC 2012 February 7.

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Page 18: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Michaylira et al. Page 18

Tabl

e 1

Gen

es w

ith a

t lea

st tw

o-fo

ld h

ighe

r ex

pres

sion

in in

vasi

ve E

PC2.

hTE

RT

.EG

FR.p

53R

175H

cel

l cel

l lin

es c

ompa

red

to n

on-in

vasi

veE

PC2.

hTE

RT

.EG

FR.p

53R

175H

/EPC

2.hT

ER

T.E

GFR

.pur

o/E

PC2.

hTE

RT

.ueo

.p53

R17

5H c

ell l

ines

by

GO

Bio

logi

cal P

roce

sses

Cel

lula

r C

ompo

nent

Org

aniz

atio

n an

dB

ioge

nesi

sB

iolo

gica

l Adh

esio

n an

dC

ellu

lar

Adh

esio

nD

evel

opm

enta

l Pro

ces

GO

:001

6043

GO

:002

2610

& G

O00

0715

5G

O:0

0488

56,0

0325

02,0

0487

31,

0009

888,

0048

513

&00

0727

5

Prob

e Se

tG

ene

Nam

eR

atio

*Pr

obe

Set

Gen

e N

ame

Rat

io*

Prob

e Se

tG

ene

Nam

eR

atio

*

2108

09_s

_at

POST

N6.

5421

0809

_s_a

tPO

STN

6.54

2108

09_s

_at

POST

N6.

54

2119

64_a

tC

OL4

A2

4.28

2262

37_a

tC

OL8

A1

5.43

2063

00_s

_at

PTH

LH4.

01

2162

50_s

_at

LPX

N4.

1720

1109

_s_a

tTH

BS1

5.11

2126

67_a

tSP

AR

C3.

72

2105

11_s

_at

INH

BA

3.93

2049

55_a

tSR

PX4.

6120

9270

_at

LAM

B3

3.27

2011

62_a

tIG

FBP7

3.72

2162

50_s

_at

LPX

N4.

1720

2267

_at

LAM

C2

2.76

2317

66_s

_at

CO

L12A

13.

6720

965l

_at

TGFB

1I1

4.15

2294

04_a

tTW

IST2

2.65

2138

69_x

_at

THY

13.

6022

7566

_at

HN

T3.

9820

0827

_at

PLO

D2.

24

2013

89_a

tIT

GA

53.

5023

1766

_s_a

tC

OL1

2A1

3.67

2131

39_a

tSN

AI2

2.14

2027

60_s

_at

AK

AP2

3.47

2138

69_x

_at

THY

13.

6020

3695

_s_a

tD

FNA

52.

07

2016

45_a

tTN

C3.

4420

1389

_at

ITG

A5

3.50

2035

62_a

tFE

Z13.

3720

1645

_at

TNC

3.44

2249

1l_s

_at

DC

BLD

23.

3120

3562

_at

FEZ1

3.37

2122

33_a

tM

API

B3.

2622

491l

_s_a

tD

C3L

D2

3.31

2092

10_s

_at

PLEK

HC

13.

2420

9270

_at

LAM

B3

3.27

2301

75_s

_at

ESD

X3.

0820

9210

_s_a

tPL

EKH

C1

3.24

2051

20_s

_at

SGC

B2.

9421

2489

_at

CO

L5A

13.

17

2219

16_a

tN

EFL

2.93

2029

98_s

_at

LOX

L23.

15

2046

82 a

tLT

BP2

2.89

2301

75_s

_at

ESD

X3.

08

3740

8_at

MR

C2

2.87

5225

5_s_

atC

OL5

A3

3.02

2192

57_s

_at

SPH

K1

2.86

2124

64_s

_at

FN1

2.93

2186

38_s

_at

SPO

N2

2.74

2113

40_s

_at

MC

AM

2.79

2086

37_x

_at

AC

TN1

2.72

2022

67_a

tLA

MC

22.

76

2242

52_s

_at

FXY

D5

2.66

2186

38_s

_at

SPO

X2

2.74

2044

66_s

_at

SNC

A2.

6120

8637

_x_a

tA

CTN

12.

72

Cancer Res. Author manuscript; available in PMC 2012 February 7.

Page 19: signature in esophageal cancer Author Manuscript NIH ...oncotherapy.us/pdf/Cancer.Stem.Cell_Bridge_Microenvironment_Esophageal-Cancer.pdfsubsequently washed at low (6 x SSPE) and high

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

NIH

-PA Author Manuscript

Michaylira et al. Page 19

Cel

lula

r C

ompo

nent

Org

aniz

atio

n an

dB

ioge

nesi

sB

iolo

gica

l Adh

esio

n an

dC

ellu

lar

Adh

esio

nD

evel

opm

enta

l Pro

ces

GO

:001

6043

GO

:002

2610

& G

O00

0715

5G

O:0

0488

56,0

0325

02,0

0487

31,

0009

888,

0048

513

&00

0727

5

Prob

e Se

tG

ene

Nam

eR

atio

*Pr

obe

Set

Gen

e N

ame

Rat

io*

Prob

e Se

tG

ene

Nam

eR

atio

*

2189

80_a

tFH

OD

32.

5622

2108

at

AM

IG02

2.71

2183

68_s

_at

TNFR

SF12

A2.

5520

5534

_at

PCD

H7

2.66

2080

79_s

_at

STK

62.

5322

4252

_s_a

tFX

YD

52.

66

2010

42_a

tTG

M2

2.53

2183

68_s

_at

TNFR

SF12

A2.

55

2020

95_s

_at

BIR

C5

2.51

2010

42_a

tTG

M2

2.53

2015

05_a

tLA

MB

12.

4820

4359

_at

FLR

T22.

52

2027

96_a

tSY

NPO

2.47

2015

05_a

tLA

MB

12.

48

2100

26_s

_at

CA

RD

102.

4720

4345

_at

CO

L16A

12.

47

2217

48_s

_at

TNS

2.40

2420

64_a

tSD

K2

2.45

2031

31_a

tPD

GFR

A2.

3520

1474

_s_a

tIT

GA

32.

35

2186

44_a

tPL

EK2

2.32

2266

09_a

tD

CB

LD1

2.29

2218

98_a

tT1

A-2

2.27

2252

93_a

tC

OL2

7A1

2.15

2011

85_a

tPR

SS11

2.23

2041

70_s

_at

CK

S22.

22

2117

25_s

_at

BID

2.20

2015

40_a

tFH

L12.

13

2147

52_x

_at

FLN

A2.

03

* Mul

tivar

iate

logi

stic

regr

essi

on m

odel

s wer

e us

ed to

test

eac

h pr

obes

et a

nd a

djus

ted

for f

alse

dis

cove

ry ra

te (p

<0.0

1)

Cancer Res. Author manuscript; available in PMC 2012 February 7.