The Differential Role of L1 in Ovarian Carcinoma and Normal Ovarian Surface Epithelium

10
The Differential Role of L1 in Ovarian Carcinoma and Normal Ovarian Surface Epithelium Silvia Zecchini, 1 Marco Bianchi, 1 Nicoletta Colombo, 2 Roberta Fasani, 2 Giovanni Goisis, 2 Chiara Casadio, 2 Giuseppe Viale, 2,3 Jinsong Liu, 4 Meenhard Herlyn, 5 Andrew K. Godwin, 6 Paolo G. Nuciforo, 1 and Ugo Cavallaro 1 1 FIRC Institute of Molecular Oncology, 2 European Institute of Oncology, and 3 University of Milano School of Medicine, Milano, Italy; 4 Department of Pathology, The University of Texas M. D. Anderson Cancer, Houston, Texas; 5 Wistar Institute and 6 Fox Chase Cancer Center, Philadelphia, Pennsylvania Abstract Epithelial ovarian carcinoma (EOC) arises from the ovarian surface epithelium (OSE), a monolayer of poorly differentiated epithelial cells that lines the ovary. The molecular mecha- nisms underlying EOC invasion into the surrounding stroma and dissemination to the peritoneum and to retroperitoneal lymph nodes are still unclear. Here, we analyzed the expression and the functional role of the cell adhesion molecule L1 during EOC development. In patient-derived samples, L1 was expressed both in OSE and in a subset of EOC, in the latter being mostly restricted to the invasive areas of the tumors. The expression of L1 correlated significantly with poor outcome and with unfavorable clinicopathologic features of the disease. The peculiar expression pattern of L1 in normal OSE and invasive EOC raised the possibility that this adhesion molecule serves a different function in nontransformed versus neoplastic ovarian epithelial cells. Indeed, we showed that in OSE cells L1 supports cell-cell adhesion and enhances apoptosis, whereas it has no effect on cell proliferation and invasion. In contrast, L1 inhibits cell-cell adhesion and apoptosis in ovarian carcinoma cells, where it promotes malignancy-related properties, such as cell proliferation, Erk1/2-dependent and phosphoinositide 3-kinase–dependent invasion, and transendothelial migration. Interestingly, a crosstalk with the fibroblast growth factor receptor signaling is implicated in the promalignant function of L1 in tumor cells. Our findings point to L1 as an EOC biomarker correlating with poor prognosis, and highlight a switch in L1 function associated to the neoplastic transformation of ovarian epithelial cells, thus implicating L1 as a potential therapeutic target. [Cancer Res 2008;68(4):1110–8] Introduction Epithelial ovarian carcinoma (EOC) is the leading cause of mortality among gynecological malignancies in the Western world. Due to the lack of clinically relevant symptoms during the early phases of cancer development, the diagnosis is frequently made at advanced stages of the disease. Surgical cytoreduction and chemotherapy usually induce tumor regression; however, most patients undergo a relapse of the disease with a high mortality rate (1). Although a number of genetic lesions associated to higher risk for specific forms of EOC have been identified, only very few biomarkers that contribute to the early diagnosis and prognosis of the disease are currently available, accounting for the difficult management of this tumor type. Moreover, the molecular mechanisms that control the onset and progression of EOC have remained elusive, and their definition would certainly open novel therapeutic avenues for the treatment of such a life-threatening disease. Cell adhesion molecules (CAM) are cell surface proteins mediating cell-cell and cell-matrix interactions. Alterations in CAM expression and/or function have been implicated in the development of various tumor types. In particular, the dysregu- lation of cell-cell adhesion caused by changes in the levels of cadherins and/or immunoglobulin-like CAMs (Ig-CAM) plays a causal role in the progression of several epithelial tumors (2). The Ig-CAM L1 (also known as L1CAM or CD171) has been extensively characterized in the nervous system, where it mediates neuronal adhesion and migration, as well as axon pathfinding and fasciculation (3). However, L1 is also expressed in nonneuronal tissues, including certain epithelia and some hematopoietic lineages, where its function remains elusive. Recent studies have reported an aberrant expression of L1 in various tumor types, including melanoma (4) and colon carcinoma (5). In the latter, L1 has been characterized as a transcriptional target of the Wnt/h-catenin pathway and as a factor involved in tumor cell invasion (5). Finally, an aberrant expression of L1 has been described in advanced EOC and has been proposed to enhance the malignant phenotype of EOC cells (6, 7). Here, we report the first demonstration that the expression of L1 is not restricted to EOC but is also abundant in the ovarian surface epithelium (OSE), the monolayer of poorly differentiated epithelial cells lining the ovaries that, upon neoplastic transformation, gives rise to most EOC forms (8). Furthermore, in agreement with previous reports, our analysis of clinical specimens revealed that the expression of L1 correlates with poor prognosis in EOC patients. The expression pattern of L1 and its association with specific clinicopathologic features of the disease suggested that this adhesion molecule serves a different function in normal versus neoplastic OSE. To verify this hypothesis, we have characterized the biological activities of L1 in OSE and in EOC cell lines. Our results indicated that L1 plays markedly different, and often opposite, roles in nontumorigenic versus transformed ovarian epithelial cells. These findings underscore the importance of the cellular context in dictating the functional role of L1 in OSE cells and provide the rationale Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Requests for reprints: Ugo Cavallaro, FIRC Institute of Molecular Oncology, via Adamello 16, I-20139 Milano, Italy. Phone: 39-02-574303-224; Fax: 39-02-574303-244; E-mail: [email protected]. I2008 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-07-2897 Cancer Res 2008; 68: (4). February 15, 2008 1110 www.aacrjournals.org Research Article Research. on January 19, 2016. © 2008 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Transcript of The Differential Role of L1 in Ovarian Carcinoma and Normal Ovarian Surface Epithelium

The Differential Role of L1 in Ovarian Carcinoma and Normal

Ovarian Surface Epithelium

Silvia Zecchini,1Marco Bianchi,

1Nicoletta Colombo,

2Roberta Fasani,

2Giovanni Goisis,

2

Chiara Casadio,2Giuseppe Viale,

2,3Jinsong Liu,

4Meenhard Herlyn,

5Andrew K. Godwin,

6

Paolo G. Nuciforo,1and Ugo Cavallaro

1

1FIRC Institute of Molecular Oncology, 2European Institute of Oncology, and 3University of Milano School of Medicine, Milano, Italy;4Department of Pathology, The University of Texas M. D. Anderson Cancer, Houston, Texas; 5Wistar Institute and 6Fox ChaseCancer Center, Philadelphia, Pennsylvania

Abstract

Epithelial ovarian carcinoma (EOC) arises from the ovariansurface epithelium (OSE), a monolayer of poorly differentiatedepithelial cells that lines the ovary. The molecular mecha-nisms underlying EOC invasion into the surrounding stromaand dissemination to the peritoneum and to retroperitoneallymph nodes are still unclear. Here, we analyzed theexpression and the functional role of the cell adhesionmolecule L1 during EOC development. In patient-derivedsamples, L1 was expressed both in OSE and in a subset of EOC,in the latter being mostly restricted to the invasive areas of thetumors. The expression of L1 correlated significantly withpoor outcome and with unfavorable clinicopathologic featuresof the disease. The peculiar expression pattern of L1 in normalOSE and invasive EOC raised the possibility that this adhesionmolecule serves a different function in nontransformed versusneoplastic ovarian epithelial cells. Indeed, we showed that inOSE cells L1 supports cell-cell adhesion and enhancesapoptosis, whereas it has no effect on cell proliferation andinvasion. In contrast, L1 inhibits cell-cell adhesion andapoptosis in ovarian carcinoma cells, where it promotesmalignancy-related properties, such as cell proliferation,Erk1/2-dependent and phosphoinositide 3-kinase–dependentinvasion, and transendothelial migration. Interestingly, acrosstalk with the fibroblast growth factor receptor signalingis implicated in the promalignant function of L1 in tumorcells. Our findings point to L1 as an EOC biomarkercorrelating with poor prognosis, and highlight a switch in L1function associated to the neoplastic transformation ofovarian epithelial cells, thus implicating L1 as a potentialtherapeutic target. [Cancer Res 2008;68(4):1110–8]

Introduction

Epithelial ovarian carcinoma (EOC) is the leading cause ofmortality among gynecological malignancies in the Westernworld. Due to the lack of clinically relevant symptoms duringthe early phases of cancer development, the diagnosis isfrequently made at advanced stages of the disease. Surgical

cytoreduction and chemotherapy usually induce tumor regression;however, most patients undergo a relapse of the disease with ahigh mortality rate (1). Although a number of genetic lesionsassociated to higher risk for specific forms of EOC have beenidentified, only very few biomarkers that contribute to the earlydiagnosis and prognosis of the disease are currently available,accounting for the difficult management of this tumor type.Moreover, the molecular mechanisms that control the onset andprogression of EOC have remained elusive, and their definitionwould certainly open novel therapeutic avenues for the treatment ofsuch a life-threatening disease.Cell adhesion molecules (CAM) are cell surface proteins

mediating cell-cell and cell-matrix interactions. Alterations inCAM expression and/or function have been implicated in thedevelopment of various tumor types. In particular, the dysregu-lation of cell-cell adhesion caused by changes in the levels ofcadherins and/or immunoglobulin-like CAMs (Ig-CAM) plays acausal role in the progression of several epithelial tumors (2).The Ig-CAM L1 (also known as L1CAM or CD171) has beenextensively characterized in the nervous system, where itmediates neuronal adhesion and migration, as well as axonpathfinding and fasciculation (3). However, L1 is also expressedin nonneuronal tissues, including certain epithelia and somehematopoietic lineages, where its function remains elusive.Recent studies have reported an aberrant expression of L1 invarious tumor types, including melanoma (4) and coloncarcinoma (5). In the latter, L1 has been characterized as atranscriptional target of the Wnt/h-catenin pathway and as afactor involved in tumor cell invasion (5). Finally, an aberrantexpression of L1 has been described in advanced EOC and hasbeen proposed to enhance the malignant phenotype of EOC cells(6, 7). Here, we report the first demonstration that theexpression of L1 is not restricted to EOC but is also abundantin the ovarian surface epithelium (OSE), the monolayer of poorlydifferentiated epithelial cells lining the ovaries that, uponneoplastic transformation, gives rise to most EOC forms (8).Furthermore, in agreement with previous reports, our analysis ofclinical specimens revealed that the expression of L1 correlateswith poor prognosis in EOC patients. The expression pattern ofL1 and its association with specific clinicopathologic features ofthe disease suggested that this adhesion molecule serves adifferent function in normal versus neoplastic OSE. To verify thishypothesis, we have characterized the biological activities of L1in OSE and in EOC cell lines. Our results indicated that L1 playsmarkedly different, and often opposite, roles in nontumorigenicversus transformed ovarian epithelial cells. These findingsunderscore the importance of the cellular context in dictatingthe functional role of L1 in OSE cells and provide the rationale

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

Requests for reprints: Ugo Cavallaro, FIRC Institute of Molecular Oncology, viaAdamello 16, I-20139 Milano, Italy. Phone: 39-02-574303-224; Fax: 39-02-574303-244;E-mail: [email protected].

I2008 American Association for Cancer Research.doi:10.1158/0008-5472.CAN-07-2897

Cancer Res 2008; 68: (4). February 15, 2008 1110 www.aacrjournals.org

Research Article

Research. on January 19, 2016. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from

for evaluating L1 both as a EOC biomarker and as a novel targetfor the molecular therapy of ovarian carcinoma.

Materials and Methods

Selection of the patients, tissue microarray analysis, and dataanalysis. Clinicopathologic and follow-up data of ovarian cancer patients

surgically treated at the European Institute of Oncology from 1995 to 2004

were used to select the cases that were included in the study. Inclusion

criteria were (a) first surgery at the European Institute of Oncology, (b) noneoadjuvant treatment, and (c) common epithelial histologic subtypes

(serous, mucinous, and endometrioid cystoadenocarcinomas).

For clinicopathologic details of the patients, construction of tissue

microarrays, and data analysis, see Supplementary Materials and Methods.Cell culture. Human ovarian carcinoma cell lines IGROV1 and OVCAR3

were from American Type Culture Collection.

HIO-80 cells (called HIOSE/A; ref. 9) and IOSE80 cells (called HIOSE/B;ref. 10) were derived from primary OSE cells transfected with SV40 large

T antigen.

For further details on culture conditions, adenoviral infection and small

interfering RNA (siRNA) transfection, and additional cell lines used in thisstudy, see Supplementary Materials and Methods.

For details on antibodies and chemicals, biochemical methods, cell

biological assays, and immunostaining procedures, see Supplementary

Materials and Methods.

Results

The Expression of L1 in Human OSE and EOCThe expression of L1 was investigated in a panel of surgically

resected specimens, including 20 normal ovaries with a clearlydetectable OSE, 4 benign lesions (cystadenoma), 211 primary EOC,and 199 metastatic lesions (see Supplementary Materials andMethods for details on selection and analysis of patients). Theresults are summarized in Fig. 1C and Supplementary Table S2. Theimmunohistochemical staining with the polyclonal antibodypcytL1, directed against the cytoplasmic tail (11), showed highlevels of L1 in 100% of the normal OSE samples, mainly localized atthe intercellular boundaries and at the basolateral surface of thecells (Fig. 1A, left). The identity of L1-positive tissue as OSE wasconfirmed by the positive staining of the same cells for cytokeratins(Supplementary Fig. S1C–E). By immunohistochemistry, benignlesions showed an L1 immunoreactivity similar to OSE (notshown). Because our observations were in contrast with a previousstudy that reported the absence of L1 in normal OSE (6), we usedadditional antibodies (2C2, CE7, and L1-S; see SupplementaryTable S1) that confirmed the L1 immunoreactivity in OSE tissue(Supplementary Fig. S1A and data not shown). Furthermore, L1was also detected in freshly isolated OSE cells where, by analogy tonormal ovary, it was enriched at cell-cell contacts of primary OSEcells (Fig. 1A, right and Supplementary Fig. S1B). The immuno-blotting of OSE cell lysates showed that L1 migrated as a doublet of200 to 220 kDa (Supplementary Fig. S3), where the highermolecular weight band is likely to represent the mature, surface-exposed protein, whereas the lower molecular weight form wouldrepresent the intracellular pool of L1 (12).Ninety primary EOC (43%) and 89 metastases (45%) exhibited a

clear L1 staining (Fig. 1C). However, in contrast to nonneoplasticepithelium (where 100% of OSE cells exhibited L1 staining), thevast majority of cancer cells within these tumor tissues showedno or very little expression of L1 in the tissue cores analyzed[primary EOC, mean 7.89% (0–80); metastatic cancer, mean 7.74(0-100)]. The immunoreactivity of L1, instead, was specifically

maintained in less cohesive cells inside the tumor mass and atthe invasive front of the tumor (Fig. 1B and SupplementaryFig S2B and C). Because this pattern suggested that L1 could beimplicated in loss of cell-cell adhesion and in cancer invasion(and, hence, progression), we asked whether the expression of L1in tumor specimens was associated with the clinicopathologicfeatures of the disease. Indeed, the presence of L1 in primary EOCsignificantly correlated with the poorly differentiated, moreaggressive phenotype of the tumor cells (grade 3, P = 0.034; seeSupplementary Table S2). In addition, tumors characterized byadvanced International Federation of Gynecology and Obstetricsstage (III–IV versus I–II) and by the presence of lymph nodemetastasis exhibited higher L1 levels, although the correlation didnot reach statistical significance (P = 0.064 and 0.057, respective-ly). Kaplan-Meier survival analyses strengthened the prognosticvalue of L1 by showing a significantly shorter 5-year overall (logrank, P = 0.0087) and disease-free (log rank, P = 0.029) survival inpatients with L1-positive tumors (Fig. 1D), thus confirming andextending the results of a previous study performed on a lowernumber of cases (6). To determine if L1 expression was aprognostic factor independent from the other clinicopathologicvariables, we performed a multivariate proportional hazardsmodeling of overall and disease-free survival, including patient’sage, histologic grade, and tumor stage. Importantly, the presenceof L1 was independently associated with shorter 5-year overallsurvival (Fig. 1D, left), whereas the correlation with shorterdisease-free survival, despite showing a clear trend, did not reachstatistical significance (Fig. 1D, right).In summary, our screening of human ovarian samples revealed

that (a) L1 expression is significantly down-regulated in primaryand metastatic EOC compared with normal OSE and (b) theexpression of L1 in ovarian cancer cells correlated with clinico-pathologic features of aggressiveness and with poor outcome.These apparently contradictory results, together with the frequent-ly observed L1 immunoreactivity in less cohesive cells within thetumor mass and at the invasive front of EOC, suggest that a switchin L1 function might occur upon transformation of ovarianepithelial cells.To address the role of neoplastic transformation in L1 expres-

sion, we took advantage of a genetically defined cellular model thatrecapitulates many features of EOC development. This consisted ofOSE cells that were immortalized by sequential transfection withSV40 T antigen (called IOSE29 cells) and human telomerase reversetranscriptase (T29 cells), followed by full transformation withoncogenic H-RasV12 (T29H; ref. 10). By analogy to most cells in theEOC mass, neither IOSE-29 nor T29 cells expressed L1, whereas thelatter was clearly detectable in isogenic, fully transformed T29Hcells (Supplementary Fig. S3). In agreement with the observationsin patient-derived specimens, these findings supported the notionthat the expression of L1 is specifically associated to a highly malig-nant phenotype.

The Functional Role of L1 in Nontransformed versusNeoplastic Ovarian Epithelial CellsCell proliferation. To verify the hypothesis of a dual role of

L1 in normal versus transformed OSE, we used gain-of-functionand loss-of-function strategies in cells derived either fromnormal OSE or from ovarian carcinoma. Primary OSE cells aredifficult to isolate and to maintain in culture, and they rapidlyundergo cellular senescence (13). To overcome this problem, as amodel for nontransformed ovarian epithelium, we used cells

The Dual Role of L1 in Ovarian Epithelial Cells

www.aacrjournals.org 1111 Cancer Res 2008; 68: (4). February 15, 2008

Research. on January 19, 2016. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from

isolated from normal OSE stably transfected with the SV40 earlyregion, containing large T and small t antigens (SV40-TAg). Thisprocedure has been reported to prolong the life span of OSEcells without inducing a tumorigenic phenotype (9, 10, 14, 15). Incontrast with OSE cells in vivo , L1 was undetectable in fourdifferent lines of SV40-TAg–transfected OSE cells (SupplementaryFigs. S3 and S4A), possibly due to the inactivation of L1-regulating transcription factors by SV40-TAg (see Discussion).For the characterization of L1 activity in nontransformed OSEcells, we selected HIO-80 (called HIOSE/A; ref. 9) and IOSE80(called HIOSE/B; ref. 10) cells to perform gain-of-functionstudies. Upon adenoviral transduction with the cDNA for humanL1, HIOSE cells exhibited a remarkable expression of L1 whichwas correctly localized at the cell surface (SupplementaryFig. S4A). To study the role of L1 ovarian carcinoma cells, weselected two cell lines: IGROV1 cells, which express high levels ofL1 (Supplementary Fig. S4B), and OVCAR3 cells, where L1 isalmost undetectable (Supplementary Fig. S4C). The former cellline was transfected with siRNA oligonucleotides that have been

successfully used to reduce L1 expression in human colon cancercells (5). Indeed, the levels of L1 in transfected IGROV1 weredramatically decreased compared with control cells transfectedwith siRNA oligonucleotides targeting the mRNA for greenfluorescent protein (GFP; Supplementary Fig. S4B). Instead,OVCAR3 cells were transduced with adenovirally expressedhuman L1 or with GFP (Supplementary Fig. S4C).We first determined the effect of L1 on serum-induced proli-

feration of HIOSE and IGROV1 cells. After serum starvation, cellswere stimulated with fetal bovine serum (FBS) and cell growthwas determined every 24 h. The forced expression of L1 showedno significant effect on either HIOSE/A or HIOSE/B cells(Fig. 2A, top left), indicating that L1 is not involved in theproliferation of this cell type. In contrast, IGROV1 cells exhibiteda marked reduction in cell proliferation upon abrogation ofL1 expression compared with cells transfected with the siRNAfor GFP (Fig. 2A, top right). The decrease in the proliferation rateof L1-deficient IGROV1 cells was further validated by a reductionin serum-induced DNA replication, as shown by BrdUrd

Figure 1. Expression of L1 in OSE and EOC andcorrelation with patient survival. Normal ovaries (A, left )and EOC tissues (B ) were immunohistochemically stainedwith the polyclonal antibody pcytL1. A, right, primaryOSE cells were subjected to immunofluorescencestaining with the monoclonal antibody CE7. Arrows,accumulation of L1 at the basolateral surface of OSEcells in the normal ovary (A, left ), at the sites of cell-cellcontacts in cultured OSE (A, right ), and at the invasiveedges of EOC in tumor tissues (B). B, arrowheads,indicate noninvasive areas of the tumor with low levelsof L1 (left). The insets in A (left) and B show amagnification of the boxed areas. S , stroma; T , tumor.Magnifications: A, 60�; B, left, 10�; B, right, 40�;A, inset, 100�; B, insets, 60�. C, expression of L1 inpatient-derived samples. D, overall (left) and disease-freesurvival (right ) curves according to L1 expression inprimary ovarian cancer. Hazard ratio (HR ) and 95%confidence intervals (in brackets) obtained from Coxmultivariate regression model before (crude ) and afteradjusting for age, pathologic stage, and tumor grade(adjusted ) are shown. Dotted line, L1-negative tumors;solid line, L1-positive tumors.

Cancer Research

Cancer Res 2008; 68: (4). February 15, 2008 1112 www.aacrjournals.org

Research. on January 19, 2016. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from

incorporation assays (Supplementary Fig. S5). To confirm thatthe inhibitory effect depended on L1, we incubated parentalIGROV1 cells with CE7, a monoclonal antibody that has beenreported to repress the tumorigenicity of ovarian cancer cells inimmunodeficient mice (16). In agreement with the data on L1knockdown cells, the CE7 monoclonal antibody, but not anisotype-matched irrelevant antibody, decreased the proliferationof IGROV1 cells (Fig. 2A, bottom left). As these results indicatedthat L1 is required for EOC cell proliferation, we asked whetherit is also sufficient. To address this question, we determinedthe effect of forcing L1 expression in OVCAR3 cells on theirgrowth capacity after serum stimulation. Indeed, the prolifera-tion rate of OVCAR3-L1 cells was markedly higher than control,GFP-expressing cells (Fig. 2A, bottom right). Overall, thesefindings implicated L1 in the proliferation of neoplastic, butnot normal, OSE cells.To gain insight into the mechanism underlying L1-dependent

ovarian cancer cell proliferation, we focused on the Erk1/2pathway, known to mediate cell proliferation in several exper-imental models. As many transformed cell lines, serum-starvedIGROV1 exhibited constitutive activation of Erk1/2 that, however,

was markedly reduced upon loss of L1 (Fig. 2B, right). Both controland L1 knockdown IGROV1 cells responded to serum stimulationwith an increase in Erk1/2 activation. In contrast, no constitutiveactivation of Erk1/2 was observed in HIOSE cells, and this was notchanged by the forced expression of L1 (Fig. 2B, left). Erk1/2activation in response to serum was retained in both control andL1-transfected HIOSE cells. These findings imply that L1 regulatesthe Erk1/2 signaling pathway in neoplastic, but not normal, ovarianepithelial cells.

Crosstalk of L1 with Fibroblast Growth FactorReceptor SignalingBecause L1 enhanced the growth of serum-stimulated ovarian

carcinoma cells (Figs. 2A and 3A) but was unable to stimulate theproliferation of serum-starved cells (Fig. 3B and C ), it isconceivable that L1 synergizes with a signaling machinery elicitedby growth factors contained in the serum. In an attempt to identifysuch signaling partners, we focused on two growth factor receptorsthat have been previously implicated in L1 function in neurons, thefibroblast growth factor receptor (FGFR) and the epidermal growthfactor receptor (EGFR; refs. 17, 18). Aberrant expression and/or

Figure 2. The effect of L1 on cellproliferation and on Erk1/2 activation.A, cell proliferation in HIOSE (top left ),IGROV1 (top right and bottom left ), orOVCAR3 cells (bottom right ) wasmeasured as described in SupplementaryMaterials and Methods. Bottom left, cellproliferation of IGROV1 cells in thepresence of 20 Ag/mL CE7 antibody orof an isotype-matched, anti-hemagglutinin(HA ) antibody. Points, mean from threeindependent experiments, eachperformed in triplicate; bars, SE.*, P < 0.005, relative to control cells.B, HIOSE/A (left ) or IGROV1 cells (right )were serum-starved and then stimulatedwith 10% FBS for 10 min beforeimmunoblotting for phosphorylated Erk1/2(top ). The blots were stripped andreprobed for total Erk1/2 (bottom ).

The Dual Role of L1 in Ovarian Epithelial Cells

www.aacrjournals.org 1113 Cancer Res 2008; 68: (4). February 15, 2008

Research. on January 19, 2016. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from

activities of both receptors have been associated with ovariancancer malignancy (19–21). Serum-induced proliferation ofOVCAR-3 cells expressing either GFP or L1 was determined aftera pretreatment of the cells with PD173074 or AG1478, whichspecifically inhibit FGFR and EGFR activity, respectively. Interest-ingly, PD173074 specifically abrogated the positive effect of L1on serum-induced proliferation, without affecting the growth ofOVCAR3-GFP cells (Fig. 3A), thus pointing to a functionalcooperation between L1 and FGFR. In contrast, the growth ofboth GFP-expressing and L1-expressing OVCAR-3 cells wasreduced by AG1478 to a very low level (Supplementary Fig. S6A),implicating EGFR in the proliferative capacity of OVCAR-3 cells butruling out any specific effect on L1-dependent proliferation. Toconfirm the interplay between L1 and FGFR, serum-starvedOVCAR3-GFP or OVCAR3-L1 cells were stimulated with FGF-2and subjected to cell proliferation assay. Notably, only L1-expressing cells exhibited a remarkable proliferative response toFGF-2 (Fig. 3B), thus confirming that L1 cooperates with FGFRsignaling. In agreement with the data on AG1478, EGF stimulatedthe proliferation of both OVCAR3-GFP and OVCAR3-L1 cells to thesame extent (Supplementary Fig. S6B), implying the lack of acrosstalk between L1 and EGFR. The role of L1 in FGFR activitywas also confirmed in IGROV1 cells by a loss-of-function approach.Indeed, the abrogation of L1 expression in these cells led to amarked decrease in FGF-induced proliferation, compared with cellstransfected with a control siRNA (Fig. 3C).

Apoptosis ResistancePrevious work implicated L1 in the protection of ovarian

carcinoma cells from apoptosis (22). To verify whether thisantiapoptotic response occurs also in nontransformed OSE cells,we compared the effect of the drug staurosporine, which inducescaspase-mediated apoptosis, in HIOSE and IGROV1 cells. Theactivation of caspase-3 after staurosporine treatment, a widelyused readout for apoptosis, was monitored by immunoblottingwith an antibody specific for the activated (i.e., cleaved) form ofcaspase-3. Staurosporine induced caspase-3 cleavage in L1-negative HIOSE cells with low efficiency, with maximal effect at2 h of treatment in HIOSE/A and at 4 h in HIOSE/B cells.However, upon expression of L1, the proapoptotic effect ofstaurosporine increased remarkably in both cell lines (Supple-mentary Fig. S7). In contrast, L1 counteracted staurosporine-induced apoptosis in IGROV1 cells. Indeed, whereas control cellsshowed no caspase-3 cleavage during a 4-h treatment, cleavedcaspase-3 in response to staurosporine became evident uponsiRNA-mediated down-regulation of L1 (SupplementaryFig. S7). Thus, L1 acted as a survival factor in EOC cells,confirming and extending previous observations (22), while itenhanced drug-induced apoptosis in nontransformed OSE cells.

Cell-Cell and Cell-Matrix AdhesionL1 has been originally characterized as an adhesion molecule that

promotes the physical interaction between adjacent cells. Inaddition, cell-cell adhesion has long been known to affect tumormalignancy and, in particular, may counteract the peritonealdissemination of ovarian carcinoma (23). Based on these consid-erations and on our observation that, in EOC tissues, high levels of L1correlated with a less cohesive phenotype (e.g., see SupplementaryFig. S2B), we checked whether L1 regulates cell-cell adhesion innontransformed versus neoplastic ovarian epithelial cells. As shownin Fig. 4A , the forced expression of L1 in either HIOSE/A or HIOSE/B

Figure 3. Crosstalk of L1 with FGFR. A, OVCAR3-GFP or OVCAR3-L1cells were pretreated with 100 nmol/L PD173074 (PD) or with vehicle(DMSO) for 2 h before serum stimulation for the indicated time lengths andcell proliferation assay. *, P < 0.05 relative to PD173074-treatedOVCAR3-L1 cells.B, serum-starved OVCAR3-GFP or OVCAR3-L1 cells were stimulated with20 ng/mL FGF-2 for the indicated time lengths before measuring cell proliferation.*, P < 0.05; **, P < 0.005, relative to untreated OVCAR3-L1 and to FGF-treatedOVCAR3-GFP cells. C, serum-starved IGROV1-siGFP or IGROV1-siL1 cellswere stimulated with 20 ng/mL FGF-2 for the indicated time lengths beforemeasuring cell proliferation. The decline in cell number observed at 72 h inuntreated cells is likely due to apoptosis after serum deprivation. *, P < 0.05;**, P < 0.005, relative to FGF-treated IGROV1-siGFP cells.

Cancer Research

Cancer Res 2008; 68: (4). February 15, 2008 1114 www.aacrjournals.org

Research. on January 19, 2016. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from

cells enhanced their intercellular adhesion. We tested the effect ofL1 on both calcium-independent and dependent cell-cell adhesion.Whereas the former is mediated by Ig-CAMs, calcium-dependentintercellular adhesion is initiated by members of the cadherin family(2). Thus, the fact that L1 stimulated also calcium-dependentadhesion in HIOSE cells (Fig. 4A) suggested a functional interactionwith cadherins in this cell type. In contrast, L1 exhibited a negativeeffect on cell-cell adhesion in IGROV1 cells, in that L1 knockdowncells formed f3.5-fold and 10-fold more clusters than control cellsin calcium-independent and calcium-dependent conditions, respec-tively (Fig. 4B). Therefore, L1 stimulates cell-cell adhesion in HIOSEcells, whereas it exerts an inhibitory effect on intercellular adhesionin IGROV1 cells.L1 has been reported to interact with integrins, the key regulators

of cell adhesion to the extracellular matrix (24). Hence, we askedwhether L1 affected cell-matrix adhesion in nontumorigenic andneoplastic ovarian epithelial cells. Adhesion assays on differentextracellular matrix components revealed that L1 stimulates cell-matrix adhesion, although the efficiency of this proadhesive activityand the specific integrins involved (as reflected by the adhesion tospecific ECM components) show some differences between HIOSEand IGROV1 cells (Supplementary Fig. S8).

Cell InvasionPrevious studies have shown that L1 is implicated in the

migration of ovarian carcinoma cells (7). However, it has remainedelusive whether L1 also modulates the invasion of tumor cellsthrough a three-dimensional extracellular matrix, a key step incancer progression. To address this issue, we subjected L1-positiveand L1-negative HIOSE and IGROV1 cells to invasion assaysthrough Matrigel, a reconstituted basement membrane. BothHIOSE/A and HIOSE/B cells exhibited a weak invasive activitythat was not significantly affected by the ectopic expression of L1(Fig. 5A, left and middle columns). In contrast, L1 was requiredfor Matrigel invasion by IGROV1 cells, as shown by the f75%reduction of invasive activity upon ablation of L1 expression(Fig. 5A, right columns). Given the biological implications of thesefindings for the progression of ovarian carcinoma, we used the L1-negative OVCAR-3 cellular model to verify whether L1 was alsosufficient for ovarian cancer cell invasion. As shown in Fig. 5B, theforced expression of L1 resulted in a 4-fold increase in OVCAR-3cell invasion, confirming that this adhesion molecule contributes tothe malignant properties of ovarian cancer cells.Based on our observation of a L1/FGFR crosstalk in ovarian

cancer cell proliferation, we asked whether a similar mechanismwas involved in L1-induced OVCAR-3 cell invasion. Indeed, apretreatment with the FGFR inhibitor PD173074 efficientlyinhibited Matrigel invasion of L1-expressing OVCAR-3 cells to thelevel of control, untreated cells, whereas the invasion of OVCAR3-GFP cells was not affected (Fig. 5B). This specifically implicatesFGFR signaling in L1-induced invasion. In contrast, the EGFRinhibitor AG1478 reduced the invasive capacity of both OVCAR3-GFP and OVCAR3-L1 cells by f3.5-fold (Supplementary Fig. S6C),which suggests that EGFR is involved in ovarian cancer cellinvasion in a L1-independent manner.In an attempt to identify the signaling mediator(s) of L1-

dependent tumor cell invasion, we assessed the Matrigel invasionof L1-transfected OVCAR-3 cells in the presence of specificinhibitors of either the Erk1/2 or the phosphoinositide 3-kinase(PI3K) pathways, two major biochemical cascades previouslyimplicated in cancer cell malignancy. Both U0126 and LY294002,which inhibit Erk1/2 and PI3K activity, respectively, efficientlyrepressed the invasion of L1-expressing OVCAR-3 cells (Fig. 5B).These findings indicate that L1 enhances the invasive potential ofovarian carcinoma cells via the Erk1/2 and PI3K signalingpathways.

Transendothelial MigrationOvarian carcinoma disseminates predominantly through detach-

ment of tumor masses from the primary site, which then spreadinto the peritoneal cavity and/or colonize peritoneal organs (25).However, a significant proportion of ovarian carcinomas formsmetastases in the retroperitoneal lymph nodes, most likelydisseminating through the lymphatic circulation (26, 27). To verifywhether L1 is implicated in this route of EOC metastatic spread, weassayed for the ability of L1 to modulate the transmigration of wild-type versus L1-deficient IGROV1 cells through a monolayer oflymphatic endothelial cells. We used HDLEC1 cells, a lymphaticendothelial cell line, whose life span was prolonged by telomeraseexpression, and HMEC-1 cells, a widely used endothelial cell linethat was recently reported to express several lymphatic endothelialmarkers (28). Reducing L1 expression in IGROV1 cells caused a2-fold decrease in their ability to cross the lymphatic endothelialbarrier constituted by either HMEC-1 or HDLEC1 cells (Fig. 5C). An

Figure 4. The effect of L1 on cell-cell adhesion. HIOSE (A) or IGROV1 cells(B) were subjected to cell-cell adhesion assays as described in SupplementaryMaterials and Methods. Columns, mean from a representative experimentperformed in triplicate; bars, SD. *, P < 0.05, relative to control cells.

The Dual Role of L1 in Ovarian Epithelial Cells

www.aacrjournals.org 1115 Cancer Res 2008; 68: (4). February 15, 2008

Research. on January 19, 2016. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from

even stronger effect was observed on the transmigration throughblood vessel endothelial cells (Fig. 5C). Therefore, L1 is required forthe transendothelial migration of ovarian carcinoma cells,emerging as a potential player in the formation of EOC lymphnode metastasis.

Discussion

This study reports for the first time that the adhesion moleculeL1 exerts different functions in nontransformed versus neoplasticOSE cells, promoting a nonmalignant phenotype in the former andan invasive one in the latter. Previous studies have implicated L1 inEOC progression, based on the correlation between L1 expressionand poor prognosis in patients (6), an observation that we haveconfirmed and extended in the present report, and on the L1-dependent growth of EOC xenografts in the peritoneum ofimmunodeficient mice (16). However, the role of L1 in OSE cellshas not been addressed, likely due to previous negative results onL1 expression in this tissue. Indeed, the groups of Altevogt andFogel have reported the absence of L1 in normal OSE (6, 29), aresult that is in sharp contrast with our data. Those studies wereperformed using two monoclonal antibodies (L1-11A and L1-14-10,both raised against the ectodomain of L1) different from theantibodies used in our screening, which probably accounts for thediscrepancy between the results of Altevogt and Fogel and ours.This highlights the importance of the antibody selection whenanalyzing the expression and localization of L1, also based onthe notion that L1 can undergo extensive posttranslationalmodifications (including glycosylation, proteolytic cleavage, andectodomain shedding; ref. 30), which probably change itsimmunoreactivity. Overall, the finding that OSE cells do expressL1 is noteworthy because, combined with the specific L1enrichment in highly invasive EOC cells, it was at the origin ofour hypothesis that L1 plays a dual role in nontransformed versusneoplastic OSE cells.In nontumorigenic OSE cells, L1 supported cell-cell adhesion

and enhanced drug-induced apoptosis, whereas it showed noeffect on cellular processes associated with tumor malignancy. Incontrast, ovarian cancer cells exhibited L1-dependent cellproliferation, invasion, resistance to apoptosis, and transendothe-lial migration, all representing important steps in cancerprogression, whereas cell-cell adhesion was repressed by L1.The inhibitory effect of L1 on intercellular adhesion is notrestricted to ovarian cancer cells, as it was recently reported inthe breast cancer cell line MCF7, where L1-mediated disruption ofadherens junctions resulted in enhanced cell motility (31). On onehand, our findings confirm and extend previous observations onthe role of L1 in enhancing the malignant phenotype of ovariancarcinoma. For example, L1 has been reported to support the i.p.growth of ovarian cancer cells in immunodeficient mice (16) andto induce their migration and resistance to apoptosis (7, 22). Onthe other hand, our results highlight a novel function of L1 innontumorigenic OSE cells, namely the induction of both calcium-dependent and independent cell-cell adhesion. Based on thenotion that intercellular adhesion, and in particular the calcium-dependent one, efficiently represses tumor invasion (2), it istempting to speculate that L1 contributes to inhibit or restrict themalignant transformation of OSE cells. In agreement with thishypothesis, L1 is down-regulated in the majority of the tumorcells during EOC development (this study and ref. 6). Yet, highlevels of L1 are present in less cohesive cells within the tumormass, as well as at the tumor-stroma interface, namely wherecancer cells are actively invading the surrounding tissue. Overall,the picture emerging from the expression pattern of L1 in vivoand from our functional studies in cultured cells is consistentwith a model, whereby L1 enhances intercellular adhesion in OSEwhile it acts as a tumor promoter in advanced EOC.

Figure 5. The effect of L1 on cellular invasion and on transendothelial migration.A, HIOSE (left and middle columns) or IGROV1 cells (right columns) weresubjected to Matrigel invasion assays as described in Supplementary Materialsand Methods. Values represent the percentage of invasion relative to controlIGROV-siGFP cells. B, OVCAR3 cells expressing GFP or L1 were left untreatedor treated with 100 nmol/L PD173074, 20 Amol/L U0126, or 10 Amol/L LY294002for 30 min before the Matrigel invasion assay. Values represent the percentage ofinvasion relative to control OVCAR3-GFP cells. Columns, mean from threeindependent experiments, each performed in triplicate; bars, SE. *P < 0.005.C, IGROV1 cells transfected with siRNA against L1 or GFP were subjected totransmigration assays through monolayers of either, HDLEC1, HMEC-1, orhuman umbilical vascular endothelial cell (HUVEC ) cells, as described inSupplementary Materials and Methods. Columns, mean from a representativeexperiment performed in triplicate; bars, SD. *, P < 0.05, relative to control,siGFP-transfected cells.

Cancer Research

Cancer Res 2008; 68: (4). February 15, 2008 1116 www.aacrjournals.org

Research. on January 19, 2016. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from

The cellular and molecular factors that determine the changesin L1 expression during EOC progression remain elusive. Inadvanced colon carcinoma, the Wnt/h-catenin/TCF pathway hasbeen proposed to induce the expression of L1 at the invasivefront of the tumor (5). Unlike colon carcinoma, whose normalcounterpart is negative for L1 (5),7 the latter is found both innormal OSE and in a subset of EOC cells, implying that differentmechanisms could regulate the expression of L1 in differenttumors. Because most of the tumor cells in EOC masses show noL1 expression, ovarian tumorigenesis is accompanied by a generalloss of L1. Such a loss could result from the inactivation oftumor-suppressing genes, such as p53, an event occurring in 50%to 70% of advanced ovarian cancers (32). Indeed, an inactivatingmutation in the p53 gene is accompanied by the down-regulationof L1 in small cell carcinoma of the prostate.8 Along the sameline, we report the loss of L1 in OSE cells expressing the SV40 Tantigen, a well-characterized antagonist of p53 function (33). Thepresence of L1 at the invasive front of more advanced EOC couldthen depend on its reexpression as part of the transition towardan invasive phenotype. Based on our results in Ras-transformedOSE cells (see Supplementary Fig. S3) and on the frequency ofoncogenic Ras mutations in advanced EOC (34), it is conceivablethat this oncogene is causally involved in the expression of L1 ininvasive EOC cells. Along this line, we have observed inductionof L1 in OVCAR3 cells upon forced expression of activated Ras.9

At the cellular level, one possibility is that microenvironment-derived factors induce L1 expression in the cancer cells located atthe tumor-stroma interface. Alternatively, the L1-positive cells atthe edge of EOC might derive from the selection of a subset oftransformed OSE cells that have maintained the expression of L1,accompanied by a functional switch of the protein to aproinvasive activity, thus enhancing tumor malignancy.The dual role of L1 in OSE cells indicates that the cellular

context and, in particular, the acquisition of a transformedphenotype, has a major effect on the function of L1 and canactually switch it from a bona fide cell-cell adhesion molecule to atumor-promoting factor. The molecular events that determine thisfunctional switch remain unknown. An attractive candidate as amediator of L1’s effect on EOC development is the FGFR, given that(a) an aberrant FGFR signaling has been associated to EOCmalignancy (19, 20) and (b) the crosstalk between L1 and FGFR haslong been described in neuronal cells, where it stimulates axonalgrowth (17), although the physical association between the twomolecules has not been reported. Our results showed for the firsttime a functional interaction of L1 with FGFR in ovarian cancercells, where it plays an important role in L1-dependent cellproliferation and invasion. Thus, interfering with the L1/FGFRcrosstalk might prove a suitable therapeutic approach for the

treatment of EOC. In agreement with our observations, theadhesion molecule N-cadherin has been shown to potentiate FGFRsignaling in breast cancer cells by favoring a sustained activation ofthe receptor by FGF-2 (35). It is noteworthy that the crosstalk of anadhesion molecule with FGFR can also lead to the repression ofFGF-induced signaling, as we have recently reported for another Ig-CAM, neural cell adhesion molecule, that abolishes the cellularresponse to FGF-2 in fibroblasts (36). Hence, the outcome of theadhesion molecule/FGFR interaction is likely to depend on thespecific adhesion molecule involved and on the cellular context. Onthe other hand, also the repertoire of growth factor receptorsinvolved in crosstalk with L1 seems to be cell type–specific,because, unlikely previous observations in neurons (18), weobtained no evidence of L1 interacting with the EGFR signalingmachinery in ovarian cancer cells.Other signaling mediators that have been implicated in L1-

induced axonal growth (37) and, based on our studies, are alsoinvolved in L1-dependent ovarian cancer progression includemitogen-activated protein kinase and PI3K pathways. But whyare these signaling cascades activated by L1 specifically in ovariancancer and not in OSE cells? A possible scenario is that therepertoire of proteins interacting with L1 changes upon neoplastictransformation of OSE cells. Indeed, many studies on neuronalsystems have documented the heterophilic interactions of L1 witha broad spectrum of molecules. The interacting partners of L1include several components of the extracellular matrix, cell surfacemolecules, such as EGFR, neuropilin, and various integrins (besidesFGFR, as mentioned above), intracellular signaling effectors, suchas Src, Numb, and RanBPM, and cytoskeletal components, such asankyrins and ezrin (reviewed in refs. 3, 38). Many of thesemolecules are involved in cellular processes that, once deregulated,contribute to cancer progression, thus providing a potential basisfor L1-dependent function in EOC. For example, FGFRs areabundantly expressed in EOC (19, 20, 39), and our data supportthe hypothesis that the simultaneous expression of L1 leads toexcessive FGFR signaling that, in turn, enhance tumor invasion.In summary, we have shown that L1 plays a dual role in OSE

cells, consistent with a tumor-suppressive function in nontrans-formed cells and with a proinvasive function in cancer cells. Thecharacterization of the cellular and molecular determinantsresponsible for this shift in L1 activity will contribute to theidentification of novel regulatory mechanisms implicated in EOCprogression, hopefully providing new therapeutic targets for thisneoplastic disease.

Acknowledgments

Received 7/30/2007; revised 11/8/2007; accepted 12/19/2007.Grant support: Associazione Italiana per la Ricerca sul Cancro grant 1378 (U.

Cavallaro), Telethon Foundation grant GGP04078 (U. Cavallaro), and CariploFoundation grant 2004.1587/11.5437 (U. Cavallaro).

The costs of publication of this article were defrayed in part by the payment of pagecharges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.

We thank V. Lemmon, M. Schachner, K. Blaser, T.J. Lawley, A. Insinga, R. Nisato, andM. Pepper for providing antibodies and cell lines.

7 Our unpublished observation.8 D.E. Hansel, personal communication.9 S. Zecchini and U. Cavallaro, unpublished data.

References1. Sun CC, Ramirez PT, Bodurka DC. Quality of life forpatients with epithelial ovarian cancer. Nat Clin PractOncol 2007;4:18–29.

2. Cavallaro U, Christofori G. Cell adhesion and signalling

by cadherins and Ig-CAMs in cancer. Nat Rev Cancer2004;4:118–32.

3. Kenwrick S, Watkins A, Angelis ED. Neural cellrecognition molecule L1: relating biological complexityto human disease mutations. Hum Mol Genet 2000;9:879–86.

4. Thies A, Schachner M, Moll I, et al. Overexpression ofthe cell adhesion molecule L1 is associated withmetastasis in cutaneous malignant melanoma. Eur JCancer 2002;38:1708–16.

5. Gavert N, Conacci-Sorrell M, Gast D, et al. L1, a noveltarget of {h}-catenin signaling, transforms cells and is

The Dual Role of L1 in Ovarian Epithelial Cells

www.aacrjournals.org 1117 Cancer Res 2008; 68: (4). February 15, 2008

Research. on January 19, 2016. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from

expressed at the invasive front of colon cancers. J CellBiol 2005;168:633–42.

6. Fogel M, Gutwein P, Mechtersheimer S, et al. L1expression as a predictor of progression and survival inpatients with uterine and ovarian carcinomas. Lancet2003;362:869–75.

7. Gast D, Riedle S, Riedle S, et al. L1 augments cellmigration and tumor growth but not h3 integrinexpression in ovarian carcinomas. Int J Cancer 2005;115:658–65.

8. Shih IM, Kurman RJ. Ovarian tumorigenesis: aproposed model based on morphological and moleculargenetic analysis. Am J Pathol 2004;164:1511–8.

9. Yang WL, Godwin AK, Xu XX. Tumor necrosis factor-a-induced matrix proteolytic enzyme production andbasement membrane remodeling by human ovariansurface epithelial cells: molecular basis linking ovulationand cancer risk. Cancer Res 2004;64:1534–40.

10. Liu J, Yang G, Thompson-Lanza JA, et al. A geneticallydefined model for human ovarian cancer. Cancer Res2004;64:1655–63.

11. Mechtersheimer S, Gutwein P, Agmon-Levin N, et al.Ectodomain shedding of L1 adhesion molecule pro-motes cell migration by autocrine binding to integrins.J Cell Biol 2001;155:661–73.

12. Zisch AH, Stallcup WB, Chong LD, et al. Tyrosinephosphorylation of L1 family adhesion molecules:implication of the Eph kinase Cek5. J Neurosci Res1997;47:655–65.

13. Auersperg N, Wong AS, Choi KC, Kang SK, Leung PC.Ovarian surface epithelium: biology, endocrinology, andpathology. Endocr Rev 2001;22:255–88.

14. Leung EH, Leung PC, Auersperg N. Differentiationand growth potential of human ovarian surfaceepithelial cells expressing temperature-sensitive SV40T antigen. In vitro Cell Dev Biol Anim 2001;37:515–21.

15. Capo-Chichi CD, Smith ER, Yang DH, et al.Dynamic alterations of the extracellular environmentof ovarian surface epithelial cells in premalignanttransformation, tumorigenicity, and metastasis. Cancer2002;95:1802–15.

16. Arlt MJ, Novak-Hofer I, Gast D, et al. Efficientinhibition of intra-peritoneal tumor growth and dis-semination of human ovarian carcinoma cells in nude

mice by anti-L1-cell adhesion molecule monoclonalantibody treatment. Cancer Res 2006;66:936–43.

17. Williams EJ, Furness J, Walsh FS, Doherty P.Activation of the FGF receptor underlies neuriteoutgrowth stimulated by L1, N-CAM, N-cadherin.Neuron 1994;13:583–94.

18. Islam R, Kristiansen LV, Romani S, Garcia-Alonso L,Hortsch M. Activation of EGF receptor kinase by L1-mediated homophilic cell interactions. Mol Biol Cell2004;15:2003–12.

19. De Cecco L, Marchionni L, Gariboldi M, et al. Geneexpression profiling of advanced ovarian cancer:characterization of a molecular signature involvingfibroblast growth factor 2. Oncogene 2004;23:8171–83.

20. Valve E, Martikainen P, Seppanen J, et al. Expressionof fibroblast growth factor (FGF)-8 isoforms and FGFreceptors in human ovarian tumors. Int J Cancer 2000;88:718–25.

21. Hennessy BT, Mills GB. Ovarian cancer: Homeoboxgenes, autocrine/paracrine growth, and kinase signaling.Int J Biochem Cell Biol 2006;38:1450–6.

22. Stoeck A, Gast D, Sanderson MP, Issa Y, Gutwein P,Altevogt P. L1-CAM in a membrane-bound or solubleform augments protection from apoptosis in ovariancarcinoma cells. Gynecol Oncol 2007;104:461–9.

23. Sundfeldt K. Cell-cell adhesion in the normal ovaryand ovarian tumors of epithelial origin; an exception tothe rule. Mol Cell Endocrinol 2003;202:89–96.

24. Silletti S, Mei F, Sheppard D, Montgomery AM.Plasmin-sensitive dibasic sequences in the third fibro-nectin-like domain of L1-cell adhesion molecule (CAM)facilitate homomultimerization and concomitant integ-rin recruitment. J Cell Biol 2000;149:1485–502.

25. Tan DSP, Agarwal R, Kaye SB. Mechanisms oftranscoelomic metastasis in ovarian cancer. LancetOncol 2006;7:925–34.

26. Sundar SS, Zhang H, Brown P, et al. Role oflymphangiogenesis in epithelial ovarian cancer. Br JCancer 2006;94:1650–7.

27. Ueda M, Hung Y-C, Terai Y, et al. Vascular endothelialgrowth factor-C expression and invasive phenotype inovarian carcinomas. Clin Cancer Res 2005;11:3225–32.

28. Nisato RE, Harrison JA, Buser R, et al. Generationand characterization of telomerase-transfected human

lymphatic endothelial cells with an extended life span.Am J Pathol 2004;165:11–24.

29. Huszar M, Moldenhauer G, Gschwend V, Ben-Arie A,Altevogt P, Fogel M. Expression profile analysis inmultiple human tumors identifies L1 (CD171) as amolecular marker for differential diagnosis and targetedtherapy. Hum Pathol 2006;37:1000–8.

30. Haspel J, Grumet M. The L1CAM extracellular region:a multi-domain protein with modular and cooperativebinding modes. Front Biosci 2003;8:s1210–25.

31. Shtutman M, Levina E, Ohouo P, Baig M, RoninsonIB. Cell adhesion molecule L1 disrupts E-cadherin-containing adherens junctions and increases scatteringand motility of MCF7 breast carcinoma cells. Cancer Res2006;66:11370–80.

32. Okamoto A, Sameshima Y, Yokoyama S, et al.Frequent allelic losses and mutations of the p53gene in human ovarian cancer. Cancer Res 1991;51:5171–6.

33. Ahuja D, Saenz-Robles MT, Pipas JM. SV40 large Tantigen targets multiple cellular pathways to elicitcellular transformation. Oncogene 2005;24:7729–45.

34. Mammas IN, Zafiropoulos A, Spandidos DA. Involve-ment of the ras genes in female genital tract cancer. Int JOncol 2005;26:1241–55.

35. Suyama K, Shapiro I, Guttman M, Hazan RB. Asignaling pathway leading to metastasis is controlled byN-cadherin and the FGF receptor. Cancer Cell 2002;2:301–14.

36. Francavilla C, Loeffler S, Piccini D, Kren A, Christo-fori G, Cavallaro U. Neural cell adhesion moleculeregulates the cellular response to fibroblast growthfactor. J Cell Sci 2007;120:4388–94.

37. Schmid RS, Pruitt WM, Maness PF. A MAP kinase-signaling pathway mediates neurite outgrowth on L1and requires Src-dependent endocytosis. J Neurosci2000;20:4177–88.

38. Maness PF, Schachner M. Neural recognition mole-cules of the immunoglobulin superfamily: signalingtransducers of axon guidance and neuronal migration.Nat Neurosci 2007;10:19–26.

39. Zhang Y, Guo KJ, Shang H, Wang YJ, Sun LG.Expression of aFGF, bFGF, FGFR1 in ovarian epithelialneoplasm. Chin Med J (Engl) 2004;117:601–3.

Cancer Research

Cancer Res 2008; 68: (4). February 15, 2008 1118 www.aacrjournals.org

Research. on January 19, 2016. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from

2008;68:1110-1118. Cancer Res   Silvia Zecchini, Marco Bianchi, Nicoletta Colombo, et al.   Ovarian Surface EpitheliumThe Differential Role of L1 in Ovarian Carcinoma and Normal

  Updated version

  http://cancerres.aacrjournals.org/content/68/4/1110

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2008/02/22/68.4.1110.DC1.html

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/68/4/1110.full.html#ref-list-1

This article cites 39 articles, 13 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/68/4/1110.full.html#related-urls

This article has been cited by 13 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] at

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

  Permissions

  [email protected] at

To request permission to re-use all or part of this article, contact the AACR Publications

Research. on January 19, 2016. © 2008 American Association for Cancercancerres.aacrjournals.org Downloaded from