11
Expression and Nuclear Location of the Transcriptional Repressor Kaiso Is Regulated by the Tumor Microenvironment Adelheid Soubry, 1 Jolanda van Hengel, 1 Eef Parthoens, 1 Cecile Colpaert, 2 Eric Van Marck, 2 David Waltregny, 3 Albert B. Reynolds, 4 and Frans van Roy 1 1 Molecular Cell Biology Unit, Department for Molecular Biomedical Research, VIB-Ghent University, Ghent; 2 Department of Pathology, University of Antwerp, Antwerp; 3 Metastasis Research Laboratory, Center for Experimental Cancer Research, University Hospital of Lie `ge, Belgium; and 4 Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee Abstract Kaiso is a BTB/POZ zinc finger protein originally described as an interaction partner of p120ctn. In cultured cell lines, Kaiso is found almost exclusively in the nucleus, where it generally acts as a transcriptional repressor. Here, we describe the first in situ immunolocalization studies of Kaiso expression in normal and cancerous tissues. Surprisingly, we found striking differences between its behavior in monolayers of different cell lines, three-dimensional cell culture systems, and in vivo . Although nuclear localization was sometimes observed in tissues, Kaiso was more often found in the cytoplasm, and in some cell types it was absent. In general, Kaiso and p120ctn did not colocalize in the nucleus. To examine this phenomenon more carefully, tumor cells exhibiting strong nuclear Kaiso staining in vitro were injected into nude mice and grown as xenografts. The latter showed a progressive translocation of Kaiso towards the cytoplasm over time, and even complete loss of expression, especially in the center of the tumor nodules. When xenografted tumors were returned to cell culture, Kaiso was re-expressed and was once again found in the nucleus. Translocation of Kaiso to the cytoplasm and down-regulation of its levels were also observed under particular experimental conditions in vitro , such as formation of spheroids and acini. These data strongly imply an unexpected influence of the microenvironment on Kaiso expression and localization. As transcriptional repres- sion is a nuclear event, this phenomenon is likely a crucial factor in the regulation of Kaiso function. (Cancer Res 2005; 65(6): 2224-33) Introduction The p120ctn binding partner Kaiso was originally discovered by a yeast two-hybrid approach using p120ctn as bait (1). p120ctn is an Armadillo protein first identified as a prominent tyrosine kinase substrate implicated in cell transformation by Src (2), and in ligand- induced receptor signaling through various tyrosine kinase recep- tors (3, 4). p120ctn binds to the juxtamembrane domain of classical cadherins (reviewed in refs. 5, 6), where it modulates cell-cell adhesion by regulating cadherin turnover and stability at the cell surface (7–9). In E-cadherin-negative cancer cells, p120ctn localizes aberrantly to both the cytoplasm and the nucleus (10, 11). Cytoplasmic p120ctn may promote metastasis through interactions with Rho GTPases (12), but aside from the Kaiso connection, roles for p120ctn in the nucleus have not been described. There is mounting evidence that Kaiso is a nuclear protein that plays a role in transcription repression. Kaiso contains an amino- terminal BTB/POZ domain (Broad Complex, Tramtrak, Bric a ` brac/ Pox virus and Zinc finger) and a carboxyl-terminal region with three zinc finger motifs of the C 2 H 2 type (1). Other known members of the BTB/POZ family have roles in development or cancer, and are also transcriptional repressors. For example, the human BCL-6 and promyelocytic leukemia zinc finger proteins are causally involved in non-Hodgkin’s lymphoma and acute promyelocytic leukemia, respectively (13, 14). The BTB/POZ proteins APM-1 and mGCL inhibit growth of cervical carcinoma and osteosarcoma cells, respectively (15, 16), whereas the BTB/POZ protein HIC1 and its relative HRG22 are candidate tumor suppressors in a variety of human cancers (17). Kaiso homodimerizes via its POZ domain (1), which may contribute to the assembly of large multiprotein complexes. As in the case of other POZ family members, Kaiso interacts with histone deacetylases through the corepressor N-CoR (18, 19), thereby controlling gene expression. Kaiso’s description as a transcriptional repressor is also based on the specific recognition by its zinc fingers of an MGMG motif (where M is a 5-methylcytosine; refs. 19, 20). Thus, despite the absence of a canonical methyl-binding domain, Kaiso may belong to a group of complexes or proteins, including MeCP2, MeCP1, and methyl-binding domain-1, which are involved in repressing DNA transcription through their attraction to methylated DNA (21, 22). CAST analysis, however, revealed that Kaiso also binds to the DNA sequence CTGCNA in a methylation- independent manner mediated by zinc finger-2 and zinc finger-3 (23). The relative importance of the methylation-dependent and -independent binding motifs is unknown. Like HIC1 (17), Kaiso may repress transcription by a dual mechanism. Several cancer-related gene fragments, such as regulatory sequences of the genes Rb, Xist, S100A4 (mts-1), CDH1 (E-cadherin), matrilysin (MMP7), MTA2 (metastasis-associated gene 2 ) and Wnt11 , have been proposed as candidates for Kaiso binding (19, 20, 23, 24). On the other hand, the core rapsyn promoter was recently shown to be up-regulated by a complex of Kaiso with y-catenin in subsynaptic nuclei of neuromuscular junctions (25). DNA binding and transcription repression by Kaiso may be modulated by interaction with p120ctn. Interaction between p120ctn and Kaiso occurs via the Arm repeats 1 to 7 of p120ctn and a noncontiguous Kaiso domain flanking the carboxyl- terminal DNA-binding zinc finger domain (1). This close physical juxtaposition of the p120ctn-binding site of Kaiso with its DNA- binding motif may explain the reported finding that DNA binding of Kaiso is inhibited by nuclear p120ctn (23, 26). Association of p120ctn with E-cadherin occurs via its Arm repeats 1 to 5 and 7 (7). Thus, Kaiso and E-cadherin likely bind Requests for reprints: Frans van Roy, Department for Molecular Biomedical Research, VIB-Ghent University, FSVM Building, Technologiepark 927, B-9052 Ghent (Zwijnaarde), Belgium. Phone: 32-9-331-3601; Fax: 32-9-331-3609; E-mail: [email protected]. #2005 American Association for Cancer Research. Cancer Res 2005; 65: (6). March 15, 2005 2224 www.aacrjournals.org Research Article Research. on August 31, 2021. © 2005 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Expression and Nuclear Location of the Transcriptional ...Kaiso is a BTB/POZ zinc finger protein originally described as an interaction partner of p120ctn. In cultured cell lines,

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Expression and Nuclear Location of the Transcriptional ...Kaiso is a BTB/POZ zinc finger protein originally described as an interaction partner of p120ctn. In cultured cell lines,

Expression and Nuclear Location of the Transcriptional Repressor

Kaiso Is Regulated by the Tumor Microenvironment

Adelheid Soubry,1Jolanda van Hengel,

1Eef Parthoens,

1Cecile Colpaert,

2Eric Van Marck,

2

David Waltregny,3Albert B. Reynolds,

4and Frans van Roy

1

1Molecular Cell Biology Unit, Department for Molecular Biomedical Research, VIB-Ghent University, Ghent; 2Department of Pathology,University of Antwerp, Antwerp; 3Metastasis Research Laboratory, Center for Experimental Cancer Research, University Hospital ofLiege, Belgium; and 4Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee

Abstract

Kaiso is a BTB/POZ zinc finger protein originally describedas an interaction partner of p120ctn. In cultured cell lines,Kaiso is found almost exclusively in the nucleus, where itgenerally acts as a transcriptional repressor. Here, wedescribe the first in situ immunolocalization studies of Kaisoexpression in normal and cancerous tissues. Surprisingly, wefound striking differences between its behavior in monolayersof different cell lines, three-dimensional cell culture systems,and in vivo . Although nuclear localization was sometimesobserved in tissues, Kaiso was more often found in thecytoplasm, and in some cell types it was absent. In general,Kaiso and p120ctn did not colocalize in the nucleus. Toexamine this phenomenon more carefully, tumor cellsexhibiting strong nuclear Kaiso staining in vitro were injectedinto nude mice and grown as xenografts. The latter showed aprogressive translocation of Kaiso towards the cytoplasmover time, and even complete loss of expression, especially inthe center of the tumor nodules. When xenografted tumorswere returned to cell culture, Kaiso was re-expressed and wasonce again found in the nucleus. Translocation of Kaiso tothe cytoplasm and down-regulation of its levels were alsoobserved under particular experimental conditions in vitro ,such as formation of spheroids and acini. These data stronglyimply an unexpected influence of the microenvironment onKaiso expression and localization. As transcriptional repres-sion is a nuclear event, this phenomenon is likely a crucialfactor in the regulation of Kaiso function. (Cancer Res 2005;65(6): 2224-33)

Introduction

The p120ctn binding partner Kaiso was originally discovered by ayeast two-hybrid approach using p120ctn as bait (1). p120ctn is anArmadillo protein first identified as a prominent tyrosine kinasesubstrate implicated in cell transformation by Src (2), and in ligand-induced receptor signaling through various tyrosine kinase recep-tors (3, 4). p120ctn binds to the juxtamembrane domain of classicalcadherins (reviewed in refs. 5, 6), where it modulates cell-celladhesion by regulating cadherin turnover and stability at the cellsurface (7–9). In E-cadherin-negative cancer cells, p120ctn localizesaberrantly to both the cytoplasm and the nucleus (10, 11).Cytoplasmic p120ctn may promote metastasis through interactions

with Rho GTPases (12), but aside from the Kaiso connection, rolesfor p120ctn in the nucleus have not been described.There is mounting evidence that Kaiso is a nuclear protein that

plays a role in transcription repression. Kaiso contains an amino-terminal BTB/POZ domain (Broad Complex, Tramtrak, Bric a brac/Pox virus and Zinc finger) and a carboxyl-terminal region with threezinc finger motifs of the C2H2 type (1). Other known members of theBTB/POZ family have roles in development or cancer, and are alsotranscriptional repressors. For example, the human BCL-6 andpromyelocytic leukemia zinc finger proteins are causally involvedin non-Hodgkin’s lymphoma and acute promyelocytic leukemia,respectively (13, 14). The BTB/POZ proteins APM-1 and mGCLinhibit growth of cervical carcinoma and osteosarcoma cells,respectively (15, 16), whereas the BTB/POZ protein HIC1 and itsrelative HRG22 are candidate tumor suppressors in a variety ofhuman cancers (17). Kaiso homodimerizes via its POZ domain (1),which may contribute to the assembly of large multiproteincomplexes. As in the case of other POZ family members, Kaisointeracts with histone deacetylases through the corepressor N-CoR(18, 19), thereby controlling gene expression. Kaiso’s description as atranscriptional repressor is also based on the specific recognition byits zinc fingers of an MGMG motif (where M is a 5-methylcytosine;refs. 19, 20). Thus, despite the absence of a canonical methyl-bindingdomain, Kaiso may belong to a group of complexes or proteins,including MeCP2, MeCP1, and methyl-binding domain-1, which areinvolved in repressing DNA transcription through their attraction tomethylated DNA (21, 22). CAST analysis, however, revealed thatKaiso also binds to the DNA sequence CTGCNA in a methylation-independent manner mediated by zinc finger-2 and zinc finger-3(23). The relative importance of the methylation-dependent and-independent binding motifs is unknown. Like HIC1 (17), Kaiso mayrepress transcription by a dual mechanism. Several cancer-relatedgene fragments, such as regulatory sequences of the genes Rb, Xist,S100A4 (mts-1), CDH1 (E-cadherin), matrilysin (MMP7), MTA2(metastasis-associated gene 2) and Wnt11 , have been proposed ascandidates for Kaiso binding (19, 20, 23, 24). On the other hand, thecore rapsyn promoter was recently shown to be up-regulated bya complex of Kaiso with y-catenin in subsynaptic nuclei ofneuromuscular junctions (25).DNA binding and transcription repression by Kaiso may be

modulated by interaction with p120ctn. Interaction betweenp120ctn and Kaiso occurs via the Arm repeats 1 to 7 of p120ctnand a noncontiguous Kaiso domain flanking the carboxyl-terminal DNA-binding zinc finger domain (1). This close physicaljuxtaposition of the p120ctn-binding site of Kaiso with its DNA-binding motif may explain the reported finding that DNAbinding of Kaiso is inhibited by nuclear p120ctn (23, 26).Association of p120ctn with E-cadherin occurs via its Armrepeats 1 to 5 and 7 (7). Thus, Kaiso and E-cadherin likely bind

Requests for reprints: Frans van Roy, Department for Molecular BiomedicalResearch, VIB-Ghent University, FSVM Building, Technologiepark 927, B-9052Ghent (Zwijnaarde), Belgium. Phone: 32-9-331-3601; Fax: 32-9-331-3609; E-mail:[email protected].

#2005 American Association for Cancer Research.

Cancer Res 2005; 65: (6). March 15, 2005 2224 www.aacrjournals.org

Research Article

Research. on August 31, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: Expression and Nuclear Location of the Transcriptional ...Kaiso is a BTB/POZ zinc finger protein originally described as an interaction partner of p120ctn. In cultured cell lines,

the same p120ctn domain in a mutually exclusive manner(1, 27).Previous reports describing almost exclusive nuclear localization

of Kaiso have been based entirely on in vitro observations in cellcultures (1, 28). Here, we describe the first in situ studies of Kaisolocalization in normal and cancerous human tissues. Surprisingly,Kaiso was frequently found in the cytoplasm, and was also oftenabsent. Moreover, Kaiso localized differently in the same cell linesdepending on whether the cells were grown in vitro , as two-dimensional monolayers with increasing cell densities on varioussubstrates, as three-dimensional cultures, or as xenografts in nudemice. These data reveal a striking microenvironmental effect that iscrucial in regulating Kaiso localization, expression, and activity.

Materials and Methods

Cell Culture. HT29 and SW48 cell lines were purchased from theAmerican Cell Type Culture collection (ATCC, Rockville, MD). HT29 human

colorectal adenocarcinoma cells were maintained in DMEM (Invitrogen,

Carlsbad, CA) with 15% FCS, nonessential amino acids (Invitrogen), 0.4 AMsodium pyruvate, 0.2 units/ml penicillin, 0.2 Ag/ml streptomycin and2 mmol L-glutamate. SW48 human colon adenocarcinoma cells were main-

tained in LB15 (Invitrogen) supplemented with 10% FCS, 0.2 units/ml

penicillin, 0.2 Ag/ml streptomycin, and 2 mmol L-glutamate. MCF-10A, a

spontaneously immortalized, nontransformed human mammary epithelialcell line (29), was kindly provided by J. Brugge (Harvard Medical School,

Boston, MA). These cells were grown in a special supplemented medium as

described previously (30).

Primary Antibodies. Kaiso was detected using two mouse monoclonalantibodies (6F and 12H, each at a concentration of 4 Ag/mL), and a rabbit

polyclonal antibody (designated pAb R and used at dilution 1:250; refs. 1, 28).

We have also generated a new pAb (S1337) against peptide C-NVTDGSTE-FEFIIPESY, which corresponds to a COOH-terminal sequence of human and

mouse Kaiso. The procedure used has been described previously (31). For

p120ctn recognition, monoclonal antibody (mAb) pp120 (Transduction

Laboratories, Lexington, KY) was used at a 1:200 dilution. Staining for E-cadherin was done with mAb human epithelial cadherin-1 (Takara, Kyoto,

Japan) at a concentration of 1.3 Ag/mL. For staining of carbonic anhydrase IX,

the mouse monoclonal antibody M75 (32) was used at a dilution of 1:50.

Antibody against HIF-1a (Transduction Laboratories, Erembodegem,Belgium) was used at a 1:80 dilution. Double staining for Kaiso and

p120ctn was done either with mAb 6F in combination with FITC-conjugated

pp120 mAb (Transduction Laboratories, Lexington, KY) or with pAb S1337together with mAb pp120.

Preparation of Cells and Tissues for Immunodetection. HT29 and

SW48 cells were grown on membrane filters, fixed in 4% formalin for 1 hour

and embedded in 5% agar in PBS before embedding in paraffin. Frozen orparaffin-embedded sections of normal and cancerous tissues from different

patients were obtained from the Department of Pathology at the University of

Antwerp, as well as a tissue microarray containing biopsies of inflammatory

breast cancers (IBC) from 32 patients prior to initiation of chemotherapy (33).Archival formalin-fixed paraffin-embedded tissue samples from human

prostate cancers (PCas) were obtained from the Department of Pathology,

University Hospital of Liege, Belgium. These sampleswere surgically obtainedfrom 45 patients who had undergone a radical retropubic prostatectomy for

localized prostate cancer. Patients who had received prior hormonal therapy,

chemotherapy, or radiation therapy were not included in the investigation.

All patients had a clinically confined tumor, classified as stage T1 (n = 19) orT2 (n = 26) according to the tumor-node-metastasis classification (34). After

histopathologic examination, 19 patients were classified as having tumors of

pathologic stage pT2. In 16 patients, extracapsular extension of the tumor

(stage pT3A) was observed. The remaining 10 patients were categorized ashaving PCas of stage pT3B because their tumors showed evidence of invading

the seminal vesicle. All PCas were evaluated according to the Gleason scoring

system (35). Tumors were classified as high grade (n = 24) when the Gleason

score was 7 or above, and as low grade (n = 21) when it was 6 or below. One

tissue block per patient containing the most representative tumor-bearingareas was selected considering the capsular status (pathologic stage) and

the Gleason score stated in the pathological report. Serial sections were

made and stained as described below. Decreased expression of p120ctn and

E-cadherin was noted in several PCas, and down-regulation of each of theseproteins correlated with high tumor grade (to be reported in detail

elsewhere).

Xenografts in Nude Mice. Athymic nu/nu mice 8 weeks of age were

purchased from Iffa Credo, France. Following a 7-day adaptation period, eachmouse was inoculated s.c. in each flank with 5 � 106 HT29 or SW48 tumor

cells in 100 ml sterile PBS. This experiment was done twice. In the first series,

mice with HT29-derived tumors were sacrificed (one per time point) on days

6, 11, 14, and 20 after inoculation, whereas mice with SW48-derived tumorswere sacrificed on days 6, 11, 20, and 32 after inoculation. All tumors grew

progressively, although HT29 xenografts grew faster than SW48 xenografts. In

a second series, mice with HT29 xenografts were sacrificed (one per timepoint) on days 2, 3, 4, 6, 9, 12, and 31 after inoculation, whereas mice with

SW48 xenografts were sacrificed on days 4, 6, 8, 9, 13, 23, and 38 after

inoculation. Half of each tumor was fixed in 4% formalin before embedding in

paraffin, and the other half was frozen in liquid nitrogen and stored at�80jC.When tumors were very small, the left tumor was prepared for paraffin

embedding and the right tumor was prepared for frozen sections.

Cryosections and paraffin sections were made for the different growth

stages of the tumors.Ex vivo Culture of Xenografted Cells. On the day of excision of

xenografted cells, 1/4 of the tumor was explanted. Cells were disaggregated in

PBS containing 1 mg/mL collagenase A and 0.05% trypsin, and incubated at

37jC on a rotator for 3 hours. After washing in medium, half of the cells were

prepared for cytospin analysis. The remaining cells were grown on cover slips

until they were subconfluent (1 week after explantation for HT29 cells, and 2

weeks after explantation for SW48 cells), and then used for immunofluores-

cence staining. The human origin of these cultures was verified by

appropriate species-specific antibodies.

Immunocytochemistry and Immunohistochemistry. Paraffin and

cryostat sections of 5 AM/L were used. Immunodetection in cryosections

has been described (31), as has the protocol for paraffin sections (cf. E-

cadherin in ref. 36). Sections were incubated with primary antibodies for 1hour at room temperature. Staining of paraffin sections was completed by a

biotinylated secondary antibody, streptavidin-peroxidase, and 3,3V-diamino-

benzidine. In the case of double staining, a standard Kaiso staining was

followed by application of anti–carbonic anhydrase IX antibody M75. The

latter was revealed using a DAKO EnVision alkaline phosphatase kit in

combination with fuchsin + substrate-chromogen (DakoCytomation, Hever-

lee, Belgium).

Samples were examined with an Olympus BX51 microscope, and imageswere recorded with a Coolsnap camera (Photometrics, Tucson, AZ) using

RSImage software (Roper Scientific, Trenton, NJ).

Cell Culture Under Hypoxic and Normoxic Conditions. HT29 and

SW48 cells were seeded on glass cover slips in six-well plates and incubated

under normal conditions overnight. They were exposed to hypoxia by

incubation in a humidified chamber that was continuously purged with a

mixture of 1% O2, 5% CO2, and 94% N2, as described by Fordel et al. (37).

Mediumwas changed every 2 dayswith appropriately gas-equilibratedmedia.

After 1, 3, 4, 5, and 7 days, cell cultures were harvested and fixed with

methanol or paraformaldehyde under hypoxic conditions. Control cells were

also cultured simultaneously under standard normoxic conditions.Two-dimensional and Three-dimensional Growth Conditions.

Monolayers of MCF-10A cells were grown on clean glass cover slips, either

uncoated or coated with GFR-Matrigel (BD Biosciences, Erembodegem,

Belgium). Cells were plated as sparse (15-20% of confluency), subconfluent(70-80%), confluent (90-95%), and very confluent (100%) cultures. Monolayers

of HT29 were grown on uncoated glass cover slips. Spheroids of HT29 cells

were made in static cultures according to the method of Yuhas et al. (38). In

order to obtain three-dimensional cultures of MCF-10A, single cells wereseeded on a Lab Tek II eight-well glass chamber slide (Nunc, Roskilde,

Denmark) with a solidified layer of GFR-Matrigel measuring 1 to 2 mm in

thickness. Cells were grown under the same conditions as those described

Kaiso Expression Regulated by Tumor Microenvironment

www.aacrjournals.org 2225 Cancer Res 2005; 65: (6). March 15, 2005

Research. on August 31, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: Expression and Nuclear Location of the Transcriptional ...Kaiso is a BTB/POZ zinc finger protein originally described as an interaction partner of p120ctn. In cultured cell lines,

earlier (30). Formation of acini-like structures was monitored for 21 days.Immunofluorescence staining was done on days 0.5, 1.5, 3, 7, 14, and 21.

Immunofluorescence. Cells were rinsed briefly with PBS and fixed eitherwith ice-cold 100% methanol for 15 minutes at �20jC, or with 4%

paraformaldehyde in PBS for 25 minutes at room temperature. Immuno-staining was done as previously described (30, 39). Antibodies were incubated

for 1 hour or overnight at 4jC. Secondary antibodies were Alexa 488/594-

coupled anti-mouse immunoglobulin or Alexa 488/594-coupled anti-rabbit

immunoglobulin antibodies (Molecular Probes, Eugene, OR). Counterstain-ing was done with Vectashield mounting medium containing 4V,6-diamidino-

2-phenylindole or propidium iodide (Vectorlabs, Burlingame, CA). Monolayer

cultures were examined with a Zeiss Axiophot photomicroscope and images

were recordedwith aMicroMax camera (Princeton Instruments, Trenton, NJ)and Metamorph software (Image Universal Corporation, New York, NY).

Differential interference contrast pictures of very confluent monolayers were

taken using a Leica DM IRE2 microscope equipped with an HCX PL APO63�/1.30 glycerin-corrected 37jC objective and a Coolsnap HQ camera.

Wide-field fluorescence images were monitored with the same microscopic

setup using a monochromator systemwith a 50WXenon lamp for excitation.

The fluorescence dyes Alexa 488 and 594were detected using a standard B/G/R filter cube (Leica). Blind deconvolution was carried out afterwards using

the Leica Deblur software. Confocal images of three-dimensional cultures

were made with an inverted Zeiss LSM 410 or LSM 510 confocal laser

scanning microscope.

Results

Predominantly Cytoplasmic Localization of Kaiso In vivo.Previous analyses of Kaiso had been carried out exclusively incultured cells. Here, we examined localization of Kaiso byimmunocytochemistry in a broad range of both normal andcancerous tissues, including ovary (two normal samples, onepapillary serous adenocarcinoma, and a liver metastasis of thelatter), oviduct (two normal samples), breast (one fibroadenoma and10 adenocarcinoma samples), testis tumors (three embryonalcarcinomas, two seminomas, and one paratesticular sarcoma),squamous cell carcinomas of lung (two samples) and esophagus(two primary tumors and one lymph node metastasis), adenocarci-nomas of the stomach (two samples) and colon (seven primarytumors and four liver metastases), normal colon of neonate (onesample) and adult (one sample), and endocrine tumor of thepancreas (one sample). Besides this broad screening, we also focusedon larger series of PCas (45 samples) and IBCs (32 samples).In contrast to in vitro observations, we detected only a few human

cell types that were positive for Kaiso at the nuclear level. Theseincluded oocytes (Fig. 1A), eosinophilic granulocytes in themuscularis propria (Fig. 1B), submucosa (not shown) and laminapropria of the colon (Fig. 1J and K), and cells from certain tumors,e.g., a seminoma (Fig. 1C), an undifferentiated paratesticularsarcoma (Fig. 1D) and 3 out of 10 breast adenocarcinomas (Fig.1E). Nuclear Kaiso staining was often associated with cytoplasmicimmunopositivity. In general, however, most cells were completelynegative for nuclear Kaiso, including tumor cells of several breastadenocarcinomas (Fig. 1F), stromal and granulosa cells of ovary (Fig.1A), normal glandular epithelium of the prostate (Fig. 1G),hepatocytes (Fig. 1H), pancreatic endocrine tumor cells (Fig. 1I),and normal epithelial cells of the colon (Fig. 1J and K). In normalprostate epithelium, some cytoplasmic staining was detectable inbasal cells, although there was no or only weak staining in thecytoplasm of secretory epithelial cells and periacinar myofibroblasts(Fig. 1G). In each of 45 PCas analyzed, Kaiso was weakly expressed inthe cytoplasm and apparently completely absent from the nucleus(exemplified in Fig. 2A for a high grade tumor; Gleason score 7). In

serial sections, the same tumor cell field showed retention ofE-cadherin and p120ctn at cell-cell contacts (Fig. 2B and C).Heterogeneous loss of these two proteins was seen in high-gradetumors, but there was no evidence for nuclear p120ctn (to bedescribed in detail elsewhere). Kaiso staining that is weak in thecytoplasm and absent from the nucleus was also seen in 8 out of the32 IBC samples analyzed (Fig. 1L). Conversely, the other 24 IBC

samples were clearly positive for Kaiso in both nucleus and

cytoplasm (Fig. 1M-O). However, staining intensity showed intra-tumoral heterogeneity, and in some cases immunopositive dots in

the perinuclear cytoplasm were observed (arrows, Fig. 1M-O).Staining of consecutive sections of a primary papillary serous

carcinoma of the ovary (Fig. 2D-F) showed that Kaiso (D) andp120ctn (E) were colocalized in the cytoplasm, whereas E-cadherinwas expressed with varying intensities at cell-cell contacts (F). Thispattern was also apparent in stainings of a liver metastasis of thissame ovarian cancer (Fig. 2G-I), where high cytoplasmic Kaiso andp120ctn levels were obvious, whereas E-cadherin was focally absent(Fig. 2I).Another interesting staining pattern was seen in a squamous cell

carcinoma of the esophagus (Fig. 2J-L), its metastasis in the lymphnodes (Fig. 2M), and some adenocarcinomas of the breast (Fig. 2Nand O). These tumors displayed a gradient of Kaiso expression levelsand a diversity of localizations, with prominent nuclear staining incancer cells at the border between tumor nodule and surroundingtissue or in infiltrating parts of the tumor, and with weaker nuclearor mainly cytoplasmic staining in the central parts of the tumor.Convincing nuclear p120ctn was not seen in any of these tumors. Atthe tumor border, the nuclear Kaiso staining was accompanied byp120ctn staining in the cytoplasm and by decreased E-cadherin andp120ctn at cell-cell contacts (e.g., Fig. 2K and L). Cytoplasmic Kaisoand p120ctn were also observed in the peritumoral stroma (Fig. 2Jand K). In general, there was little evidence of p120ctn and Kaisocolocalization in the nucleus, but the proteins were frequentlyobserved together in the cytoplasm.Staining with a newly made pAb (S1337) raised against a

carboxyl-terminal epitope of Kaiso not recognized by mAb 6F, mAb12H, or pAb R gave similar results. An example is shown of IBC withhighly positive cells (Fig. 1O , to be compared with Fig. 1N).Examples of negative staining in the nuclei are given for epithelialcells of normal colon crypts, whereas eosinophilic granulocytes inthe lamina propria showed positive nuclei (Fig. 1K , to be comparedwith Fig. 1J). This confirms the specificity of the Kaiso staining intissue sections.Cells with high cytoplasmic Kaiso levels and absent or weak

expression in the nucleus were also common in several normal andbenign tissues, e.g. ciliated epithelial cells of the oviduct (Fig. 3A) orbronchus (Fig. 3B), biliary duct cells (Fig. 3C), smoothmuscle cells ofthe muscularis propria of the colon of a neonate (Fig. 3D), neuralganglia of the stomach (Fig. 3E), and breast fibroadenoma cells (Fig.3F). In general, cytoplasmic staining in these tissues was manifestedin a punctuate pattern, often in a polarized fashion (Fig. 3A-C),although smooth muscle cells showed dots only close to the nucleus(Fig. 3D), which was quite comparable to the staining results in somecases of IBC (Fig. 1M-O).Subcellular Translocation and Loss of Kaiso Expression in

Human Xenografts. In order to investigate the prominent differ-ences between in vitro and in vivo Kaiso expression, we did xenograftexperiments using human HT29 and SW48 tumor cell lines. HT29cells normally express p120ctn at cell-cell contacts, whereas SW48cells express only a minor amount of p120ctn (7).

Cancer Research

Cancer Res 2005; 65: (6). March 15, 2005 2226 www.aacrjournals.org

Research. on August 31, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: Expression and Nuclear Location of the Transcriptional ...Kaiso is a BTB/POZ zinc finger protein originally described as an interaction partner of p120ctn. In cultured cell lines,

In a first xenograft experiment, cryosections and paraffinsections were made from tumors at different time points afters.c. injection in athymic mice. Stainings in both types of sectionswere comparable. Results of paraffin sections of HT29 xenograftsare presented in Fig. 4, and data from SW48 experiments weresimilar. In both cases, we observed a striking change in thesubcellular localization and the amount of Kaiso as the injectedcells grew into tumors in vivo . All injected cells showed Kaiso-positive nuclei in vitro , as tested by immunofluorescence andimmunohistochemistry of cell cultures embedded in paraffin.In contrast, Kaiso localization in the growing tumors evolved froma diversity of patterns (Fig. 4A and B) to a nearly complete lossof expression (Fig. 4E-H). Cells with nuclear Kaiso were observedonly occasionally, but always at the border of younger tumornodules, where they had close contact with the surroundingstroma. At an intermediate time point (Fig. 4C and D), a possibletranslocation of Kaiso from the nucleus towards the cytoplasmwas detected, especially in the form of granules located around the

nucleus. Here again, use of pAb S1337 yielded quite similar results,with nuclear Kaiso staining in young xenografts (Fig. 4I) and lossof nuclear staining at later stages (Fig. 4J).Because diverse patterns of Kaiso expression were already

present in tumors excised 6 days after inoculation (Fig. 4A), thedata were extended by a second xenograft experiment in whichmice were sacrificed at earlier stages (see Materials andMethods). Again, in vivo findings clearly differed from the in vitrostainings. In general, the results were similar to those of the firstxenograft experiment, with diverse Kaiso localization patternsapparent 2 days after inoculation (data not shown). At 6 days afterinoculation, xenografts were stained for the endogenous hypoxia-regulated marker carbonic anhydrase IX (32). Interestingly, thecenter of the tumor nodules stained positively for carbonicanhydrase IX but lacked nuclear Kaiso, whereas the tumor edgesshowed the inverse staining pattern (Fig. 4K). Tumor cells nearbyblood vessels showed lower carbonic anhydrase IX levels butprominent nuclear Kaiso (Fig. 4L).

Figure 1. Rare nuclear expression ofKaiso in normal and tumoral humantissues. Cryosections (A, C, D, G, I ) andparaffin sections (B, E, F, H, J-O )were stained with mAb 6F against Kaiso,with the exception of K and O, wherepAb S1337 was used. The two antibodieswere raised against completely differentepitopes but yielded essentially identicalresults. 3,3V-Diaminobenzidine was usedas chromogen, and counterstaining waswith hematoxylin. Stromal cells andgranulosa cells of the ovary are negative,whereas the oocyte has a positive nucleus(arrow in A, �400 magnification).Eosinophilic granulocytes in the laminapropria and the muscularis propriaof peritumoral colon tissue showed bilobedKaiso-positive nuclei (arrows in B, �400magnification), whereas the surroundingsmooth muscle cells, mast cells, plasmacells, and neutrophils (data not shown)showed Kaiso-negative nuclei. Nucleusand cytoplasm are Kaiso-positive inseminoma cells (C, �1,000 magnification),in cells of an undifferentiated paratesticularsarcoma (D, �1,000 magnification),and in tumor cells of a subset of breastcancers (E, �400 magnification). Otherbreast adenocarcinoma cells lackeddetectable nuclear Kaiso (F, arrow, rarestromal cell with nuclear positivity; �400magnification). Nuclei of normal glandularepithelial cells of the prostate (G, �400magnification), of hepatocytes (H, thecytoplasmic staining in this section is dueto endogenous peroxidase activity; �400magnification), and of pancreatic endocrinetumor cells (I, �400 magnification) wereessentially negative for Kaiso. A normalcolonic crypt with numerous goblet cellsshowed negative nuclei and positivecytoplasm (J and K, arrows, eosinophilicgranulocytes in the lamina propria; �400magnification). Differences in Kaisoexpression were seen in a series of 32IBC patients: 8 samples showed onlyweak cytoplasmic staining (L, �600magnification), whereas 24 sampleswere clearly positive in the nucleus andcytoplasm (M-O, �600 magnification).Several of the 32 samples showed dotsclose to the nucleus (see arrows in M-O ).

Kaiso Expression Regulated by Tumor Microenvironment

www.aacrjournals.org 2227 Cancer Res 2005; 65: (6). March 15, 2005

Research. on August 31, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: Expression and Nuclear Location of the Transcriptional ...Kaiso is a BTB/POZ zinc finger protein originally described as an interaction partner of p120ctn. In cultured cell lines,

HT29 xenografts at 31 days and SW48 xenografts at 38 days afterinoculation were essentially Kaiso-negative, also at tumor-host in-terfaces. Remarkably, when these tumor cells were put into ex vivoculture, they remained Kaiso-negative for at least 2 days (Fig. 5Afor HT29), but later on, they recovered nuclear staining (Fig. 5E , at 6days after explantation). In the case of HT29, p120ctn staining wasconsistently positive at cell-cell contacts (Fig. 5B and F), andcolocalization of p120ctn with Kaiso was not detected. SW48 cellsshowed no p120ctn staining, as expected (7).Influence of the Microenvironment on Kaiso Expression. In

order to determine the possible effect of hypoxia on Kaisoexpression, we did a comparative study on cultures of HT29 andSW48 cells grown under normoxic versus hypoxic conditions.Cultures at low density invariably showed nuclear Kaiso (illustratedfor HT29 cells in Fig. 6A-D). When cell cultures became very

confluent, Kaiso disappeared from the nucleus. This was againobserved under both hypoxic and normoxic conditions (Fig. 6E-H).Hypoxia was monitored by staining for carbonic anhydrase IX andHIF-1 (data not shown). Loss of Kaiso was seen a few days earlierunder normoxic conditions, which can be explained by the morerapid formation of a superconfluent layer. Upon double staining forp120ctn, no influence of either oxygen pressure or culture densityon p120ctn expression was observed (Fig. 6I and J).We then analyzed the subcellular localization of Kaiso in two-

dimensional versus three-dimensional cultures. We first aimed atconfirming a relationship between cell density and Kaiso proteinexpression in HT29, SW48, and MCF-10A cells. Only cultures thatachieved the highest density at the time of fixation showed regionsnegative for Kaiso in the nucleus. In these regions, cells had piled up,forming semi-three-dimensional cultures.

Figure 2. Immunohistochemical stainingof human tumors for Kaiso, p120ctn andE-cadherin. Consecutive paraffin sectionswere stained in (A-C ), (D-F ), and (J-L). AllPCas obtained from up to 45 patientsshowed Kaiso-negative nuclei andslightly positive cytoplasm (A, �400magnification). The same tumor fieldshowed p120ctn (B, �400 magnification)and E-cadherin at the cell-cell contacts(C, �400 magnification). Kaiso andp120ctn colocalized in the cytoplasm ofthe primary tumor of a papillary serousadenocarcinoma of the ovary (D and E,�400 magnification), whereas E-cadherinwas heterogeneously expressed at cell-cellcontacts (F, �400 magnification). Stainingsof the liver metastasis of this same ovariantumor gave about the same results (G-I,�630 magnification), with prominentcytoplasmic staining for Kaiso andp120ctn. Paraffin sections of a squamouscell carcinoma of the esophagus (J-L,�400 magnification) showed strongnuclear Kaiso staining at the border ofthe tumor nodule and mainly cytoplasmicstaining more centrally in the tumor; thiswas also observed in the lymph nodemetastasis (M, �400 magnification) thatwas obtained from the same patient. Incontrast, both E-cadherin and p120ctnwere expressed at cell-cell contacts in thecenter of the tumor, whereas they werereduced or cytoplasmic at the border(K and L). Likewise, a central part of abreast adenocarcinoma showed largelycytoplasmic Kaiso staining (N, �1,000magnification), whereas an infiltratingpart of the same tumor showed severalKaiso-positive cell nuclei (O, �1,000magnification).

Cancer Research

Cancer Res 2005; 65: (6). March 15, 2005 2228 www.aacrjournals.org

Research. on August 31, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: Expression and Nuclear Location of the Transcriptional ...Kaiso is a BTB/POZ zinc finger protein originally described as an interaction partner of p120ctn. In cultured cell lines,

Next, we examined Kaiso location in full three-dimensionalstructures. HT29 spheroids, formed during 5 days on a bacterial Petridish or on an agar base, were collected and immunostained forKaiso, p120ctn, and E-cadherin. In comparison with subconfluentHT29 monolayers, Kaiso expression was strongly decreased in thesethree-dimensional structures (Fig. 6K and L), and the inner cells ofthe spheroids in particular completely lost nuclear Kaiso. Incontrast, p120ctn and E-cadherin were still expressed at cell-cellcontacts.The formation of acini-like structures by MCF-10A cells was

analyzed in three-dimensional Matrigel cultures. These cells werealso grown on glass cover slips, either uncoated or coated withMatrigel. In the latter controls, strong nuclear and weak cytoplasmicKaiso staining was observed as long as the cultures weresubconfluent (Fig. 7A-D). Morphogenesis in Matrigel of hollowstructures of 1 to 2mm thickness was comparedwith published data(30). Sizes of the forming glandular structures perfectly matchedthese data in time, and (almost) completely hollow acini were formed bydays 10 to 14. At early time points (day 0.5, 1.5, and 3), when cellswere still single or had formed only very small aggregates, Kaisoexpression had already decreased significantly in the nucleus,whereas cytoplasmic levels slightly increased (Fig. 7E and F). At days7 and 14, nuclear Kaiso could no longer be detected, and only cells atthe outer rim (reported as a polarized layer; ref. 30) showed somecytoplasmic Kaiso staining (Fig. 7G and H). Strikingly, apparentlysingle cells sticking to the acini or shed by them contained nuclearKaiso (arrows, Fig. 7G and H). Finally at day 21, when typical acinihad formed, Kaiso immunopositivity completely disappeared (Fig. 7Iand J). Stainings with E-cadherin and p120ctn antibodies showedsatisfactory antibody penetration into these relatively large three-dimensional structures (exemplified in Fig. 7K and L).

Discussion

We have shown that the intracellular localization of Kaiso candeviate dramatically from what has been reported for variouscultured cells. Kaiso is almost always nuclear in cells grown as two-dimensional monolayer cultures, but usually cytoplasmic or evenabsent in vivo . The data we obtained using several highly specificmAbs and pAbs recognizing at least two different Kaiso epitopes wereessentially identical. By directly switching the cells between in vitroand in vivo growth conditions (xenografts and xenograft explants), wehave shown that the nuclear exclusion of Kaiso is completely

reversible. Comparable results were obtained from three-dimensionalcell culture models: Kaiso translocated from the nucleus towards thecytoplasm, and eventually disappeared nearly completely in HT29spheroids cultured in liquid medium and in MCF10A acini-likespheroids grown in reconstituted basement membrane (Matrigel).Our findings reveal a novel and striking cell density–dependenttranslocation and/or down-regulation of Kaiso in a wide varietyof cell types and tissues, both normal and tumoral ones.In vivo , nuclear Kaiso was sometimes observed at tumor borders,

in infiltrating parts of particular human tumors, and in metastases,whereas the majority of normal human tissues and primary tumorshad Kaiso-negative nuclei. The reproducibility of these findings wasshown by analysis of two larger tumor series: a set of 45 PCas ofeither low or high grade, and a set of 32 IBCs. The PCas invariablyshowed Kaiso-negative nuclei, whereas the IBCs showed intratumorand intertumor variability ranging from negative to quite strongKaiso staining in the tumor cell nuclei.Kaiso is generally described as a transcription repressor (19, 20,

22–24). Interestingly, the majority of candidate Kaiso target genesidentified thus far, i.e., CDH1 (E-cadherin), S100A4 (mts-1), matrilysin(MMP7), MTA2, andWnt11 have been linked with development and/or cancer. The absence of nuclear Kaiso in many of the humantissues examined may reflect a requirement for the expression ofcertain of these Kaiso-repressible genes. E-cadherin is the prototypiccell-cell adhesion molecule in epithelial cells and is a renownedtumor and invasion suppressor (40). Both MTA1 and MTA2 arecomponents of NuRD ATP-dependent chromatin remodeling andhistone deacetylase complexes. Whereas the transcription factorcomplex that contains MTA-1 is highly expressed in metastatic cells,a housekeeping role is suggested for the complex that contains thehomologous MTA2 (41). Both S100A4 and matrilysin are known fortheir importance in malignant invasion (42, 43). Down-regulation ofthe Wnt11 proximal promoter by Kaiso was recently shown in theXenopus model system (24). The Wnt11 protein contributes via anoncanonical, h-catenin-independent Wnt signaling pathway toplanar cell polarity and morphogenetic movements such asconvergent extension. More particularly, inhibition of Xenopus Kaisoby antisense morpholino oligonucleotides or by nuclear p120ctn wasshown to interfere with normal gastrulation movements, axialelongation, and neural fold closure (24). Expression of WNT11 is up-regulated in various human tumor cell lines and tumors (44),including high-grade prostate tumors and androgen-independent

Figure 3. Cytoplasmic expressionof Kaiso in normal human tissues andbenign lesions. Paraffin sections(A, B, C, D ) and cryosections (E and F )were stained. 3,3V-Diaminobenzidine wasused as chromogen and counterstainingwas with hematoxylin. Strong cytoplasmicKaiso staining was detected in somecases, e.g., ciliated epithelial cells of theoviduct (A, �1,000 magnification) and thebronchus (B, �1,000 magnification). Biliaryduct cells showed cytoplasmic dot-likeKaiso immunoreactivity (C, �1,000magnification). In smooth muscle cellsof the muscularis propria of the colonof a neonate, Kaiso-positive dots wereseen in apparent association with thenuclei (arrows in D, �1,000 magnification).Neural ganglia of the stomach (arrows in E,� 400 magnification) and breastfibroadenoma cells (F, �630 magnification)showed more homogenous cytoplasmicstaining.

Kaiso Expression Regulated by Tumor Microenvironment

www.aacrjournals.org 2229 Cancer Res 2005; 65: (6). March 15, 2005

Research. on August 31, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: Expression and Nuclear Location of the Transcriptional ...Kaiso is a BTB/POZ zinc finger protein originally described as an interaction partner of p120ctn. In cultured cell lines,

prostate cancer cell lines and xenografts (45). Treatment of IEC6 ratintestinal epithelial cells with Wnt11 down-regulates E-cadherin-mediated cell-cell contacts and stimulates cell migration andcontact-independent cell growth (46). This implicates nuclearKaiso in control of particular cell migration events. The diversityof Kaiso localization in many of the tumors described herein maybe related to these observations, although the molecular detailsand functional consequences remain poorly understood.Our data suggest that the microenvironment has a crucial

causative role in regulating Kaiso expression and localizationpatterns. Modulating the microenvironment of HT29, SW48, andMCF-10A cells changed Kaiso levels and switched its localizationbetween the cytoplasm and the nucleus. The fact that thesephenomena were readily reversible suggests that they are not relatedto changes in DNA methylation. Differences between autonomousgrowth of cells in two and three dimensions may be importantdeterminants. Other factors, such as the presence or absence ofstroma and/or oxygen levels were suggested to be instrumental withthe finding that nuclear Kaiso was frequently maintained along theperiphery of tumor xenografts, and was also associated with themore malignant parts in several tumors. In keeping with this, weobserved an inverse relationship between expression of carbonicanhydrase IX, a frequently used marker for hypoxia, and nuclearKaiso in young HT29 xenografts.In order to define the microenvironmental factors that modulate

Kaiso expression patterns more precisely, we tested severalrelevant parameters directly. In vitro cultivation of HT29 orSW48 cells under hypoxic conditions did not by itself abolishexpression of nuclear Kaiso. Loss of nuclear Kaiso was observed indense cultures, but this happened under both normoxic andhypoxic conditions. This suggests that progressive Kaiso loss intumor cell nuclei in xenografts is also mainly related to high celldensity rather than to hypoxia. One possibility is that thresholdlevels of paracrine down-regulating factors are more easily reached

under these conditions. Indeed, three-dimensional spheroids ofHT29 cells grown without supplementation with extracellularmatrix components also lost nuclear Kaiso. Likewise, three-dimensional cultures of MCF-10A cells in Matrigel showedtranslocation of nuclear Kaiso to the cytoplasm, but this couldoccur early after seeding, even at the single-cell stage. Later on,when a polarized differentiated phenotype appeared in acini-likestructures, Kaiso expression was lost completely. In contrast,when these cells were cultured on either uncoated or Matrigel-coated glass cover slips, strong nuclear Kaiso was retained. Ourdata indicate that close cell contact with either the extracellularmatrix in a three-dimensional matrix, or with fellow cells indense cultures or spheroids simulating a three-dimensional tissue,could trigger Kaiso down-regulation. This agrees well with thepaucity of nuclear Kaiso in normal tissues, where cells arebounded in three dimensions either by fellow cells or by extra-cellular matrix.Presently, we do not know why a small fraction of normal cell

types express nuclear Kaiso or why only particular tumor cells do so.Consistent expression of nuclear Kaiso in two-dimensional culturesof tumor cell lines may point to a dedifferentiation process resultingfrom insufficient contact with extracellular matrix or surroundingcells. For some tumor types, a similar dedifferentiation withconsequent insensitivity to microenvironmental cues and acquisi-tion of a higher migratory behavior may explain the occurrence ofnuclear Kaiso. Alternatively, nuclear Kaiso in particular tumors maybe directly induced by activated oncogenes or loss of tumorsuppressor genes, and lead to a more invasive phenotype by virtueof the transcriptional activities of Kaiso. The expression of nuclearKaiso in cell types with diffusely infiltrating growth patterns, such aseosinophilic granulocytes and paratesticular sarcoma cells, fits thishypothesis. Nuclear Kaiso in infiltrating parts of otherwise coherenttumor nodules and in metastases may reflect the dedifferentiation

Figure 4. Changes in Kaiso expression patterns in progressively growing HT29 xenografts. Immunohistochemical staining of paraffin sections with Kaiso-specificmAb 6F was done on different stages of tumor growth: 6 days after s.c. inoculation for (A and B ), 11 days for (C and D ), 14 days for (E and F ), 20 days for (G and H )and 6 days for (K and L). Antibody specificity was confirmed using pAb S1337, as exemplified for xenografts on post-inoculation day 6 (I ) and 14 (J ). In all thesecases, 3,3V-diaminobenzidine was used as chromogen, and counterstaining was with hematoxylin. The growing tumors evolved from a high nuclear Kaiso expression (Aand B ) to a more dot-like cytoplasmic expression (C and D ), and finally to an expression that is very weak or totally absent (E-H ). In (K and L) expression of thehypoxia marker carbonic anhydrase IX was revealed by mAb M75 and visualized with a fuchsia-colored reaction product. Arrows, tumor edge (K ) and a blood vessel (L),respectively, where decreased carbonic anhydrase IX staining correlates with the occurrence of nuclear Kaiso. Magnifications: �100 for A, C, E and G; �200 forK; �400 for L; �600 for I and J ; and �1,000 for B, D, F and H.

Cancer Research

Cancer Res 2005; 65: (6). March 15, 2005 2230 www.aacrjournals.org

Research. on August 31, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: Expression and Nuclear Location of the Transcriptional ...Kaiso is a BTB/POZ zinc finger protein originally described as an interaction partner of p120ctn. In cultured cell lines,

and higher migratory activities at these locations. However, thenature of most genes known to be candidate targets for inhibitionby Kaiso, predicts an inhibition rather than a stimulation ofinvasion by nuclear Kaiso. It may therefore be worthwhile in thefuture to analyze three-dimensional cultures of MCF-10A cellstransformed with various oncogenes (47, 48) for subcellularexpression patterns of Kaiso in relation to loss of cell polarity andacquisition of invasive properties. It is also important to keep inmind that tumors are influenced by the state of activation of thesurrounding stroma (49). From the above, it is evident that not allfindings on Kaiso subcellular localization can be readily explained.For instance, the observation that clinically localized PCas expressno nuclear Kaiso regardless of grade, shows that aggressive tumorsdo not invariably have nuclear Kaiso.Kaiso’s interaction partner p120ctn has been reported to be partly

nuclear in E-cadherin-deficient tumor cell lines (10) and in a minorfraction of lobular breast cancers with complete loss of E-cadherin

Figure 5. Expression of Kaiso (A and E ) and p120ctn (B and F ) in HT29cells explanted from xenograft tumors. Monoclonal antibodies were used forboth antigens, followed by an Alexa 488-conjugated anti-mouse secondaryantibody. DNA was stained with 4V,6-diamidino-2-phenylindole (C, D, G and H ).Pictures were made with a Zeiss Axiophot microscope. Kaiso negativity wasseen directly after tissue explantation (A ). When kept in culture for 7 days, cellsrecovered Kaiso expression in the nucleus (E ). p120ctn was positive at thecell-cell contacts in both cases (B and F ; �1,000 magnification).

Figure 6. Influence of microenvironmental factors on expression of Kaiso andp120ctn in HT29 cells. Subconfluent (A-D ) and highly confluent two-dimensionalcell cultures (E-J ) were analyzed after growth under normoxic (left ) andhypoxic conditions (right ). A double staining was done using pAb S1337 forKaiso detection and mAb pp120 for p120ctn detection. DNA was stained with4V,6-diamidino-2-phenylindole (B, D, G, H ). Kaiso was largely nuclear insubconfluent cultures (A and C ), whereas high confluency resulted in nuclearloss of Kaiso irrespective of oxygen pressure (E and F ). No change inexpression of p120ctn was observed (I and J ). HT29 cells cultured asthree-dimensional spheroids showed loss of nuclear Kaiso in the nucleus (K andL). mAb 6F and secondary Alexa 488-conjugated anti-mouse antibody wereused for Kaiso staining (K ). DNA was counterstained with PI (L ). Pictures weremade with a Zeiss Axiophot microscope (A-D ), a Leica DM IRE2 microscope(E-J ), and a Zeiss LSM 410 confocal microscope (K and L ). Bars, 10 AM.

Kaiso Expression Regulated by Tumor Microenvironment

www.aacrjournals.org 2231 Cancer Res 2005; 65: (6). March 15, 2005

Research. on August 31, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 9: Expression and Nuclear Location of the Transcriptional ...Kaiso is a BTB/POZ zinc finger protein originally described as an interaction partner of p120ctn. In cultured cell lines,

(50). Hence, it is surprising that we were unable to detectcolocalization of Kaiso with p120ctn in the nucleus. In contrast,the presence of both proteins in the cytoplasm was readily seen invarious tumors, including the xenografts. For pancreatic carcino-mas, p120ctn was reported to occur in both the cytoplasm andnucleus, particularly in the case of high-grade undifferentiatedtumors (51). Recent studies on breast cancer revealed frequentcytoplasmic p120ctn in lobular carcinomas and atypical lobularhyperplasias, but rarely in ductal tumors (50, 52). Cytoplasmicp120ctn is known to modulate the activity of small GTPases, tocontribute to growth factor-induced cell migration, and to regulateassembly and stability of E-cadherin-dependent cell junctions (6). Itis conceivable that Kaiso is retained in the cytoplasm or excludedfrom the nucleus by virtue of its interaction with cytoplasmicp120ctn. Although it has been shown that nuclear p120ctn relievesKaiso-mediated transcriptional repression in cultured cells, whereascytoplasmic p120ctn does not (24, 26), the common cytoplasmiclocation of Kaiso in vivo is consistent with a mechanism whereby

p120ctn inhibits Kaiso transcriptional repression by sequestering itin the cytoplasm rather than by directly blocking its binding to DNA.Physical dissociation between cytoplasmic p120ctn and nuclearKaiso, as we observed in particular tumors or tumor fields, may thuscontribute to tumor progression. On the other hand, we cannotpresently exclude that other p120ctn-independent mechanismsexist for cytoplasmic sequestration and down-regulation of Kaiso.Further studies are needed to elucidate the exact role of possibleKaiso-p120ctn interactions in the cytoplasm, as well as a possiblecorrelation with activity of small GTPases.In summary, we have shown that the microenvironment contains

crucial information that modulates Kaiso expression and subcellu-lar localization. Our observations point to an important role fornuclear Kaiso in particular cases of dynamic cell behavior anddedifferentiation. Further research may determine the exactunderlying rules that are likely to differ on a case-by-case basis.Given that high levels of nuclear Kaiso observed in monolayersin vitro are not representative of nonpathologic situations in vivo

Figure 7. Dynamic intracellular localization of Kaiso in MCF-10A cells. mAb 6F was used for Kaiso, followed by an Alexa 488-conjugated anti-mouse secondaryantibody (A, C, E, G, I, and J ). DNA was stained with PI (B, D, F, H, J, and L ). Kaiso positivity was seen in subconfluent cell layers, on glass cover slips (A) and also oncover slips coated with Matrigel (C ). Arrows, nucleoli that are Kaiso-negative (A-D ). When seeded in a thick Matrigel layer, Kaiso translocated from the nucleus towardsthe cytoplasm within half a day (E ). On day 14, typical acini had formed, and Kaiso could no longer be detected, with the exception of cells at the periphery, whichshowed cytoplasmic staining (G ). Arrow, a solitary cell sticking to the spheroid clearly showed nuclear Kaiso expression. On day 21 (I ), Kaiso had become completelyundetectable. Several controls were included for staining efficiency and acinus organization, such as staining with mAb human epithelial cadherin-1 recognizingE-cadherin (K ). Pictures were made by confocal microscopy. Bars, 10 AM.

Cancer Research

Cancer Res 2005; 65: (6). March 15, 2005 2232 www.aacrjournals.org

Research. on August 31, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 10: Expression and Nuclear Location of the Transcriptional ...Kaiso is a BTB/POZ zinc finger protein originally described as an interaction partner of p120ctn. In cultured cell lines,

(or in vitro three-dimensional), experimental conditions forstudying the physiologic role(s) of Kaiso in either normal tissuesor malignant tumors need to be thoughtfully reconsidered.

AcknowledgmentsReceived 6/8/2004; revised 12/3/2004; accepted 1/6/2005.

Grant support: FWO, the Geconcerteerde Onderzoeksacties of Ghent University,Fortis Verzekeringen (Belgium), and Interuniversity Attraction Poles Programme,

Belgian Science Policy. Stichting Emmanuel van der Schueren and by a BOF grant fromGent University (A. Soubry). J. van Hengel is a postdoctoral fellow with the FWO (Fundfor Scientific Research, Flanders).

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 Drs. J. Brugge and J. Debnath for MCF-10A cells, protocols, and fortheir three-dimensional culture; Dr. P. Brouckaert for his help with the xenografts;Dr. G. Berx for constructive discussions; Hilde Hellemans, Petra D’Hooge, andBarbara Gilbert for technical assistance; and Dr. Amin Bredan for editorial assistance.

References1. Daniel JM, Reynolds AB. The catenin p120(Ctn)interacts with Kaiso, a novel BTB/POZ domainzinc finger transcription factor. Mol Cell Biol 1999;19:3614–23.

2. Reynolds AB, Roesel DJ, Kanner SB, Parsons JT.Transformation-specific tyrosine phosphorylation of anovel cellular protein in chicken cells expressingoncogenic variants of the avian cellular src gene. MolCell Biol 1989;9:629–38.

3. Kanner SB, Reynolds AB, Parsons JT. Tyrosinephosphorylation of a 120-kilodalton pp60src substrateupon epidermal growth factor and platelet-derivedgrowth factor receptor stimulation and in polyomavirusmiddle-T-antigen-transformed cells. Mol Cell Biol1991;11:713–20.

4. Downing JR, Reynolds AB. PDGF, CSF-1, and EGFinduce tyrosine phosphorylation of p120, a pp60srctransformation-associated substrate. Oncogene 1991;6:607–13.

5. Anastasiadis PZ, Reynolds AB. The p120 cateninfamily: complex roles in adhesion, signaling and cancer.J Cell Sci 2000;113:1319–34.

6. Reynolds AB, Roczniak-Ferguson A. Emerging roles forp120-catenin in cell adhesion and cancer. Oncogene2004;23:7947–56.

7. Ireton RC, Davis MA, van Hengel J, et al. A novel rolefor p120 catenin in E-cadherin function. J Cell Biol 2002;159:465–76.

8. Davis MA, Ireton RC, Reynolds AB. A core function forp120-catenin in cadherin turnover. J Cell Biol 2003;163:525–34.

9. Xiao KY, Allison DF, Buckley KM, et al. Cellular levelsof p120 catenin function as a set point for cadherinexpression levels in microvascular endothelial cells.J Cell Biol 2003;163:535–45.

10. van Hengel J, Vanhoenacker P, Staes K, van Roy F.Nuclear localization of the p120ctn Armadillo-likecatenin is counteracted by a nuclear export signaland by E-cadherin expression. Proc Natl Acad Sci U S A1999;96:7980–5.

11. Thoreson MA, Reynolds AB. Altered expression of thecatenin p120 in human cancer: implications for tumorprogression. Differentiation 2002;70:583–9.

12. Anastasiadis PZ, Reynolds AB. Regulation of RhoGTPases by p120-catenin. Curr Opin Cell Biol 2001;13:604–10.

13. Dhordain P, Albagli O, Lin RJ, et al. CorepressorSMRT binds the BTB/POZ repressing domain of theLAZ3/BCL6 oncoprotein. Proc Natl Acad Sci U S A 1997;94:10762–7.

14. Hong SH, David G, Wong CW, Dejean A, PrivalskyML. SMRT corepressor interacts with PLZF and withthe PML-retinoic acid receptor a (RARa) and PLZF-RARa oncoproteins associated with acute promyelo-cytic leukemia. Proc Natl Acad Sci U S A 1997;94:9028–33.

15. de la Luna S, Allen KE, Mason SL, La Thangue NB.Integration of a growth-suppressing BTB/POZ domainprotein with the DP component of the E2F transcrip-tion factor. EMBO J 1999;18:212–28.

16. Reuter S, Bartelmann M, Vogt M, et al. APM-1, a novelhuman gene, identified by aberrant co-transcriptionwith papillomavirus oncogenes in a cervical carcinomacell line, encodes a BTB/POZ-zinc finger protein withgrowth inhibitory activity. EMBO J 1998;17:215–22.

17. Deltour S, Pinte S, Guerardel C, Wasylyk B, LeprinceD. The human candidate tumor suppressor gene HIC1recruits CtBP through a degenerate GLDLSKK motif.Mol Cell Biol 2002;22:4890–901.

18. Huynh KD, Bardwell VJ. The BCL-6 POZ domainand other POZ domains interact with the co-repressorsN-CoR and SMRT. Oncogene 1998;17:2473–84.

19. Yoon HG, Chan DW, Reynolds AB, Qin J, Wong JM. N-CoR mediates DNA methylation-dependent repressionthrough a methyl CpG binding protein Kaiso. Mol Cell2003;12:723–34.

20. Prokhortchouk A, Hendrich B, Jorgensen H, et al.The p120 catenin partner Kaiso is a DNA methylation-dependent transcriptional repressor. Genes Dev 2001;15:1613–8.

21. Bird A. DNA methylation patterns and epigeneticmemory. Genes Dev 2002;16:6–21.

22. Ruzov A, Dunican DS, Prokhortchouk A, et al. Kaisois a genome-wide repressor of transcription that isessential for amphibian development. Development2004;131:6185–94.

23. Daniel JM, Spring CM, Crawford HC, Reynolds AB,Baig A. The p120(Ctn)-binding partner Kaiso is a bi-modal DNA-binding protein that recognizes both asequence-specific consensus and methylated CpGdinucleotides. Nucleic Acids Res 2002;30:2911–9.

24. Kim SW, Park J-I, Spring CM, et al. Non-canonicalWnt signals are modulated by the Kaiso transcriptionalrepressor and p120-catenin. Nat Cell Biol 2004;6:1212–20.

25. Rodova M, Kelly KF, VanSaun M, Daniel JM, WerleMJ. Regulation of the rapsyn promoter by Kaiso and y-catenin. Mol Cell Biol 2004;24:7188–96.

26. Kelly KF, Spring CM, Otchere AA, Daniel JM. NLS-dependent nuclear localization of p120ctn is necessaryto relieve Kaiso-mediated transcriptional repression.J Cell Sci 2004;117:2675–786.

27. Daniel JM, Reynolds AB. The tyrosine kinasesubstrate p120(cas) binds directly to E-cadherin butnot to the adenomatous polyposis coli protein or a-catenin. Mol Cell Biol 1995;15:4819–24.

28. Daniel JM, Ireton RC, Reynolds AB. Monoclonalantibodies to Kaiso: a novel transcription factor andp120(Ctn)-binding protein. Hybridoma 2001;20:159–66.

29. Soule H, Maloney T, Wolman S, et al. Isolation andcharacterization of a spontaneously immortalizedhuman breast epithelial cell line, MCF-10. Cancer Res1990;50:6075–86.

30. Debnath J, Muthuswamy SK, Brugge JS. Morphogen-esis and oncogenesis of MCF-10A mammary epithelialacini grown in three-dimensional basement membranecultures. Methods 2003;30:256–68.

31. Janssens B, Goossens S, Staes K, et al. aT-Catenin:a novel tissue-specific h-catenin-binding protein me-diating strong cell-cell adhesion. J Cell Sci 2001;114:3177–88.

32. Wykoff CC, Beasley NJ, Watson PH, et al. Hypoxia-inducible expression of tumor-associated carbonicanhydrases. Cancer Res 2000;60:7075–83.

33. Colpaert CG, Vermeulen PB, Benoy I, et al. Inflam-matory breast cancer shows angiogenesis with highendothelial proliferation rate and strong E-cadherinexpression. Br J Cancer 2003;88:718–25.

34. Schroder FH, Hermanek P, Denis L, Fair WR,Gospodarowicz MK, Pavone-Macaluso M. The TNMclassification of prostate cancer. Prostate Suppl1992;4:129–38.

35. Gleason DF, Mellinger GT. Prediction of prognosis forprostatic adenocarcinoma by combined histologicalgrading and clinical staging. J Urol 1974;111:58–64.

36. Colpaert CG, Vermeulen PB, Van Beest P, et al.Cutaneous breast cancer deposits show distinctgrowth patterns with different degrees of angiogene-sis, hypoxia and fibrin deposition. Histopathology2003;42:530–40.

37. Fordel E, Geuens E, Dewilde S, et al. Cytoglobinexpression is upregulated in all tissues upon hypoxia: anin vitro and in vivo study by quantitative real-time PCR.Biochem Biophys Res Commun 2004;319:342–8.

38. Yuhas J, Li A, Martinez A, Ladman A. A simplifiedmethod for production and growth of multicellulartumor spheroids. Cancer Res 1977;37:3639–43.

39. van Hengel J, Gohon L, Bruyneel E, et al. Proteinkinase C activation upregulates intercellular adhesionof a-catenin-negative human colon cancer cell va-riants via induction of desmosomes. J Cell Biol 1997;137:1103–16.

40. Strumane K, Berx G, van Roy F. Cadherins in cancer.In: Behrens J, Nelson J, editors. Cell Adhesion. Vol. 165.Heidelberg (Germany): Springer-Verlag; 2004. p. 69–103.

41. Yao YL, Yang WM. The metastasis-associated pro-teins 1 and 2 form distinct protein complexes withhistone deacetylase activity. J Biol Chem 2003;278:42560–8.

42. Barraclough R. Calcium-binding protein S100A4 inhealth and disease. Biochim Biophys Acta-Mol Cell Res1998;1448:190–9.

43. Fingleton BM, Goss KJH, Crawford HC, Matrisian LM.Matrilysin in early stage intestinal tumorigenesis.APMIS 1999;107:102–10.

44. Kirikoshi H, Sekihara H, Katoh M. Molecular cloningand characterization of human WNT11. Int J Mol Med2001;8:651–6.

45. Zhu H, Mazor M, Kawano Y, et al. Analysis of Wntgene expression in prostate cancer: mutual inhibitionby WNT11 and the androgen receptor. Cancer Res 2004;64:7918–26.

46. Ouko L, Ziegler TR, Gu LH, Eisenberg LM, Yang VW.Wnt11 signaling promotes proliferation, transforma-tion, and migration of IEC6 intestinal epithelial cells.J Biol Chem 2004;279:26707–15.

47. Muthuswamy SK, Li DM, Lelievre S, Bissell MJ,Brugge JS. ErbB2, but not ErbB1, reinitiates proliferationand induces luminal repopulation in epithelial acini.Nat Cell Biol 2001;3:785–92.

48. Wrobel CN, Debnath J, Lin E, Beausoleil S, Roussel MF,Brugge JS. Autocrine CSF-1R activation promotes Src-dependent disruption of mammary epithelial architec-ture. J Cell Biol 2004;165:263–73.

49. Mueller MM, Fusenig NE. Friends or foes—bipolareffects of the tumour stroma in cancer. Nat Rev Cancer2004;4:839–49.

50. Sarrio D, Perez-Mies B, Hardisson D, et al. Cytoplas-mic localization of p120ctn and E-cadherin losscharacterize lobular breast carcinoma from preinvasiveto metastatic lesions. Oncogene 2004;23:3272–83.

51. Mayerle J, Friess H, Buchler MW, et al. Up-regulation,nuclear import, and tumor growth stimulation of theadhesion protein p120ctn in pancreatic cancer. Gastro-enterology 2003;124:949–60.

52. Shibata T, Kokubu A, Sekine S, Kanai Y, Hirohashi S.Cytoplasmic p120ctn regulates the invasive phenotypesof E-cadherin-deficient breast cancer. Am J Pathol2004;164:2269–78.

Kaiso Expression Regulated by Tumor Microenvironment

www.aacrjournals.org 2233 Cancer Res 2005; 65: (6). March 15, 2005

Research. on August 31, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 11: Expression and Nuclear Location of the Transcriptional ...Kaiso is a BTB/POZ zinc finger protein originally described as an interaction partner of p120ctn. In cultured cell lines,

2005;65:2224-2233. Cancer Res   Adelheid Soubry, Jolanda van Hengel, Eef Parthoens, et al.   MicroenvironmentRepressor Kaiso Is Regulated by the Tumor Expression and Nuclear Location of the Transcriptional

  Updated version

  http://cancerres.aacrjournals.org/content/65/6/2224

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/65/6/2224.full#ref-list-1

This article cites 51 articles, 27 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/65/6/2224.full#related-urls

This article has been cited by 6 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

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

.http://cancerres.aacrjournals.org/content/65/6/2224To request permission to re-use all or part of this article, use this link

Research. on August 31, 2021. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from