+ All Categories
Home > Documents > Fibroblast-Derived Dermal Matrix Drives Development of ... · COL7A1 as ranked by Sigma-Aldrich...

Fibroblast-Derived Dermal Matrix Drives Development of ... · COL7A1 as ranked by Sigma-Aldrich...

Date post: 22-Oct-2020
Category:
Upload: others
View: 2 times
Download: 0 times
Share this document with a friend
14
Microenvironment and Immunology Fibroblast-Derived Dermal Matrix Drives Development of Aggressive Cutaneous Squamous Cell Carcinoma in Patients with Recessive Dystrophic Epidermolysis Bullosa Yi-Zhen Ng 1,3 , Celine Pourreyron 1 , Julio C. Salas-Alanis 4 , Jasbani H.S. Dayal 1 , Rodrigo Cepeda-Valdes 4 , Wenfei Yan 5 , Sheila Wright 1 , Mei Chen 6 , Jo-David Fine 7 , Fiona J. Hogg 2 , John A. McGrath 8 , Dedee F. Murrell 5 , Irene M. Leigh 1 , E. Birgit Lane 3 , and Andrew P. South 1 Abstract Patients with the genetic skin blistering disease recessive dystrophic epidermolysis bullosa (RDEB) develop aggressive cutaneous squamous cell carcinoma (cSCC). Metastasis leading to mortality is greater in RDEB than in other patient groups with cSCC. Here we investigate the dermal component in RDEB using mRNA expression proling to compare cultured broblasts isolated from individuals without cSCC and directly from tumor matrix in RDEB and non-RDEB samples. Although gene expression of RDEB normal skin broblasts resembled that of cancer-associated broblasts, RDEB cancer-associated broblasts exhibited a distinct and divergent gene expression prole, with a large proportion of the differentially expressed genes involved in matrix and cell adhesion. RDEB cancer-associated broblasts conferred increased adhesion and invasion to tumor and non- tumor keratinocytes. Reduction of COL7A1, the defective gene in RDEB, in normal dermal broblasts led to increased type XII collagen, thrombospondin-1, and Wnt-5A, while reexpression of wild type COL7A1 in RDEB broblasts decreased type XII collagen, thrombospondin-1, and Wnt-5A expression, reduced tumor cell inva- sion in organotypic culture, and restricted tumor growth in vivo. Overall, our ndings show that matrix composition in patients with RDEB is a permissive environment for tumor development, and type VII collagen directly regulates the composition of matrix proteins secreted by dermal and cancer-associated broblasts. Cancer Res; 72(14); 352234. Ó2012 AACR. Introduction Recessive dystrophic epidermolysis bullosa (RDEB) is an inherited skin blistering disease caused exclusively by muta- tions in the gene-encoding type VII collagen, COL7A1 (1). Type VII collagen is the main component of anchoring brils, structures that support anchorage of the epidermis to the underlying dermis (2). The central dogma is that defective anchoring brils lead to skin fragility characterized by long- term wounds and healing with scarring, resulting in consid- erable disruption to dermal architecture (3). This devastating condition is further complicated by the development of numer- ous, aggressive cutaneous squamous cell carcinoma (cSCC) suggestive of a eld effect. Tumors frequently metastasize, resulting in more than 80% mortality by age 50 (4). This rate of metastasis far exceeds other patient groups in which cSCC is also a major complication, such as organ transplant patients and patients with the hereditary disease xeroderma pigmen- tosum, the reasons for which remain unclear (5). Protocols for the early detection and aggressive management, including amputation, radiation therapy, and chemotherapy, have not been proven to increase survival in RDEB cSCC (6). Previous studies have failed to identify consistent differences between RDEB cSCC and cSCC from the general population or organ transplant recipients in spite of the different clinicopathologic behavior. In fact, in virtually all studies to date, RDEB cSCC exhibit similar characteristics to non-RDEB cSCC and a caus- ative relationship with tumor progression has yet to be unequivocally identied (715). With few exceptions these studies have focused on tumor keratinocytes and have ignored the surrounding stroma. Cancer-associated broblasts are the main cell type present in tumor stroma and have been shown to contribute toward cancer invasion (16), initiation, and pro- gression via stromalepithelial interactions (1719) and can provide oncogenic signals such as broblast growth factors, Authors' Afliations: 1 Division of Cancer Research, Medical Research Institute, Ninewells Hospital and Medical School, University of Dundee; 2 Department of Plastic Surgery, Ninewells Hospital and Medical School, Dundee, United Kingdom; 3 Institute of Medical Biology, A Star, Singapore; 4 Universidad Autonoma de Nuevo Leon, Monterrey, Mexico; 5 St George Hospital, University of New South Wales, Sydney, New South Wales, Australia; 6 Department of Dermatology, Keck School of Medicine, Univer- sity of Southern California, Los Angeles, California; 7 Department of Med- icine, Vanderbilt University School of Medicine, Nashville, Tennessee; and 8 St John's Institute of Dermatology, King's College London (Guy's Cam- pus), London, United Kingdom Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Author: Andrew P. South, Division of Cancer Research, Clinical Research Centre, Ninewells Hospital and Medical School, Dundee DD1 9SY, UK. Phone: 44-1382-496432; Fax: 44-1382-436892; E-mail: [email protected] doi: 10.1158/0008-5472.CAN-11-2996 Ó2012 American Association for Cancer Research. Cancer Research Cancer Res; 72(14) July 15, 2012 3522 on April 4, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst May 7, 2012; DOI: 10.1158/0008-5472.CAN-11-2996
Transcript
  • Microenvironment and Immunology

    Fibroblast-Derived Dermal Matrix Drives Development ofAggressiveCutaneous SquamousCell Carcinoma in Patientswith Recessive Dystrophic Epidermolysis Bullosa

    Yi-Zhen Ng1,3, Celine Pourreyron1, Julio C. Salas-Alanis4, Jasbani H.S. Dayal1, Rodrigo Cepeda-Valdes4,Wenfei Yan5, Sheila Wright1, Mei Chen6, Jo-David Fine7, Fiona J. Hogg2, John A. McGrath8, Dedee F. Murrell5,Irene M. Leigh1, E. Birgit Lane3, and Andrew P. South1

    AbstractPatients with the genetic skin blistering disease recessive dystrophic epidermolysis bullosa (RDEB) develop

    aggressive cutaneous squamous cell carcinoma (cSCC). Metastasis leading tomortality is greater in RDEB than inother patient groups with cSCC. Here we investigate the dermal component in RDEB using mRNA expressionprofiling to compare cultured fibroblasts isolated from individuals without cSCC and directly from tumor matrixin RDEB and non-RDEB samples. Although gene expression of RDEB normal skin fibroblasts resembled that ofcancer-associated fibroblasts, RDEB cancer-associated fibroblasts exhibited a distinct and divergent geneexpression profile, with a large proportion of the differentially expressed genes involved in matrix and celladhesion. RDEB cancer-associated fibroblasts conferred increased adhesion and invasion to tumor and non-tumor keratinocytes. Reduction of COL7A1, the defective gene in RDEB, in normal dermal fibroblasts ledto increased type XII collagen, thrombospondin-1, andWnt-5A, while reexpression of wild type COL7A1 in RDEBfibroblasts decreased type XII collagen, thrombospondin-1, and Wnt-5A expression, reduced tumor cell inva-sion in organotypic culture, and restricted tumor growth in vivo. Overall, our findings show that matrixcomposition in patients with RDEB is a permissive environment for tumor development, and type VII collagendirectly regulates the composition of matrix proteins secreted by dermal and cancer-associated fibroblasts.Cancer Res; 72(14); 3522–34. �2012 AACR.

    IntroductionRecessive dystrophic epidermolysis bullosa (RDEB) is an

    inherited skin blistering disease caused exclusively by muta-tions in the gene-encoding type VII collagen, COL7A1 (1). TypeVII collagen is the main component of anchoring fibrils,structures that support anchorage of the epidermis to theunderlying dermis (2). The central dogma is that defectiveanchoring fibrils lead to skin fragility characterized by long-

    term wounds and healing with scarring, resulting in consid-erable disruption to dermal architecture (3). This devastatingcondition is further complicated by the development of numer-ous, aggressive cutaneous squamous cell carcinoma (cSCC)suggestive of a field effect. Tumors frequently metastasize,resulting in more than 80%mortality by age 50 (4). This rate ofmetastasis far exceeds other patient groups in which cSCC isalso a major complication, such as organ transplant patientsand patients with the hereditary disease xeroderma pigmen-tosum, the reasons for which remain unclear (5). Protocols forthe early detection and aggressive management, includingamputation, radiation therapy, and chemotherapy, have notbeen proven to increase survival in RDEB cSCC (6). Previousstudies have failed to identify consistent differences betweenRDEB cSCC and cSCC from the general population or organtransplant recipients in spite of the different clinicopathologicbehavior. In fact, in virtually all studies to date, RDEB cSCCexhibit similar characteristics to non-RDEB cSCC and a caus-ative relationship with tumor progression has yet to beunequivocally identified (7–15). With few exceptions thesestudies have focused on tumor keratinocytes and have ignoredthe surrounding stroma. Cancer-associated fibroblasts are themain cell type present in tumor stroma andhave been shown tocontribute toward cancer invasion (16), initiation, and pro-gression via stromal–epithelial interactions (17–19) and canprovide oncogenic signals such as fibroblast growth factors,

    Authors' Affiliations: 1Division of Cancer Research, Medical ResearchInstitute, Ninewells Hospital and Medical School, University of Dundee;2Department of Plastic Surgery, Ninewells Hospital and Medical School,Dundee, United Kingdom; 3Institute of Medical Biology, A�Star, Singapore;4Universidad Autonoma de Nuevo Leon, Monterrey, Mexico; 5St GeorgeHospital, University of New South Wales, Sydney, New South Wales,Australia; 6Department of Dermatology, Keck School of Medicine, Univer-sity of Southern California, Los Angeles, California; 7Department of Med-icine, Vanderbilt University School of Medicine, Nashville, Tennessee; and8St John's Institute of Dermatology, King's College London (Guy's Cam-pus), London, United Kingdom

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

    Corresponding Author: Andrew P. South, Division of Cancer Research,Clinical Research Centre, Ninewells Hospital and Medical School, DundeeDD1 9SY, UK. Phone: 44-1382-496432; Fax: 44-1382-436892; E-mail:[email protected]

    doi: 10.1158/0008-5472.CAN-11-2996

    �2012 American Association for Cancer Research.

    CancerResearch

    Cancer Res; 72(14) July 15, 20123522

    on April 4, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 7, 2012; DOI: 10.1158/0008-5472.CAN-11-2996

    http://cancerres.aacrjournals.org/

  • which act in a paracrine manner to transformed tumorkeratinocytes (20). Prompted by our recent observations thatvery few genetic and behavioral differences exist betweenprimary cultures of cSCC keratinocytes derived from RDEBand non-RDEB individuals (15), we now apply a similarapproach to study primary cultures of fibroblasts derived fromRDEB patient skin and cSCC.

    Materials and MethodsTissue samplesThis study was conducted according to the Declaration of

    Helsinki Principles and was approved by the appropriateEthics Committees. RDEB patient diagnosis was confirmedby characteristic immunofluorescence findings and clinicalcriteria (3). Biopsies from normal skin or cSCC tissue fromnon-RDEB and RDEB patients were obtained after informedconsent. Supplementary Table S1 gives details of all samplesused for fibroblast isolation in this study. Normal non-RDEB,non-cSCC samples from redundant control skin were obtainedfrom nonmalignant esthetic plastic surgery procedures.

    Cell cultureFibroblasts were isolated as follows. Briefly, after mechan-

    ical disassociation and trypsin digestion, biopsy fragmentswere subjected to collagenase D (Roche Diagnostics) over-night. Fibroblasts were grown in high-glucose Dulbecco'sModified Eagle Media supplemented with 10% FBS. All fibro-blast cultures used in this study were of early passage lessthan 8.

    Cell proliferation assayFibroblasts were seeded at 20,000 cells/cm2 density in 96-

    well plates in triplicate and a methylthiazolyldiphenyl-tetra-zolium bromide (MTT) cell proliferation assay (#30-1010K;American Type Culture Collection, Teddington, UK) was con-ducted according to manufacturer's instructions every 24hours over 5 days. Biologic replicates from each fibroblasttype were then analyzed in Prism 5 (Graphpad software).

    Agilent one-colour microarray and expression analysisFull details of microarray and analysis are given in Supple-

    mentary Data. Briefly, fibroblasts seeded at 20,000 cells/cm2

    density were lysed after 4 days and 100 ng/mL of RNA wasamplified, labeled, and hybridized onto Agilent Single ColourWhole Human Genome Oligo Microarray 4 � 44k cDNA array(G4112F, Design ID 14850; Agilent Technologies) according tothe manufacturer's guidelines. The microarray data discussedin this publication have been deposited in NCBI's Gene Expres-sion Omnibus (21) and are accessible through GEO Seriesaccession number GSE37738 (http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc¼GSE37738).

    AntibodiesThe following primary antibodies were used for immuno-

    blotting and immunostaining: anti-aSMA (1A4; Abcam),anti-FAP (ab53066; Abcam), anti-FSP1 (ab27597; Abcam),anti-glyceraldehyde-3-phosphate dehydrogenase (GAPDH;

    71.1; Sigma-Aldrich), anti-keratin cocktail (LL001 and LP34,produced in house), anti-MUC1 (VU4H5; Santa Cruz), anti-TSP1 (A6.1 and A4.1; Santa Cruz), antitype V collagen (1E2E4;Millipore and ACL50511AP; Accurate Chemicals), polyclonalanti-type VII collagen raised against the NC1 domain, antitypeXII collagen (A11), anti-vimentin (V9; Santa Cruz), and anti-WNT-5a (AF645; R&D). The following secondary antibodieswere used: goat anti-mouse Alexa Fluor 488 (Invitrogen), goatanti-rabbit Alex Fluor 568 (Invitrogen), goat anti-mouse HRP(Dako), and swine anti-rabbit HRP (Dako).

    ImmunoblottingSamples were resolved by 1-dimensional (1D) SDS-PAGE

    gels using standard techniques. Equal amounts of proteinwere subjected to SDS-PAGE and transferred to nitrocellu-lose membrane. The membrane was subsequently blockedwith 5% nonfat milk or 5% bovine serum albumin (BSA) for 1hour and probed with specific primary antibodies, followedby the appropriate peroxidase-conjugated secondary anti-body. Proteins were detected using the ECL Western Blot-ting Detection Reagents (GE Healthcare) or SuperSignalWest Dura Extended Duration Substrate (Thermo FisherScientific). The membrane was then stripped and reprobedfor loading control.

    Tissue preparation, histology, and immunostainingTissues were fixed in 4% paraformaldehyde and embedded

    in paraffin. Four-micrometer-thick sections were cut and air-dried overnight before being deparaffinized and then rehy-drated through a decreasing concentration of alcohol to water.Standard heat-based antigen retrieval method was used. Sec-tions were stained using Vectastain ABC kits (Vector Labs)or Bond Autostainer (Leica Microsystems GmbH) accordingto the manufacturer's protocol. Liquid diaminobenzidine(DAB; Dako) was subsequently applied for 3 to 10 minutesand sections were counterstained with hematoxylin. Slideswere photographed using AxioImager Z1 (Carl Zeiss Micro-Imaging GmbH).

    Morphogenic analysis and immunohistochemicalquantification

    A coherent single square lattice was applied to each pho-tograph of DAB stained tissue to produce a total of 100 testpoints determined by intersect of the 10 � 10 lattice. At eachintersect a 20 � 20 pixel area was sampled. Those areas thatdid not fall onto stroma (i.e., were either not occupied by tissueor were obviously epithelial or tumor cells) were discarded.Using color deconvolution in FIJI (http://fiji.sc/), DAB wasconverted to a 256 gray scale and the average intensity acrossall areas for all sections was calculated. The fraction of the areaoccupied by those pixels greater than the average signalintensity was calculated and used to generate box and whiskerplots shown in Fig. 4.

    Cell-derived matrix and invasion assayCell-derived matrix (CDM) was made in vitro based on the

    protocol by Larouche and colleagues (22) with somemodifica-tions. Briefly, 10,000 fibroblasts per cm2 were seeded on plastic

    Dermal Matrix Composition Promotes RDEB Cancer

    www.aacrjournals.org Cancer Res; 72(14) July 15, 2012 3523

    on April 4, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 7, 2012; DOI: 10.1158/0008-5472.CAN-11-2996

    http://cancerres.aacrjournals.org/

  • and the media was supplemented with 0.1 mmol/L L-ascorbicacid 2-phosphate (A8960; Sigma-Aldrich) with refeedingevery 2 to 3 days. After 5 to 7 weeks, the single layer of CDMformed was released and left in media for 5 days. RDEBSCCtumor cells were then seeded onto this CDM, which was raisedto air–liquid interface the next day. Tumor cells were left toinvade for 7 to 14 days before fixation in 4% paraformaldehyde.

    Invasion quantificationSections of the CDMwere immunostainedwith the antibody

    LL001 against keratin 14 and photographed using AxioImagerZ1 (Carl Zeiss MicroImaging GmbH). The obtained imageswere analyzed using FIJI. The imageswerefirst processed usingcolor deconvolution into 8-bit images and then a threshold setto select for the tumor cells stained by LL001. The invasionindex was calculated based on the total number of invadingparticles (excluding the surface epithelium) times the totalarea of the particles for each image.

    RNA interferenceWe used the top 3 siRNA oligonucleotides against

    COL7A1 as ranked by Sigma-Aldrich (SASI_Hs01_00155170,SASI_Hs01_00155171, SASI_Hs01_00155173) and a nontarget-ing control (NTC; MISSION siRNA Universal Negative Control#1; Sigma-Aldrich). Normal human fibroblasts (NHF) wereseeded in 6-well plates at 100,000 cells/well and transfected24 hours later with pooled siRNA or NTC (40 nmol/L finalconcentration) using Lipofectamine 2000 (Invitrogen) dilutedin Opti-MEM (Invitrogen) according to the manufacturer'sinstructions.

    Recombinant type VII collagen expressionFull details of recombinant type VII collagen expression are

    given in Supplementary Data. Briefly, wild-type COL7A1 wasdelivered with infectious replicative defective retrovirus andtarget cells were selected with puromycin (2 mg/mL).

    Tumorigenicity assaysAll animals were used in accordance with UK Home Office

    regulations. Full details of the protocol are given in Supple-mentary Data. Briefly, a suspension of 4� 106 SCCIC1 cells and1� 106 of the indicated population of primary fibroblasts weremixed with high-concentration Matrigel� (Becton Dickinson)and injected subcutaneously into the flanks of SCID balb/cmice. Tumors were regularly measured by caliper.

    ResultsRDEB fibroblast cultures display comparableproliferation rates to equivalent non-RDEB fibroblastcultures

    We isolated dermal fibroblasts from RDEB patients withouta clinical diagnosis of cSCC (herein referred to as RDEBF), fromRDEB cSCC (RDEBSCCF) as well as from nonpathologic skinfrom esthetic plastic surgery procedures (NHF) and sponta-neous UV-induced cSCCs excised from non-RDEB individuals(UVSCCF), as described in Materials and Methods. Supple-mentary Table S1 details the samples used in this study. No

    distinct morphologic differences between cells isolated fromthe 4 patient groups were observed (Fig. 1A). Using anapproach similar to our previous analysis of cultured kerati-nocytes (15), we seeded fibroblasts at a relative high-densityand assayed proliferation over 5 days using theMTT assay (Fig.1B). This showed no significant difference between the pro-liferation rates under these conditions and identified thatcultures were relatively quiescent at day 4. We screened 2RDEB cSCC fibroblast isolations for genetic rearrangementusing 10K SNPmapping arrays and observed no abnormalities(data not shown). No contaminating keratinocytes wereobserved in any fibroblast isolations and no expression ofkeratinocyte markers such as mucin-1 (23) or keratin wasevident (Fig. 1C). All populations tested expressed similarlevels of vimentin and varied in the expression of otherfibroblast markers such as a-smooth muscle actin (a-SMA),fibroblast specific protein 1 (FSP1), and fibroblast-activatedprotein (FAP; refs. 24 and 25; Fig. 1D).

    mRNA expression profiling demonstrates thatnoncancer RDEB skin fibroblasts behave ascancer-associated fibroblasts

    Wechose to assaymRNAexpression at day 4when cells wererelatively quiescent as we have previously shown that a similarapproach analyzing keratinocytes was capable of identifyingtargets necessary for cSCC cell survival (15). Unsupervisedclustering of all 20,095 probes passing quality control on theAgilent array platform clearly separated NHF from RDEBSCCFbut was unable to separate RDEBF from UVSCCF (Fig. 2A).One-way ANOVA analysis identified 1,098 probes as beingdifferentially expressed between disease state and normal(Fig. 2B). The majority of these probes were differentiallyexpressed in RDEBSCCF, whereas the majority of probesdifferentially expressed in RDEBF were also differentiallyexpressed in UVSCCF and shared with RDEBSCCF (Fig. 2C).Analysis of all 1,098 probes as well as 159 probes common to all3 disease groups showed a stepwise progression starting withUVSCCF andRDEBF, followed by RDEBSCCF, showing that thelevel of deregulation in this gene set was greatest in RDEBSCCF(Fig. 2D).

    An activated gene signature clusters normal fibroblastsbut cannot separate noncancer RDEB skin fibroblastsfrom cancer-associated fibroblasts

    Previous array experimentation using normal dermalcultured fibroblasts has identified a core serum response(CSR) gene set, which is differentially expressed in responseto serum exposure (26). Upon exposure to serum fibroblastsbecome activated and display similar features to thoseobserved in a wounded or cancer environment. Importantly,this CSR gene signature is evident in various tumor tissuesand is associated with a worse prognosis in breast, lung, andgastric cancer, suggesting that tumors and their associatedmicroenvironment have a wound-like phenotype (26). Weobserved little overlap between the CSR gene signature andour 1,098 probes; 61 of 904 probes on our array representingthe 591 CSR genes were differentially expressed. However,we investigated whether the CSR signature alone was

    Ng et al.

    Cancer Res; 72(14) July 15, 2012 Cancer Research3524

    on April 4, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 7, 2012; DOI: 10.1158/0008-5472.CAN-11-2996

    http://cancerres.aacrjournals.org/

  • capable of separating disease state. Clustering of our fibro-blast samples based on the CSR gene signature resulted in aseparation of NHF from the 3 disease states but was unableto cluster RDEBSCCF (Supplementary Fig. S1). The CSR genesignature clustering highlights the role of serum response inall 3 disease states, corroborating similarities in the tran-scriptional profiles of RDEBF, UVSCCF, and RDEBSCCF thatwe observe in our ANOVA analysis.

    Matrix-associated gene expression is increased in RDEB,UV cSCC, and RDEB cSCC fibroblast culturesKEGG pathway analysis of the 1,098 differentially expressed

    genes identified a significant enrichment of genes associatedwith adhesion and extracellular matrix, with 6/13 pathways

    being involved in these processes (Fig. 3A and SupplementaryTable S2). The differences identified in COL5A1, COL12A1,ITGA3, ITGA6, and TSP1 were confirmed at the level of proteinexpression by Western blotting (Fig. 3B). Other differentiallyexpressed genes of note included Wnt5A (confirmed by West-ern blot, Fig. 3B), TLR4, TGFBR3, DAP, and ACVRL1.

    Type V collagen, type XII collagen, and thrombospondin-1 are upregulated in cSCC stroma

    We analyzed type V collagen, thrombospondin-1, and typeXII collagen expression in normal skin (n¼ 3), UV cSCC (n¼ 6),and RDEB cSCC (n ¼ 8) tissue using immunohistochemistry.Type V collagen expression was detected in the dermis ofnormal skin and was significantly increased in the tumor

    Figure 1. RDEB fibroblast cultures display proliferation rates and marker expression comparable with corresponding non-RDEB fibroblast cultures. A,representative images of confluent cultured primary fibroblasts (day 4; magnification,�100). B, MTT assay average colorimetric reading relative to day 1for each fibroblast sample group seeded at high density (20,000 cells/cm2) and cultured over 5 days. Results are triplicate mean � SD, n ¼ 3 to 5 pergroup. C, no evidence of MUC-1 or cytokeratin expression in fibroblast cultures. NHK, primary normal human keratinocyte; RSK, RDEBSCCkeratinocyte line, SCCRDEB2. D, expression of markers a-SMA, FSP1, FAP-a, and vimentin are consistent with cultured dermal fibroblasts and donot separate sample groups.

    Dermal Matrix Composition Promotes RDEB Cancer

    www.aacrjournals.org Cancer Res; 72(14) July 15, 2012 3525

    on April 4, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 7, 2012; DOI: 10.1158/0008-5472.CAN-11-2996

    http://cancerres.aacrjournals.org/

  • CB

    NHF

    NHF

    Algorithm: hierarchical clusteringparameters:

    UVSCCF vs. NHF201 probes

    RDEBF vs. NHF287 probes

    RDEBSCCF vs. NHF953 probes

    Legend - Dendrogram

    Color range

    –3.1 0 3.1

    Cluster on = both rows and columnsDistance metric = EuclideanLinkage rule = average

    UVSCCF

    UVSCCF

    / RDEBF

    RDEBF

    RDEBSCCF

    RDEBSCCF

    RDEBF vs. NHF

    5

    4

    3

    2

    1

    0

    Ave

    rage

    ran

    k

    5

    4

    3

    2

    1

    0

    Ave

    rage

    ran

    k

    RDEB

    F

    UVSCCF vs. NHF

    UVSC

    CFNHF

    RDEBSCCF vs. NHF

    RDEB

    SCCF

    RDEB

    F

    UVSC

    CFNHF

    RDEB

    SCCF

    D Nonparametric ranking of gene dysregulation in each fibroblast group

    i) Using all 1,098 differentially expressed probes

    A

    NH

    F66

    NH

    F88

    NH

    F2

    SC

    CIC

    15

    RD

    EB

    48F

    SC

    CT

    7F

    SC

    CT

    6F

    SC

    CIC

    17

    RD

    EB

    23F

    RD

    EB

    26F

    SC

    CIC

    16

    RD

    EB

    27F

    RD

    EB

    3SC

    C1F

    RD

    EB

    2SC

    C1F

    RD

    EB

    43S

    CC

    1F

    RD

    EB

    7SC

    CC

    2F

    ii) Using 159 differentially expressed probes shared by all 3disease states compared to NHF

    Figure 2. mRNA expression profiling shows that RDEB normal skin fibroblasts behave as cancer-associated fibroblasts and RDEB cancer-associatedfibroblasts represent a distinct sample group. A, unsupervised clustering based on 20,095 probes that passed filtering criteria clearly separates fibroblastcultures into 3 groups: NHF, UVSCCF/RDEBF, and RDEBSCCF. B, supervised hierarchical clustering of 1,098 differentially expressed transcripts acrossall 3 disease states shows RDEBSCCF represent a distinct sample group. C, Venn diagram depicting the overlaps in 1,098 differentially expressed genetranscripts frompair-wisecomparisonswithNHF.Mostof thegene transcripts identifiedaredifferentiallyexpressed inRDEBSCCF(953of1,098probes).RDEBFand UVSCCF show very similar gene expression changes, which overlap considerably with RDEBSCCF. D, nonparametric ranking of all 1,098 probes (top)and 159 probes dysregulated in all 3 fibroblast disease groups (bottom) compared with NHF reveals a stepwise progression in gene dysregulation fromNHF to RDEBSCCF. Each probe was ranked according to level of gene dysregulation (either increased or decreased expression) compared with NHF.

    Ng et al.

    Cancer Res; 72(14) July 15, 2012 Cancer Research3526

    on April 4, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 7, 2012; DOI: 10.1158/0008-5472.CAN-11-2996

    http://cancerres.aacrjournals.org/

  • stroma in 7/8 RDEB cSCC and 2/6 UV cSCC (Fig. 4, top).Thrombospondin-1 expression was virtually undetectable inthe dermis of normal skin and was significantly increasedin the stroma of 12/13 cSCC samples (Fig. 4, middle). Bothtype V collagen and thrombospondin-1 were also expressedby normal epidermal keratinocytes as well as tumor keratino-

    cytes (data not shown). Type XII collagen was weakly detectedin the dermis of normal skin and significantly increased inthe stroma of 5/6 UV cSCC and 5/7 RDEB cSCC samples (Fig. 4,bottom). These data show that the changes in protein expres-sion identified in cultured fibroblasts are also apparent incSCC in vivo.

    KEGG terms

    1. RibosomeECM / cell adhesion related

    1

    ITGA3 mRNA expression level

    *

    ITGA3

    GAPDH

    ITGA6

    GAPDH

    WNT-5A

    GAPDH

    TSP1

    GAPDH

    Type XIIcollagen

    GAPDH

    Type Vcollagen(1e2e4)

    GAPDH

    * *

    ***

    TSP1 mRNA expression level COL12A1 mRNA expression level

    ITGA6 mRNA expression level COL5A1 mRNA expression level WNT5A mRNA expression level

    2 3 4 5 6 7 8 9 10 11 12 13

    20

    15

    10

    5

    0

    5

    4

    3

    2

    1

    0

    4

    3

    2

    1

    0

    2.0

    1.5

    1.0

    0.5

    0.0Nor

    mal

    ized

    sig

    nal (

    linea

    r)

    10

    8

    6

    4

    2

    0Nor

    mal

    ized

    sig

    nal (

    linea

    r)

    Nor

    mal

    ized

    sig

    nal (

    linea

    r)

    2.0

    1.5

    1.0

    0.5

    0.0Nor

    mal

    ized

    sig

    nal (

    linea

    r)

    Nor

    mal

    ized

    sig

    nal (

    linea

    r)

    3

    2

    1

    0

    −1Nor

    mal

    ized

    sig

    nal (

    linea

    r)

    No.

    of p

    robe

    s in

    eac

    h K

    EG

    G te

    rm

    Others

    2. Heparan sulfate biosynthesis

    3. Chondroitin sulfate biosynthesis

    4. Glutamate metabolism

    5. D-Glutamine and D-glutamate metabolism

    6. 3-Chloroacrylic acid degradation

    7. Hematopoietic cell lineage

    8. Nicotinate and nicotinamide metabolism

    9. Vibrio cholerae infection

    10. Focal adhesion

    11. ECM–receptor interaction

    12. Axon guidance

    13. Cell adhesion molecules

    A

    B

    NHF UVSCCF RDEBF RDEBSCCF

    NHF UVSCCF RDEBF RDEBSCCF NHF UVSCCF RDEBF RDEBSCCF NHF UVSCCF RDEBF RDEBSCCF

    NHF UVSCCF RDEBF RDEBSCCF NHF UVSCCF RDEBF RDEBSCCFNHF UVSCCF RDEBF RDEBSCCF

    NHF UVSCCF RDEBF RDEBSCCF NHF UVSCCF RDEBF RDEBSCCF NHF UVSCCF RDEBF RDEBSCCF

    NHF UVSCCF RDEBF RDEBSCCF NHF UVSCCF RDEBF RDEBSCCF

    Figure 3. Matrix-associated gene and protein expression is increased in RDEB, UV cSCC, and RDEB cSCC fibroblast cultures. A, KEGG pathway analysisidentifies an enrichment of gene expression associated with extracellular matrix (ECM) and cell adhesion in RDEB and cSCC sample groups. B, arraysignal intensity (graph,multiple dots correspond tomultiple probeswherepresent) is reflectedbyprotein expressionof typeVcollagen, integrina3, integrina6,Wnt-5A, thrombospondin-1, and type XII collagen as determined by Western blotting.

    Dermal Matrix Composition Promotes RDEB Cancer

    www.aacrjournals.org Cancer Res; 72(14) July 15, 2012 3527

    on April 4, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 7, 2012; DOI: 10.1158/0008-5472.CAN-11-2996

    http://cancerres.aacrjournals.org/

  • A CCSBEDRCCSlamroType V collagen staining

    Norm

    alSC

    C

    RDEB

    SCC

    Norm

    alSC

    C

    RDEB

    SCC

    Norm

    alSC

    C

    RDEB

    SCC

    150

    100

    50

    0

    Ave

    rage

    are

    a fr

    actio

    n

    150

    100

    50

    0

    Ave

    rage

    are

    a fr

    actio

    n

    150

    100

    50

    0

    Ave

    rage

    are

    a fr

    actio

    n

    Type XII collagen staining

    Thrombospondin-1 staining

    N

    Negative control

    CCSBEDRCCSlamroN

    Negative control

    CCSBEDRCCSlamroN

    Negative control

    B

    C

    Figure 4. Immunohistochemical labeling identifies increased matrix-associated protein expression in cSCCs. Labeling with anti-type V collagen (A), anti-thrombospondin-1 (B), and anti-type XII collagen (C) antibodies identifies increased expression in the stroma of UV-induced (n ¼ 6) and RDEB cSCC(n ¼ 7-8) compared with normal skin (n¼ 3). The percentage of pixels staining with intensity above the average signal is expressed as average area fraction(graph, left). Box and whisker plots show top and bottom quartile (box), median (central line), and 10th and 90th percentile values (error bars). Right, arepresentative 200 � 200 pixel region of stroma from individual normal or tumor samples. Negative control (omitting primary antibody) shows no labeling.Scale bar, 50 mm.

    Ng et al.

    Cancer Res; 72(14) July 15, 2012 Cancer Research3528

    on April 4, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 7, 2012; DOI: 10.1158/0008-5472.CAN-11-2996

    http://cancerres.aacrjournals.org/

  • RDEB cSCC fibroblast–derived extracellular matrixpromotes adhesion of cSCC and skin keratinocytesisolated from RDEB and non-RDEB individualsWehypothesized that the difference in protein expression in

    RDEB and cSCC fibroblasts would influence the behavior ofcSCC keratinocytes. To assess the fibroblasts' own secretedmatrix in the absence of an artificial input, we coated tissueculture plastic dishes with primary fibroblast secreted matrix

    as described in Supplementary Data. We assessed fibroblastsecreted matrix from 7 separate primary populations, 3 NHF, 1RDEBF, 1 UVSCCF, and 2 RDEBSCCF. Adhesion was expressedrelative to collagen I for 6 separate keratinocyte lines: 1 HPVimmortalized axilla, 1 spontaneously immortalized foreskin, 1HPV immortalized RDEB axilla, 1 cSCC, and 2 RDEB cSCC.Every keratinocyte population we tested was more adhesive inthe presence of RDEBSCCF matrix compared with all other

    siRNA knockdown

    Type XIIcollagen

    Type VII collagen

    Type XIIcollagen

    TSP1

    TSP1

    Wnt-5A

    Wnt-5A

    GAPDH

    GAPDH

    Knockdown of COL7A1 mRNA

    Overexpression of C7

    1.5

    1.0

    0.5

    0.0

    25

    20

    15

    10

    5

    0

    2.5

    2.0

    1.5

    1.0

    0.5

    0.0

    1.5

    1.0

    0.5

    0.0

    1.5

    1.0

    0.5

    0.0

    1.5

    1.0

    0.5

    0.0

    1.8

    1.6

    1.4

    1.2

    1.0

    0.8

    1.6

    1.4

    1.2

    1.0

    0.8

    C7 Pooled

    C7 pBabeRDEB1SCCF

    A

    C

    B

    RDEB1SCCF

    RDEB4SCCF

    RDEB4SCCF

    RDEB16SCCF

    RDEB16SCCF

    RDEB29F

    RDEB29F

    RDEB1SCCFRDEB4SCCFRDEB16SCCFRDEB29F

    RDEB1SCCFRDEB4SCCFRDEB16SCCFRDEB29F

    RDEB1SCCFRDEB4SCCFRDEB16SCCFRDEB29F

    C7 pBabe C7 pBabe

    C7 pBabe

    C7 pBabe C7 pBabe

    C7 pBabe

    C7 pBabe

    Scr

    C7 Pooled Scr

    C7 Pooled Scr

    C7 Pooled Scr

    Rel

    ativ

    e pr

    otei

    n le

    vel

    of ty

    pe V

    II co

    llage

    n

    Rel

    ativ

    e pr

    otei

    n le

    vel

    of ty

    pe V

    II co

    llage

    n

    Relative protein levelof type XII collagen

    Relative protein levelof TSP1

    Relative protein levelof WNT-5A

    Relative protein levelof type XII collagen

    Relative protein levelof TSP1

    Relative protein levelof WNT-5A

    Type VII collagen

    GAPDH

    GAPDH

    C7 Pooled Scr

    Figure 5. COL7A1 knockdown in normal dermal fibroblasts increases type XII collagen, thrombospondin-1, and Wnt-5A expression, whereas COL7A1overexpression in RDEB fibroblasts reduces type XII collagen, thrombospondin-1, and Wnt-5A expression. A, siRNA-mediated knockdown of COL7A1 in 3separate normal dermal fibroblast populations yields reduction in type VII collagen expression in whole-cell lysates after 4 days as identified by Westernblotting. C7 Pooled, pool of 3 COL7A1 targeting siRNA; Scr, nontargeting control siRNA. B, Western blotting identifies increases in type XII collagen,thrombospondin-1 and Wnt-5A after COL7A1 knockdown. Right panel graphs show Western densitometry from each separate fibroblast populationnormalized to GAPDH and scrambled control. C, overexpression of COL7A1 in 4 separate RDEB patient–derived fibroblast populations results in decreasedthrombospondin-1, type XII collagen, and Wnt-5A expression. C7, pbabe-COL7A1 vector expressing full-length wild-type COL7A1; pBabe, pBabe-puroempty vector alone. Right, graphs show Western densitometry normalized to GAPDH and scrambled control.

    Dermal Matrix Composition Promotes RDEB Cancer

    www.aacrjournals.org Cancer Res; 72(14) July 15, 2012 3529

    on April 4, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 7, 2012; DOI: 10.1158/0008-5472.CAN-11-2996

    http://cancerres.aacrjournals.org/

  • fibroblast secreted matrix (P < 0.001, combined analysis of3 experiments conducted in quintuplicate; SupplementaryFig. S2).

    RDEB cSCC fibroblast–derived matrix promotes RDEBcSCC keratinocyte invasion

    Wenext examined the invasion of RDEB cSCC keratinocytesinto 3D organotypic models constructed from fibroblast-derived cellular matrix. Rather than embed patient fibroblastsinto a synthetic matrix or cadaveric dermis with or without thebasement membrane we generated 3D organotypic culturesusing CDMs as described (22). 3D matrices derived fromRDEBSCCF increased invasion compared with correspondingNHF or UVSCCF (Supplementary Fig. S3) supporting thehypothesis that RDEB cSCC–derived matrix influences tumorcell behavior.

    Manipulation of COL7A1 in primary normal dermalfibroblasts and RDEB fibroblasts modulates matrixcomposition

    Fibroblasts from different anatomic sites retain their site-specific gene expression profiles in culture (27). To assesswhether the difference in gene expression observed inRDEBSCCF, RDEBF, and UVSCCF were a result of positionalinformation acquired as a result of a wounded environment (inthe case of RDEB and RDEB cSCC) and/or a tumor environ-ment (in the case of cSCC and RDEB cSCC) or whether thepresence of full-length type collagen VII influences the expres-sion of extracellularmatrix components identified in our array,we examined the effect of COL7A1 knockdown in normaldermal fibroblasts populations (n ¼ 3). Of 6 proteins tested,we observed an increase in the level of type XII collagen,thrombospondin-1, and Wnt-5A expression after COL7A1knockdown (Fig. 5A and B). We confirmed the observationthat COL7A1 modulates the expression of matrix-associatedproteins, by reexpressing wild-type cDNA in 4 RDEB fibroblastpopulations (3 RDEBSCCF and 1 RDEBF): we then observed asignificant reduction in type XII collagen, thrombospondin,and Wnt-5A (Fig. 5C).

    Expression of full-length recombinant type VII collagenin RDEB fibroblasts retards cancer cell invasion and invivo tumor progression

    To assess the functional consequences of type VII col-lagen reexpression in RDEB patient fibroblasts, we assessedRDEB29F expressing either recombinant type VII collagen orempty vector control in organotypic invasion assays and cSCCxenograft tumor progression.

    Recombinant type VII collagen significantly retarded theinvasion of RDEB cSCC keratinocytes into 3D organotypicmodels constructed from fibroblast-derived cellular matrix inseparate experiments (conducted in duplicate and triplicate,respectively; Fig. 6). Coinoculation of SCCIC1, a UV-inducedcSCC keratinocyte line derived from an immunocompetentindividual, with RDEB fibroblasts expressing empty vectoralone significantly accelerated tumor growth after 50 dayswhen compared with the same RDEB fibroblasts expressing

    recombinant type VII collagen or NHF (Fig. 7A and B). Histo-logic examination of these tumors showed that NHF and RDEBfibroblasts expressing type VII collagen induced significantdifferentiation compared with RDEB fibroblasts expressingempty vector alone (Fig. 7C).

    DiscussionHerewe show through gene expression profiling of skin- and

    tumor-isolated fibroblasts that the RDEB cSCC dermal micro-environment is strikingly distinct compared with normal skinand UV-induced cSCC. Cultured RDEB cSCC fibroblastsaccount for 86.8% (953) of the 1,098 differentially expressedgenes identified in this study of which 61.4% (674) are differ-entially expressed only in this sample group when comparedwith normal dermal fibroblast cultures (Fig. 2C). Furthermore,considerable overlap between the expression changes, whichseparate RDEB skin and UV cSCC from control skin, showsthat RDEB dermal fibroblasts are indistinguishable from can-cer-associated fibroblasts (Fig. 2). We show that the differ-ences identified here are not a result of physical genetic changeor occult tumor cells within our primary isolations (Fig. 1 anddata not shown) and by identifying changes in quiescentcultures supported by the cells' own secreted matrix we have

    A

    B

    1 W

    k

    RDEB29FpBabe

    RDEB29F C7

    Invasion index

    Cell-derived matrix

    Inva

    sion

    inde

    x(N

    orm

    aliz

    ed to

    pB

    abe)

    RDEB29FpBabe RDEB29F C7

    150

    100

    50

    0

    *

    Figure 6. Full-length type VII collagen expression in RDEBF cell–derivedmatrix retards cSCC keratinocyte invasion. Invasion of SCCRDEB4keratinocytes into RDEB cell–derived matrix is retarded in RDEBF cellsexpressing recombinant full-length type VII collagen (RDEB29F C7)compared with identical cells expressing empty vector alone (RDEB29FpBabe). An invasion index was calculated from 3 to 13 images for eachcell-derived matrix organotypic culture over 2 experiments andnormalized to pBabe. A, normalized invasion index from a representativeexperiment � SEM. B, representative sections stained with a keratin14 antibody highlightingSCCRDEB4keratinocytes;magnification,�200;�, P < 0.05.

    Ng et al.

    Cancer Res; 72(14) July 15, 2012 Cancer Research3530

    on April 4, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 7, 2012; DOI: 10.1158/0008-5472.CAN-11-2996

    http://cancerres.aacrjournals.org/

  • removed potential consequences of migratory and prolifer-ative stimuli, which have previously been shown to separatetumorfibroblasts fromnormal (28, 29). Greater than 89% of thedifferentially expressed genes separating UV cSCC and RDEBskin from normal skin are also found in RDEB cSCC and thesechanges increase in a stepwise manner from normal to UVcSCC and RDEB through to RDEB cSCC suggesting a progres-sion in the degree of gene expression changes (Fig. 2B and C). Alarge proportion of the differences between disease andnormalskin are in genes encoding components of, or receptors thatinteract with, the extracellular matrix (Figs. 3 and 4). Thechanges in extracellular matrix correlate with increased adhe-sion and invasion of cSCC keratinocytes, which can be attrib-uted to the lack of functional type VII collagen expression

    (Supplementary Figs. S2 and S3 and Fig. 6). We show thatknockdown of COL7A1 in normal fibroblasts and reexpressionof COL7A1 in RDEB fibroblasts results in similar matrix-asso-ciated changes to those identified by microarray, providing amechanism by which RDEB fibroblasts parallel cancer-asso-ciated fibroblasts (Fig. 5). The functional outcomes of thesematrix changes are clearly showed by the decreased cSCC cellinvasion in 3D organotypic cultures and decreased tumorprogression in xenograft studies upon restoration of type VIIcollagen in RDEB fibroblasts (Figs. 6 and 7). Our proposedmodel is that RDEB skin already mimics aspects of cSCCbecause of the shift in gene expression in skin fibroblastsresulting from absence of type VII collagen, and is furthermodified by the process of repeated tissue injury and

    Figure 7. Expression of full-lengthrecombinant type VII collagen inRDEB fibroblasts retards tumorprogression in vivo, leading toincreased tumor differentiationcompared with RDEB fibroblastsexpressing empty vector control.Female SCID Balb/c mice weresubcutaneously injected in the rightflank with 4 � 106 SCCIC1 cellsmixed with 1 � 106 indicatedfibroblast populations and high-concentration Matrigel (Becton-Dickinson). NHF, normal dermalfibroblasts; RDEB29FC7, type VIIcollagen expressing RDEBfibroblasts; RDEB29FpBabe, vectoronly control RDEB fibroblasts. A,tumors were regularly measured bycaliper and individual volume wascalculated using the formula V ¼ p4/3[(L þ W)/4]3 where L is the lengthand W is the width. Graph showsaverage�SEM. �,P

  • inflammation, leading to cancer initiating changes in thekeratinocytes that rapidly progress in the absence of a normalinhibitory dermal architecture. Decreased tumor cell differen-tiation in the presence of RDEB fibroblasts compared withnormal or RDEB fibroblasts expressing full-length type VIIcollagen further supports this (Fig. 7C).

    Tumor stromal cells are said to be "activated", being moreproliferative and secreting more extracellular matrix proteinssuch as type I collagen and fibronectin (25). Here we havefocused on differences in extracellular matrix and adhesionprotein expression, and show that types V and XII collagens aswell as integrin subunits alpha 3 and alpha 6 and thrombos-pondin-1 are upregulated in RDEBF and UVSCCF fibroblastsand further upregulated in RDEBSCCF fibroblasts. Types V andXII collagens are reported to have roles in cellular adhesion andmigration (30, 31) and the increased fibroblast-mediated con-traction of type V collagen–containing gels is integrin depen-dent (32) in agreement with integrin a3–dependent increasedSCC invasion into fibroblast remodeled gels (16). Type XIIcollagen is a FACIT collagen (Fibril Associated Collagens withInterrupted Triple helices), which does not aggregate to formfibrils but instead binds in a periodic manner to the surface offibrils formed by fibrillar collagens (such as type I collagen;ref. 33). How an upregulation of FACIT type XII collagen altersthe cSCC tumormicroenvironment is not yet known. Studies inv-myc or v-src transformed avian fibroblasts and prostatecancer cells have indicated downregulation of type XII collagen(34, 35), although more recently type XII collagen has beenreported to be highly expressed by cancer-associated fibro-blasts in colon cancer (36), suggesting that cancer-associatedtype XII collagen expression is context dependent. Thrombos-pondin upregulation in the stroma of esophageal adenocarci-noma identifies patients with poor prognosis (37) and a recentgene expression study correlating stromal activation withmetastasis in head and neck SCC identified COL5A1 and TSP1as the top 2 differentially expressed genes (38) supporting theirclinical relevance in other aggressive cancers.

    We show through both siRNA knockdown and reexpressionof COL7A1 that, as with normal breast epithelial cells (39), thecomposition of the extracellular matrix directly influences thecomposition of cell-secreted extracellular matrix. Moreover,we show that the expression of type VII collagen directlyinfluences the extracellularmatrix secreted by fibroblasts, datathat are not mirrored in keratinocytes. Our previous ana-lysis of RDEB and non-RDEB tumor and normal keratinocytesdid not identify changes in extracellular matrix gene expres-sion (15) and knockdown of COL7A1 in UV cSCC keratinocytesinduced cytokine rather than extracellularmatrix changes (40).

    A normal tissue microenvironment is protective and sup-presses the development of cSCC (41). Moreover, inhibiting theinteractions between tumor cells and their surroundingmatrixis effective at arresting or delaying tumor progression (42). InRDEB, there seems to be a situation in which the compositionof thematrix per se is tumor promoting or tumor permissive inthe epidermis. Certainly our in vivo data would support thisnotion. Additional contributions from other well-defined pro-cesses in tumor progression and wound healing such asinflammation (43) and/or tumor stroma cross talk (44) need

    to be further investigated. The clinical behavior of cSCC inRDEB also differs from UV cSCC. In RDEB, the majority oftumors are well- or moderately-differentiated, yet surgicalexcision with clear tumor excision margins does not lead toa cure (unlike the vast majority of UV cSCCs treated similarly).Thus, a tumor that would normally be considered lessaggressive in a non-RDEB setting frequently recurs in RDEBpatients, often with further multiple primary cSCCs that areless well-differentiated and that can metastasize and becomefatal (45). RDEB cSCC usually occur in areas of chronic scarringand nonhealing ulceration, correlating positively with theextent of scarring, clinical observations that support a tumorpromoting or permissive stromal microenvironment in thepathophysiology of RDEB cSCC (4). Moreover, no other pri-mary epithelial tumors have been reported to frequentlymanifest in RDEB patients (4) even though COL7A1 isexpressed in other tissues (46) and extracutaneous/mucosalsymptoms occur, especially in more severe RDEB subtypes(47). These observations would support the hypothesis thatCOL7A1 has a more direct role in epidermal homeostasis thanin other epithelial tissues, arguing against a systemic tumorpromoting role and agreeing with the recent propositionthat factors that modulate malignant transformation throughthe tumor microenvironment are likely tissue-dependent (48).Certainly, the context-dependent nature of the expression ofCOL7A1 and type VII collagen across a breadth of differentcancers would support this idea (5).

    COL7A1 is mutated in RDEB fibroblasts yet similar changesin matrix composition were observed in UV cSCC fibroblaststhat, compared with normal fibroblasts, express increasedlevels of wild-type COL7A1 and type VII collagen (Supplemen-tary Fig. S4). This and our knockdown experiments wouldsuggest that convergent evolution of the overall matrix com-position of UV cSCC fibroblasts and RDEB fibroblasts, ratherthan simply reduced COL7A1 in UV cSCC fibroblasts, isresponsible for these changes in vivo. Therefore, it is likelythat these tumor-promoting matrix changes can arise throughother processes, as evidenced in other cancers (discussedearlier). We cannot rule out that the common environmentof inflammation is responsible for a proportion of the expres-sion differences we identify and further work will be necessaryto explore this likely possibility. The large proportion of geneexpression changes unique to RDEB cancer-associated fibro-blasts have not been the focus of this study andwill be followedup separately. It is important to note that themajority ofmatrixgenes we identify here are either direct or indirect targets ofTGF-b signaling, COL7A1 included (49). As TGF-b signalingdirects cellular processes in a context-dependent manner (50)this pathway could have important bearing on the switch fromnormal to a cancer-associated fibroblast behavior.

    In summary, our data suggest that dermal composition iscancer predisposing in patients with RDEB that, rather thansuppressing tumor development, is permissive to the progres-sion toward metastatic disease. Downregulation of COL7A1switches gene expression in dermal fibroblasts toward a can-cer-associated fibroblast phenotype. This cancer-associatedfibroblast phenotype in turn promotes substrate adhesion andinvasion of tumor keratinocytes and tumor progression in vivo.

    Ng et al.

    Cancer Res; 72(14) July 15, 2012 Cancer Research3532

    on April 4, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 7, 2012; DOI: 10.1158/0008-5472.CAN-11-2996

    http://cancerres.aacrjournals.org/

  • By understanding the underlying mechanisms behind theaggressive nature of cSCC in RDEB patients, more effectivetherapies that target the dermal architecture and micro-environment may help in combating this devastating compli-cation to an already severe disease and may also translate toaggressive cSCC and other epithelial cancers in the generalpopulation.

    Disclosure of Potential Conflicts InterestI.M. Leigh has Employment (other than primary affiliation; e.g., consulting)

    in i-pri as Research Director. No potential conflicts of interest were disclosed bythe other authors.

    Authors' ContributionsConception and design: Y.Z. Ng, I.M. Leigh, A.P. SouthDevelopment of methodology: Y.Z. Ng, J.-D. Fine, A.P. SouthAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): Y.Z. Ng, C. Pourreyron, J.C. Salas-Alanis, J.H.S. Dayal,R. Cepeda-Valdes, J.-D. Fine, F.J. Hogg, J.A. McGrath, D.F. Murrell, I.M. Leigh, E.B.Lane, A.P. SouthAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): Y.Z. Ng, A.P. South

    Writing, review, and/or revision of the manuscript: Y.Z. Ng, J.C. Salas-Alanis,W. Yan,M. Chen, J.-D. Fine, F.J. Hogg, J.A.McGrath, D.F.Murrell, E.B. Lane,A.P. SouthAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): Y.Z. Ng, J.C. Salas-Alanis, R. Cepeda-Valdes, W. Yan, S. Wright, M. Chen, D.F. Murrell, I.M. LeighStudy supervision: C. Pourreyron, E.B. Lane, A.P. South

    AcknowledgmentsThe authors thank John Lim (microLAMBDA, Singapore), Xin Mao (QMUL,

    UK), and the Histopathology and Rodent Necropsy Facility at IMCB, A�Star,Singapore, for technical assistance.

    Grant SupportThis work was funded by DebRA, the dystrophic epidermolysis bullosa

    research association (http://www.debra.org.uk/), and the European ResearchCouncil.

    The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicate thisfact.

    Received September 5, 2011; revised April 4, 2012; accepted April 20, 2012;published OnlineFirst May 7, 2012.

    References1. ChristianoAM,GreenspanDS,HoffmanGG,ZhangX, TamaiY, Lin AN,

    et al. A missense mutation in type VII collagen in two affected siblingswith recessive dystrophic epidermolysis bullosa. Nat Genet 1993;4:62–6.

    2. Burgeson RE. Type VII collagen, anchoring fibrils, and epidermolysisbullosa. J Invest Dermatol 1993;101:252–5.

    3. Fine JD, Eady RA, Bauer EA, Bauer JW, Bruckner-Tuderman L,Heagerty A, et al. The classification of inherited epidermolysis bullosa(EB): report of the Third International ConsensusMeeting onDiagnosisand Classification of EB. J Am Acad Dermatol 2008;58:931–50.

    4. Fine JD, Johnson LB, Weiner M, Li KP, Suchindran C. Epidermolysisbullosa and the risk of life-threatening cancers: the National EBRegistry experience, 1986–2006. J Am Acad Dermatol 2009;60:203–11.

    5. South AP, O'Toole EA. Understanding the pathogenesis of recessivedystrophic epidermolysis bullosa squamous cell carcinoma. DermatolClin 2009;28:171–8.

    6. Venugopal SS, Murrell DF. Treatment of skin cancers in epidermolysisbullosa. Dermatol Clin 2010;28:283–7, ix-x.

    7. Arbiser JL, FanCY,SuX, VanEmburghBO,Cerimele F,MillerMS, et al.Involvement of p53 and p16 tumor suppressor genes in recessivedystrophic epidermolysis bullosa-associated squamous cell carcino-ma. J Invest Dermatol 2004;123:788–90.

    8. Arbiser JL, Fine JD, Murrell D, Paller A, Connors S, Keough K, et al.Basic fibroblast growth factor: a missing link between collagen VII,increased collagenase, and squamous cell carcinoma in recessivedystrophic epidermolysis bullosa. Mol Med 1998;4:191–5.

    9. Cooper HL, Cook IS, Theaker JM,Mallipeddi R, McGrath J, FriedmannP, et al. Expression and glycosylation of MUC1 in epidermolysisbullosa-associated and sporadic cutaneous squamous cell carcino-mas. Br J Dermatol 2004;151:540–5.

    10. Mallipeddi R,Wessagowit V, SouthAP,RobsonAM,OrchardGE, EadyRA, et al. Reduced expression of insulin-like growth factor-bindingprotein-3 (IGFBP-3) in squamous cell carcinoma complicating reces-sive dystrophic epidermolysis bullosa. J Invest Dermatol 2004;122:1302–9.

    11. Pourreyron C, Cox G, Mao X, Volz A, Baksh N, Wong T, et al. Patientswith recessive dystrophic epidermolysis bullosa develop squamous-cell carcinoma regardless of type VII collagen expression. J InvestDermatol 2007;127:2438–44.

    12. Purdie KJ, Pourreyron C, Fassihi H, Cepeda-Valdes R, Frew JW, VolzA, et al. No evidence that human papillomavirus is responsible for theaggressive nature of recessive dystrophic epidermolysis bullosa-

    associated squamous cell carcinoma. J Invest Dermatol 2010;130:2853–5.

    13. Smoller BA, McNutt NS, Carter DM, Gottlieb AB, Hsu A, Krueger J.Recessive dystrophic epidermolysis bullosa skin displays a chronicgrowth-activated immunophenotype: implications for carcinogenesis.Arch Dermatol 1990;126:78–83.

    14. Tyring SK, Chopra V, Johnson L, Fine JD. Natural killer cell activity isreduced in patients with severe forms of inherited epidermolysisbullosa. Arch Dermatol 1989;125:797–800.

    15. Watt SA, Pourreyron C, Purdie K, Hogan C, Cole CL, Foster N, et al.Integrative mRNA profiling comparing cultured primary cells withclinical samples reveals PLK1 and C20orf20 as therapeutic targetsin cutaneous squamous cell carcinoma. Oncogene 2011;30:4666–77.

    16. Gaggioli C, Hooper S, Hidalgo-Carcedo C, Grosse R, Marshall JF,HarringtonK, et al. Fibroblast-led collective invasionof carcinomacellswith differing roles for RhoGTPases in leading and following cells. NatCell Biol 2007;9:1392–400.

    17. Bhowmick NA, Chytil A, Plieth D, Gorska AE, Dumont N, Shappell S,et al. TGF-beta signaling in fibroblasts modulates the oncogenicpotential of adjacent epithelia. Science 2004;303:848–51.

    18. Erez N, Truitt M, Olson P, Arron ST, Hanahan D. Cancer-associatedfibroblasts are activated in incipient neoplasia to orchestrate tumor-promoting inflammation in an NF-kappaB-dependent manner. CancerCell 2010;17:135–47.

    19. Camps JL, Chang SM, Hsu TC, FreemanMR, Hong SJ, Zhau HE, et al.Fibroblast-mediated acceleration of human epithelial tumor growth invivo. Proc Natl Acad Sci U S A 1990;87:75–9.

    20. Arbeit JM, Olson DC, Hanahan D. Upregulation of fibroblast growthfactors and their receptors during multi-stage epidermal carcinogen-esis in K14-HPV16 transgenic mice. Oncogene 1996;13:1847–57.

    21. Edgar R, Domrachev M, Lash AE. Gene Expression Omnibus: NCBIgene expression and hybridization array data repository. Nucleic AcidsRes 2002;30:207–10.

    22. Larouche D, Paquet C, Fradette J, Carrier P, Auger FA, Germain L.Regeneration of skin and cornea by tissue engineering. Methods MolBiol 2009;482:233–56.

    23. Beer TW, Shepherd P, Theaker JM. Ber EP4 and epithelial membraneantigen aid distinction of basal cell, squamous cell and basosquamouscarcinomas of the skin. Histopathology 2000;37:218–23.

    24. Sugimoto H, Mundel TM, Kieran MW, Kalluri R. Identification offibroblast heterogeneity in the tumor microenvironment. Cancer BiolTher 2006;5:1640–6.

    Dermal Matrix Composition Promotes RDEB Cancer

    www.aacrjournals.org Cancer Res; 72(14) July 15, 2012 3533

    on April 4, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 7, 2012; DOI: 10.1158/0008-5472.CAN-11-2996

    http://cancerres.aacrjournals.org/

  • 25. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer 2006;6:392–401.

    26. Chang HY, Sneddon JB, Alizadeh AA, Sood R, West RB, MontgomeryK, et al. Gene expression signature of fibroblast serum responsepredicts human cancer progression: similarities between tumors andwounds. PLoS Biol 2004;2:E7.

    27. Chang HY, Chi JT, Dudoit S, Bondre C, van de RijnM, Botstein D, et al.Diversity, topographic differentiation, and positional memory in humanfibroblasts. Proc Natl Acad Sci U S A 2002;99:12877–82.

    28. Schor SL, Schor AM, Durning P, Rushton G. Skin fibroblasts obtainedfrom cancer patients display foetal-like migratory behaviour on colla-gen gels. J Cell Sci 1985;73:235–44.

    29. Commandeur S, Ho SH, de Gruijl FR, Willemze R, Tensen CP, ElGhalbzouri A. Functional characterization of cancer-associated fibro-blasts of human cutaneous squamous cell carcinoma. Exp Dermatol2011;20:737–42.

    30. Breuls RG, Klumpers DD, Everts V, Smit TH. Collagen type V mod-ulates fibroblast behavior dependent on substrate stiffness. BiochemBiophys Res Commun 2009;380:425–9.

    31. Izu Y, Sun M, Zwolanek D, Veit G, Williams V, Cha B, et al. Type XIIcollagen regulatesosteoblast polarity andcommunicationduringboneformation. J Cell Biol 2011;193:1115–30.

    32. Berendsen AD, Bronckers AL, Smit TH, Walboomers XF, Everts V.Collagen type V enhances matrix contraction by human periodontalligament fibroblasts seeded in three-dimensional collagen gels. MatrixBiol 2006;25:515–22.

    33. Ricard-Blum S, Ruggiero F. The collagen superfamily: from the extra-cellular matrix to the cell membrane. Pathol Biol (Paris) 2005;53:430–42.

    34. Kopp MU, Croci LP, Trueb B. Down-regulation of collagen XII intransformed mesenchymal cells. Int J Cancer 1995;60:275–9.

    35. Wang J, Levenson AS, Satcher RL Jr. Identification of a unique set ofgenes altered during cell-cell contact in an in vitro model of prostatecancer bone metastasis. Int J Mol Med 2006;17:849–56.

    36. Karagiannis GS, Petraki C, Prassas I, Saraon P, Musrap N, Dimitro-manolakis A, et al. Proteomic signatures of the desmoplastic invasionfront reveal collagen type XII as a marker of myofibroblastic differen-tiation during colorectal cancer metastasis. Oncotarget 2012;3:267–85.

    37. Saadi A, ShannonNB, Lao-Sirieix P,O'DonovanM,Walker E, ClemonsNJ, et al. Stromal genes discriminate preinvasive from invasive dis-

    ease, predict outcome, and highlight inflammatory pathways in diges-tive cancers. Proc Natl Acad Sci U S A 2010;107:2177–82.

    38. Roepman P, de Koning E, van Leenen D, de Weger RA, Kummer JA,Slootweg PJ, et al. Dissection of a metastatic gene expression sig-nature into distinct components. Genome Biol 2006;7:R117.

    39. Streuli CH, Bissell MJ. Expression of extracellular matrix componentsis regulated by substratum. J Cell Biol 1990;110:1405–15.

    40. Martins VL, Vyas JJ, Chen M, Purdie K, Mein CA, South AP, et al.Increased invasive behaviour in cutaneous squamous cell carcinomawith loss of basement-membrane type VII collagen. J Cell Sci2009;122:1788–99.

    41. Javaherian A, Vaccariello M, Fusenig NE, Garlick JA. Normal kerati-nocytes suppress early stages of neoplastic progression in stratifiedepithelium. Cancer Res 1998;58:2200–8.

    42. Weaver VM, Petersen OW, Wang F, Larabell CA, Briand P, Damsky C,et al. Reversion of the malignant phenotype of human breast cells inthree-dimensional culture and in vivo by integrin blocking antibodies. JCell Biol 1997;137:231–45.

    43. DeNardoDG, JohanssonM,Coussens LM. Immune cells asmediatorsof solid tumor metastasis. Cancer Metastasis Rev 2008;27:11–8.

    44. Wallace JA, Li F, Leone G, Ostrowski MC. Pten in the breast tumormicroenvironment: modeling tumor-stroma coevolution. Cancer Res2011;71:1203–7.

    45. Mallipeddi R. Epidermolysis bullosa and cancer. Clin Exp Dermatol2002;27:616–23.

    46. Wetzels RH, RobbenHC, Leigh IM, SchaafsmaHE, Vooijs GP, Ramae-kers FC. Distribution patterns of type VII collagen in normal andmalignant human tissues. Am J Pathol 1991;139:451–9.

    47. Fine JD, Mellerio JE. Extracutaneous manifestations and complica-tions of inherited epidermolysis bullosa: part II: other organs. J AmAcad Dermatol 2009;61:387–402; quiz 403–4.

    48. Bissell MJ, HinesWC.Whydon't we getmore cancer? Aproposed roleof the microenvironment in restraining cancer progression. Nat Med2011;17:320–9.

    49. Vindevoghel L, Lechleider RJ, Kon A, de Caestecker MP, Uitto J,Roberts AB, et al. SMAD3/4-dependent transcriptional activationof the human type VII collagen gene (COL7A1) promoter by trans-forming growth factor beta. Proc Natl Acad Sci U S A 1998;95:14769–74.

    50. Inman GJ. Switching TGFbeta from a tumor suppressor to a tumorpromoter. Curr Opin Genet Dev 2011;21:93–9.

    Ng et al.

    Cancer Res; 72(14) July 15, 2012 Cancer Research3534

    on April 4, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 7, 2012; DOI: 10.1158/0008-5472.CAN-11-2996

    http://cancerres.aacrjournals.org/

  • 2012;72:3522-3534. Published OnlineFirst May 7, 2012.Cancer Res Yi-Zhen Ng, Celine Pourreyron, Julio C. Salas-Alanis, et al. Recessive Dystrophic Epidermolysis BullosaAggressive Cutaneous Squamous Cell Carcinoma in Patients with Fibroblast-Derived Dermal Matrix Drives Development of

    Updated version

    10.1158/0008-5472.CAN-11-2996doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://cancerres.aacrjournals.org/content/suppl/2012/05/07/0008-5472.CAN-11-2996.DC1

    Access the most recent supplemental material at:

    Cited articles

    http://cancerres.aacrjournals.org/content/72/14/3522.full#ref-list-1

    This article cites 48 articles, 12 of which you can access for free at:

    Citing articles

    http://cancerres.aacrjournals.org/content/72/14/3522.full#related-urls

    This article has been cited by 4 HighWire-hosted articles. Access the articles at:

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected]

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://cancerres.aacrjournals.org/content/72/14/3522To request permission to re-use all or part of this article, use this link

    on April 4, 2021. © 2012 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 7, 2012; DOI: 10.1158/0008-5472.CAN-11-2996

    http://cancerres.aacrjournals.org/lookup/doi/10.1158/0008-5472.CAN-11-2996http://cancerres.aacrjournals.org/content/suppl/2012/05/07/0008-5472.CAN-11-2996.DC1http://cancerres.aacrjournals.org/content/72/14/3522.full#ref-list-1http://cancerres.aacrjournals.org/content/72/14/3522.full#related-urlshttp://cancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://cancerres.aacrjournals.org/content/72/14/3522http://cancerres.aacrjournals.org/

Recommended