Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Nuclear Kaiso Expression Is Associated with High Grade and Triple-Negative Invasive Breast Cancer

  • Jeroen F. Vermeulen ,

    Contributed equally to this work with: Jeroen F. Vermeulen, Robert A. H. van de Ven

    Affiliation Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands

  • Robert A. H. van de Ven ,

    Contributed equally to this work with: Jeroen F. Vermeulen, Robert A. H. van de Ven

    Affiliation Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands

  • Cigdem Ercan,

    Affiliation Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands

  • Petra van der Groep,

    Affiliations Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, Utrecht, The Netherlands

  • Elsken van der Wall,

    Affiliation Division of Internal Medicine and Dermatology, University Medical Center Utrecht, Utrecht, The Netherlands

  • Peter Bult,

    Affiliation Department of Pathology, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands

  • Matthias Christgen,

    Affiliation Institute of Pathology, Hannover Medical School, Hannover, Germany

  • Ulrich Lehmann,

    Affiliation Institute of Pathology, Hannover Medical School, Hannover, Germany

  • Juliet Daniel,

    Affiliation Department of Biology, McMaster University, Hamilton, Onatrio, Canada

  • Paul J. van Diest,

    Affiliation Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands

  • Patrick W. B. Derksen

    p.w.b.derksen@umcutrecht.nl

    Affiliation Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands

Abstract

Kaiso is a BTB/POZ transcription factor that is ubiquitously expressed in multiple cell types and functions as a transcriptional repressor and activator. Little is known about Kaiso expression and localization in breast cancer. Here, we have related pathological features and molecular subtypes to Kaiso expression in 477 cases of human invasive breast cancer. Nuclear Kaiso was predominantly found in invasive ductal carcinoma (IDC) (p = 0.007), while cytoplasmic Kaiso expression was linked to invasive lobular carcinoma (ILC) (p = 0.006). Although cytoplasmic Kaiso did not correlate to clinicopathological features, we found a significant correlation between nuclear Kaiso, high histological grade (p = 0.023), ERα negativity (p = 0.001), and the HER2-driven and basal/triple-negative breast cancers (p = 0.018). Interestingly, nuclear Kaiso was also abundant in BRCA1-associated breast cancer (p<0.001) and invasive breast cancer overexpressing EGFR (p = 0.019). We observed a correlation between nuclear Kaiso and membrane-localized E-cadherin and p120-catenin (p120) (p<0.01). In contrast, cytoplasmic p120 strongly correlated with loss of E-cadherin and low nuclear Kaiso (p = 0.005). We could confirm these findings in human ILC cells and cell lines derived from conditional mouse models of ILC. Moreover, we present functional data that substantiate a mechanism whereby E-cadherin controls p120-mediated relief of Kaiso-dependent gene repression. In conclusion, our data indicate that nuclear Kaiso is common in clinically aggressive ductal breast cancer, while cytoplasmic Kaiso and a p120-mediated relief of Kaiso-dependent transcriptional repression characterize ILC.

Introduction

Kaiso was initially identified as a binding partner of the adherens junction (AJ) complex member p120-catenin (p120) [1]. Kaiso is a member of the BTB/POZ-ZF (Broad complex, Tramtrak, Bric á brac/Pox virus and zinc finger) family of transcription factors [2] consisting of approximately 60 BTB/POZ-ZF members that include the cancer-associated B cell lymphoma 6 (BCL6), lymphoma-related factor (LRF), and hypermethylated in cancer (HIC1) genes (reviewed in [3]). Kaiso (also known as zinc finger- and BTB domain-containing protein 33; ZBTB33) interacts with its target gene promoters via two distinct mechanisms: via sequence-specific Kaiso binding sites (KBS), consisting of the consensus sequence CTGCNA, or via methylated CpG dinucleotides [4][7]. Although Kaiso can act as a transcriptional activator [8], it mainly acts as a transcriptional repressor by binding to the promoters of its target genes. This interaction can be inhibited by p120 binding to a region flanking Kaiso’s ZF motifs [1] and results in expression of distinct target genes [9], [10]. Kaiso has been shown to directly repress canonical Wnt targets via TCF/LEF family members [11], [12]. These target genes include the matrix metalloprotease Matrilysin (aka MMP7), CCND1, Siamois, Fos and Myc [4], [12]. In addition, Kaiso can regulate expression of Wnt11, a regulator of directed cell movement and morphogenesis [10].

While there is data demonstrating a role for Kaiso in early vertebrate development [12], [13], data implicating Kaiso-mediated regulation of gene transcription in cancer are scarce. Kaiso expression and sub-cellular localization seems dynamic and highly dependent on tumor type and micro-environmental conditions [14], [15]. Interestingly, Kaiso-null mice show resistance to intestinal cancer characterized by a delayed onset of tumor development, decreased tumor size, and prolonged survival when crossed with APCMIN/+ mice [16].

Loss of E-cadherin and subsequent disruption of AJ function is strongly linked to breast cancer development and progression (reviewed in [17]). Using tissue-specific and conditional mouse models, we have established a causal relationship between early inactivation of E-cadherin and formation of invasive lobular carcinoma (ILC) [18], [19]. While β-catenin is rapidly degraded upon loss of E-cadherin [20], [21], p120 translocates and resides in the cytosol [22], where it regulates anchorage-independent tumor growth and metastasis through Mrip-dependent activation of the Rock pathway [20]. In addition, cytoplasmic p120 has been implicated in the acquisition of motility and invasiveness in E-cadherin negative breast cancer [23], [24]. The structure of p120 reveals a number of domains including a protein-protein interaction Armadillo (Arm) domain consisting of 10 Armadillo repeats. This domain mediates not only the interaction with cadherins but also p120 binding to the transcriptional repressor Kaiso, probably in a mutually exclusive manner [1].

Given the importance of p120 in the pathobiology of breast cancer, and its regulation of Kaiso-mediated transcriptional repression, we performed a comprehensive analysis of Kaiso expression and localization to pathological features and molecular subtypes in 477 cases of invasive breast cancer.

Materials and Methods

Patients

The study population was derived from the archives of the Departments of Pathology of the University Medical Center Utrecht, Utrecht, and the Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands. These comprised 477 cases of invasive breast cancer, including cases with a BRCA1 germ-line mutation as previously described [25]. Histological grade was assessed according to the Nottingham scheme, and mitotic activity index (MAI) was assessed as before [26]. From representative donor paraffin blocks of the primary tumors, tissue microarrays were constructed by transferring tissue cylinders of 0.6 mm (3 cylinders per tumor) from the tumor area, determined by a pathologist based on haematoxylin-eosin stained slides, using a tissue arrayer (Beecher Instruments, Sun Prairie, WI, USA) as described before [27]. The use of anonymous or coded leftover material for scientific purposes is part of the standard treatment contract with patients in The Netherlands [28]. Ethical approval was not required.

thumbnail
Figure 1. Kaiso expression in normal breast epithelium.

Sections were stained using immunohistochemistry for Kaiso (middle panel) or E-cadherin (right panel). Kaiso expression in normal breast epithelium is heterogeneous; high nuclear expression (top row) versus only cytoplasmic expression (bottom row) is found with equal frequency. Left panels show a haematoxylin and eosin (H&E) staining. Size bars equal 50 µm, in inserts 25 µm.

https://doi.org/10.1371/journal.pone.0037864.g001

Immunohistochemistry

Immunohistochemistry was carried out on 4 µm thick sections. After deparaffination and rehydration, endogenous peroxidase activity was blocked for 15 min in a 46 mM citric acid-100 mM sodiumphosphate buffer solution pH5.8 containing 0.3% hydrogen peroxide. After antigen retrieval, i.e. boiling for 20 min in 10 mM citrate pH6.0 (Kaiso, p120, PR), Tris/EDTA pH9.0 (E-cadherin, ERα, HER2), or Prot K (0.15 mg/ml, DAKO, Glostrup Denmark) for 5 min at room temperature (EGFR), a cooling period of 30 min preceded the primary antibody incubation. Kaiso (clone 6F, Upstate, Billerica, MA, USA) [29] 1∶100; E-cadherin (clone 4A2C7, Zymed, Invitrogen, Breda, The Netherlands) 1∶200; ERα (clone ID5, DAKO) 1∶80; PR (clone PgR636, DAKO) 1∶25; HER2 (SP3, Neomarkers, Duiven, The Netherlands) 1∶100 were diluted in PBS containing 1% BSA and incubated for 1 h at room temperature. Primary antibodies against p120 (cat 610134, BD Transduction Labs, San Diego, CA, USA) 1∶500 and EGFR (clone 31G7, Zymed, Invitrogen) 1∶30 were diluted in PBS containing 1% BSA and incubated over night at 4°C. The signal was amplified using Powervision poly-HRP anti-mouse, rabbit, rat (DPVO-HRP, Immunologic, Duiven, The Netherlands) or the Novolink kit (Leica, Rijswijk, The Netherlands) (in the case of EGFR) and developed with diaminobenzidine, followed by counterstaining with haematoxylin, dehydration in alcohol, and mounting. Appropriate negative and positive controls were used throughout.

Scoring of Immunohistochemistry

All scoring was done blinded to patient characteristics and results of other staining by two independent observers. E-cadherin and EGFR stainings were scored using the DAKO/HER2 scoring system for membranous staining. Membranous scores 1+, 2+, and 3+ were considered positive, except for HER2 where only a score of 3+ was considered positive. Kaiso staining was scored based on localization and by counting the positive tumor nuclei, considering samples with more than 5% positive tumor nuclei as positive. Using thresholds of 1 or 10% for scoring nuclear accumulation as positive did not change the results. p120 staining was scored based on the localization as membranous or cytoplasmic.

Based on ERα, PR, and HER2 immunohistochemistry, tumors were classified as luminal (ERα and/or PR positive), HER2-driven (ERα-, PR-, HER2+), or basal-like/triple negative (ERα-, PR-, HER2- with or without EGFR expression), the immunohistochemical surrogate [30] of the original Sorlie/Perou classification [31].

Statistics

Statistical analysis was performed using IBM SPSS Statistics version 18.0 (SPSS Inc., Chicago, IL, USA). Associations between categorical variables were examined using the Pearson’s Chi-square test and associations between continuous variables using the Student’s T-test. P-values <0.05 were considered to be statistically significant.

Cell Culture

Origin and culture of the mouse cell lines Trp53Δ/Δ-3, Trp53Δ/Δ-4 and mILC-1 were described before [20]. ILC cell line IPH-926 was cultured as described [32]. Human breast cancer cell line MCF10a was obtained from ATCC (validated by STR profiling), and cultured in DMEM/F12 (Invitrogen, Breda, the Netherlands) supplemented with 10 mg/L insulin, 20 µg/L EGF, 100 µg/L cholera toxin, and 500 µg/L hydrocortisone (Sigma, Zwijndrecht, The Netherlands). All media contained 10% fetal calf serum, 100 IU/ml penicillin, and 100 µg/ml streptomycin. All cell lines were maintained at 37°C in a 5% CO2 humidified atmosphere.

thumbnail
Figure 2. E-cadherin and p120 membrane localization correlates with nuclear Kaiso expression.

IDC (left panels) and ILC (right panels) were stained for E-cadherin, p120, and Kaiso using immunohistochemistry. Note the association between membrane-localized E-cadherin and p120, and high nuclear Kaiso in IDC. In contrast, ILC is characterized by loss of E-cadherin, and expression of cytoplasmic p120, which correlates with absence of nuclear Kaiso. Size bars equal 50 µm.

https://doi.org/10.1371/journal.pone.0037864.g002

thumbnail
Table 1. Clinicopathological characteristics of 477 invasive breast cancer patients studied for the expression of Kaiso.

https://doi.org/10.1371/journal.pone.0037864.t001

thumbnail
Table 2. Correlation of cytoplasmic Kaiso with clinicopathological features in invasive breast cancer.

https://doi.org/10.1371/journal.pone.0037864.t002

thumbnail
Table 3. Correlation of nuclear Kaiso with clinicopathological features in invasive breast cancer.

https://doi.org/10.1371/journal.pone.0037864.t003

Immunofluorescence

Cells were cultured on coverslips and fixed in methanol for 10 minutes, permeabilized using 0.3% Triton-X100/PBS and subsequently blocked using 4% BSA (Roche, Woerden, The Netherlands). Cover slips were incubated with mouse anti Kaiso 1∶500 (clone 6F) in 4% BSA for 1 hour at room temperature. Subsequently, cells were incubated in 4% BSA with goat-anti-mouse Alexa 488 (1∶600; Molecular Probes, Breda, The Netherlands) for 1 hour. Next, cells were incubated with TRITC-conjugated mouse anti-p120 1∶300 (clone 98/pp120, BD Biosciences) overnight at 4°C. Cover slips were mounted using Vectashield mounting medium (Vector Laboratories, Burlingame, CA, USA). Samples were analyzed by confocal laser microscopy.

Luciferase Reporter Assay

Cells were cultured in 6-wells culture plates and grown to 40–50% confluency. Next, cells were transfected with either 600 ng of the Kaiso-specific reporter (pGL3-4XKBS), a mutated Kaiso reporter (pGL3-4XKBS CAmut) or empty vector (pGL3-Control) and co-transfected with 5 ng Renilla (pRL-CMV, Promega, Leiden, The Netherlands) for normalization of transfection efficiency, using Lipofectamine 2000 (Invitrogen) according to manufactures instructions. In addition, cells were transfected with 400 ng effector plasmid consisting either of pC2-p120 isoform 1a, pcDNA3.1-Kaiso or empty vector (pcDNA3.1). The transfection mixture was added to the cells and incubated for 2.5 hours. Followed by replacement of the transfection mixture by complete medium.

Two days post-transfection cells were washed twice with PBS, lysed by scraping in 200µl Passive Lysis Buffer (Promega) and subjected to a freeze-thaw cycle. Cellular debris was spun down at 5,000 g at 4°C for 5 minutes and supernatants were collected. Bioluminescence was measured in 50µl sample with Dual Luciferase Reporter assay system (Promega) on a Lumat LB9507 Luminometer (Berthold Technologies, Vilvoorde, Belgium) according manufactures instructions.

thumbnail
Table 4. Correlation of nuclear Kaiso with the molecular subtypes of breast cancer in invasive breast cancer.

https://doi.org/10.1371/journal.pone.0037864.t004

Results

Kaiso Expression in Normal Breast Epithelium and Invasive Breast Cancer

In normal breast tissue, localization of Kaiso was observed in the cytosol of both luminal and myoepithelial cells (Figure 1). We detected nuclear Kaiso expression mainly in the luminal epithelial cells, which was heterogeneous while the number of cells showing nuclear Kaiso varied between ductal structures (5–35% of the cells)(Figure 1).

Next, we set out to analyze Kaiso expression in invasive breast cancer. We used a study population comprised of 312 (65.4%) IDC, 130 (27.3%) ILC, and 35 (7.3%) invasive breast cancer cases with other histology (Table 1). First, we scored absence or presence of cytoplasmic expression of Kaiso, since this variable has recently been linked to poor prognosis in non-small cell lung cancer (NSCLC) [33]. Although cytoplasmic expression of Kaiso was significantly different between the histological sub-types of breast cancer (p = 0.006), it was not associated with other clinicopathological features (Table 2 and Figure 2). Given that Kaiso functions as a transcriptional repressor, we scored nuclear expression in our breast cancer cohort. Since using thresholds of 1%, 5% or 10% for scoring of nuclear accumulation resulted in identical outcome and statistical significance (data no shown), we used 5% nuclear localization as a positive cut-off percentage. IDC expressed nuclear Kaiso more often than ILC (p = 0.007; Table 3; Figure 2), while exclusive cytoplasmic expression of Kaiso was a common feature of ILC. In addition, no significant difference was found between classical and pleomorphic lobular cancers (p = 0.237) (data not shown). For the other clinicopathological features, we observed that high-grade tumors and cancers with a MAI ≥13, had significantly more nuclear Kaiso than low-grade tumors (p = 0.023 and p = 0.003, respectively), while no significant differences were found for lymph node status and tumor size (Table 3).

thumbnail
Table 5. Correlation between functional adherens junctions and nuclear Kaiso expression in invasive breast cancer.

https://doi.org/10.1371/journal.pone.0037864.t005

Nuclear Kaiso Expression and Molecular Subtypes of Breast Cancer

Since Kaiso is implicated in transcriptional repression of specific target genes, and our data indicated that nuclear Kaiso correlated with histology and grading in our invasive breast cancer cohort, we performed a cross-comparison between nuclear Kaiso expression and the molecular subtypes of breast cancer. Nuclear Kaiso was significantly enriched in the basal/triple negative and HER2-driven breast cancers than luminal-type breast cancers (p = 0.018; Table 4). While we did not find differences in nuclear Kaiso expression in the context of PR and HER2 (p = 0.104 and p = 0.246, respectively), an inverse correlation between nuclear Kaiso and ERα expression was detected (p = 0.001) (Table 4). Moreover, BRCA1-associated breast cancers showed a significantly higher number of tumors expressing nuclear-localized Kaiso than sporadic carcinomas (71,4% versus 29,3%, respectively; p<0.001, Table 4).

Localization of Kaiso, EGFR and the Adherens Junction in Breast Cancer

Expression of EGFR has been linked to prognosis in basal/triple-negative breast cancer [34], [35]. Because EGFR partly co-localizes with the AJ [36], and EGF stimulation can modulate AJ function through phosphorylation of Src, p120 and PKCδ [37], [38], we determined whether EGFR expression correlated with levels of membranous E-cadherin and nuclear Kaiso. Indeed, a strong association between EGFR and E-cadherin (p<0.001) was observed, which coincided with a higher prevalence of nuclear Kaiso expression in EGFR-expressing breast cancers (p = 0.019; Table 4).

Kaiso was identified as a p120-binding partner in a yeast two-hybrid screen, using p120 as bait [1]. Since then, several studies indicated that p120 controls relief of Kaiso-mediated transcriptional repression through binding and shuttling from and to the cytosol [9], [10]. Interestingly, this feature can be antagonized by E-cadherin expression, a key determinant in the differential diagnosis between IDC and ILC [39]. While approximately 90% of ILC cases show loss of E-cadherin expression, the majority of IDC cases have retained E-cadherin on the membrane [40][42]. Our data indicated that IDC and ILC show significant differences in cytoplasmic and nuclear Kaiso localization (Figure 2, Tables 3 and 4). The presence of AJs, i.e. membranous localization of E-cadherin and p120, strongly correlated with high nuclear Kaiso (p = 0.008 and p = 0.001, respectively; Table 5). Moreover, nuclear Kaiso inversely correlated with cytoplasmic p120 (p = 0.005), thus supporting the notion that loss of E-cadherin and subsequent translocation of p120 to the cytosol may control Kaiso localization.

To substantiate these findings, we analyzed expression of Kaiso in MCF10a, a breast cancer cell line that expressed membranous E-cadherin and p120. Furthermore, since our data indicated that nuclear Kaiso and ILC were inversely correlated, we also used immunofluorescence to examine Kaiso localization in a recently generated and characterized bona fide human ILC cell line; IPH-926 [32]. Although Kaiso expression was observed in E-cadherin-expressing as well as in E-cadherin-mutant cells, nuclear Kaiso was enriched in MCF10a cells, whereas IPH-926 virtually lacked nuclear Kaiso (Figure 3A). In addition, we employed cell lines derived from conditional mouse models in which E-cadherin and/or p53 were somatically inactivated [19]. In agreement with our findings in human cell lines, we could detect nuclear Kaiso in an E-cadherin expressing and p53-deficient mammary carcinoma cell line (Trp53Δ/Δ-3), whereas mouse ILC (mILC) cells mainly lacked nuclear Kaiso (Figure 3B). Evidence that p120 could direct nuclear localization of Kaiso was obtained by overexpressing p120 in Trp53Δ/Δ-4 cells, which resulted in high cytoplasmic p120 and a reduction in nuclear Kaiso (Figure 3C). To determine the effect of p120 overexpression on Kaiso-dependent transcriptional repression, we performed reporter assays using a Kaiso reporter system (containing 4 tandem-repeats of the consensus Kaiso Binding Sequence; 4XKBS reporter). In line with our expression data, we observed that Kaiso-dependent transcriptional repression was significantly higher in Trp53Δ/Δ-4 than in mILC-1 cells (p = 0.015; Figure 3D). Furthermore, transcriptional repression of the 4XKBS reporter was attenuated by exogenous Kaiso expression in mILC-1 cells (Figure 3D). Finally, we observed that overexpression of p120 in Trp53Δ/Δ-4 cells resulted in decreased Kaiso-dependent transcriptional repression (Figure 3C and 3D), consistent with a decrease in nuclear Kaiso expression. These results support our findings in primary breast cancer samples and indicate that p120 controls localization of Kaiso and subsequent de-repression of Kaiso-dependent transcription in breast cancer.

thumbnail
Figure 3. p120 and Kaiso localization in breast cancer cell lines.

Human (A) and mouse (B) E-cadherin-expressing (top panels) and E-cadherin-deficient (bottom panels) breast cancer cell lines were stained for p120 (left panels) and Kaiso (middle panels). Right panels depict the merge of Kaiso (green) and p120 (red). Note the nuclear accumulation in MCF10a and Trp53Δ/Δ-3 (arrows; also upper panels in C) versus cytoplasmic Kaiso expression in human and mouse ILC (arrowheads; IPH-926 and mILC-1). (C) Overexpression of p120 in Trp53Δ/Δ-4 cells resulted in decreased nuclear accumulation of Kaiso (arrowheads; bottom panels) compared to untransfected Trp53Δ/Δ-4, which shows predominantly nuclear Kaiso (arrows; upper panels). Size bars equal 20 µm. (D) Kaiso-dependent reporter assay using the 4XKBS reporter in mILC-1 and Trp53Δ/Δ-4 cells. Upon overexpression of p120 in Trp53Δ/Δ-4 cells, Kaiso-dependent gene repression is attenuated, whereas exogenous expression of Kaiso in mILC-1 increased gene repression.

https://doi.org/10.1371/journal.pone.0037864.g003

Discussion

In addition to the established role of BTB-POZ-ZF transcription factors in vertebrate development, increasing evidence emerges that these factors can function as oncogenes or tumor suppressors [43]. For instance, the BTB/POZ promyelocytic leukemia zinc finger (PLZF) has been identified as a translocation partner of the retinoic receptor alpha (RARα). In this setting, PLZF confers oncogenic potential through fusion to the hormone-binding domain of RARα, subsequent binding to its target sites and local recruitment of histone deacetylases [44]. Another well-studied BTB-POZ oncogene is BCL6, a protein that exerts its pro-tumorigenic functions by repression of target genes necessary for terminal B cell differentiation [45], [46]. In contrast, HIC1 is a candidate tumor suppressor that is often found mutated or hypermethylated in human cancer [47]. However, unlike PLZF, BCL-6 and HIC1, it remains unclear whether Kaiso mislocalization or absence could drive malignancy.

Kaiso could function as an oncogene or as a tumor suppressor as it has been implicated in both transcriptional activation and repression [4], [8], [13]. In colon cancer, Kaiso may regulate methylation-dependent inhibition of tumor suppressors such as CDKN2A by binding to its methylated promoter. As a consequence, tumor cells are resistant to cell cycle arrest and chemotherapy-mediated cell death [48]. Interestingly, genetic Kaiso ablation results in a delay in intestinal tumorigenesis in the context of APCMIN/+ mice [16], which suggests that Kaiso may indeed contribute to intestinal tumor progression through silencing of tumor suppressors. Conversely, Kaiso has been strongly implicated in regulation of Wnt signaling-related target genes [4], [11], [12], [49], [50]. Given its bi-modal nature of β-catenin-dependent regulation of Wnt signaling [51] and the overlap between TCF/LEF regulated genes and Kaiso targets, the effects of Kaiso on tumor development may be highly dependent on cell type and their dependency on (canonical) Wnt signals. In lung cancer, cytoplasmic Kaiso was correlated with poor prognosis [33]. Here it was proposed that the invasive phenotype of NSCLC might be regulated by nuclear export of Kaiso, which was mediated by phosphorylation of p120 isoform 3 [52]. Lung and other epithelial tissue differ substantially from breast with respect to cadherin expression and p120 function. For instance, condition p120 knockout in the skin, gastro-intestinal tract or oral cavity is tolerated and induces hyperplasia or tumor formation [53][56]. In contrast, p120 knock-out in the mammary gland is not tolerated and leads to apoptosis and subsequent cell clearance (our unpublished results), indicating that p120 family members may play tissue-specific redundant roles, as has been suggested for δ-catenin in non-small-cell lung cancer [57].

In this study we have performed to the best of our knowledge, the first comprehensive analysis of Kaiso expression in breast cancer, using a tissue micro array (TMA)-based collection of 477 invasive breast cancer cases. Previous studies had already indicated that localization of Kaiso may be highly variable depending on tumor type and environmental context [14]. Our data indicate that nuclear Kaiso expression correlates with the pathological and phenotypical traits of specific breast cancer sub-types that are linked to poor prognosis, i.e. high-grade, and basal/triple-negative breast cancer. These tumors were also associated with high EGFR expression, which is associated with worse prognosis for basal/triple-negative breast cancers [34], [35]. Our observation that BRCA1-associated hereditary breast cancers often showed high nuclear Kaiso, is in line with the finding that nuclear Kaiso is in general associated with high grade, basal-like and EGFR positive breast cancers. Since our data does not indicate differential E-cadherin expression and localization between sporadic and BRCA1-related invasive breast cancer, this cannot explain the increase in nuclear Kaiso localization. Future research will have to determine if and how other (p120-unrelated) events such as promoter methylation of specific genes may recruit Kaiso to the nucleus and initiate subsequent epigenetic silencing in BRCA1-related invasive breast cancer. We have furthermore shown that nuclear Kaiso correlated with the presence of membrane-localized E-cadherin and p120, a finding that is in line with the reported regulation of Kaiso by p120 [58]. In this scenario, p120 relieves transcriptional repression by Kaiso and as such may control shuttling of the p120/Kaiso complex to the cytosol [9]. Because most IDC retain a membrane-localized E-cadherin/p120 complex, our data confirmed this concept by showing that nuclear Kaiso correlated with tumors expressing E-cadherin. Also, since the basal-like and ERα negative high-grade tumors mainly reside in the E-cadherin-expressing IDC cohort, it supports the notion that p120 may regulate Kaiso distribution in breast cancer. The mechanism of this needs to be further elucidated.

ILC is characterized by loss of the AJ complex through early mutational inactivation of E-cadherin and subsequent translocation of p120 to the cytosol. If p120 were a major factor controlling Kaiso distribution, one would expect that the absence of nuclear Kaiso associated with ILC. Our data indeed conforms to this hypothesis by showing that exclusive cytoplasmic Kaiso expression is strongly correlated with the lobular phenotype. Together, these findings suggest that genes may be differentially regulated in IDC versus ILC as a result of differential Kaiso localization. This notion may therefore partly explain the differences in expression profiles that have been reported when comparing IDC and ILC [59], [60].

Recent data have indicated that phosphorylation of p120 can increase its binding to Kaiso and induce inhibition of canonical Wnt signaling [11]. It is well established that ILC expresses cytoplasmic p120 and does not activate canonical Wnt signals [20], [21], [61]. Although it is unclear if this mechanism controls expression of Kaiso targets in ILC, it clearly emphasizes the possible ramifications of Kaiso and its regulation by p120 in breast cancer. Moreover, we have recently shown that cytoplasmic translocation of p120 controls ILC tumor growth and metastasis through Mrip-dependent regulation of Rock1 signaling, while IDC does not appear to be contingent on these signals for anchorage-independence [20]. We envisage that differential cadherin-catenin localization in IDC and ILC and the signals that emanate from p120 may not only explain the lobular phenotype, but probably also control regulation of transcriptional regulation and cellular biochemistry. Although Kaiso’s target genes in breast cancer are unknown, our findings suggest that Kaiso may function as an oncogene in IDC through inhibition of tumor suppressor gene expression whereas in ILC, Kaiso might harbor tumor suppressor functions by p120-mediated relieve of transcriptional repression of oncogenic target genes. As such, it may have significant impact on the development of personalized cancer care since it suggests that the main breast cancer types may depend on diametrical mechanisms for tumor progression.

Author Contributions

Conceived and designed the experiments: JFV RAHvdV PWBD. Performed the experiments: JFV RAHvdV CE. Analyzed the data: JD MC UL PB PvdG PJvD EvdW. Contributed reagents/materials/analysis tools: EvdW PB MC UL PJvD. Wrote the paper: JFV RAHvdV PWBD.

References

  1. 1. Daniel JM, Reynolds AB (1999) The catenin p120(ctn) interacts with Kaiso, a novel BTB/POZ domain zinc finger transcription factor. Mol Cell Biol 19: 3614–3623.
  2. 2. Daniel JM (2007) Dancing in and out of the nucleus: p120(ctn) and the transcription factor Kaiso. Biochim Biophys Acta 1773: 59–68.
  3. 3. van Roy FM, McCrea PD (2005) A role for Kaiso-p120ctn complexes in cancer? Nat Rev Cancer 5: 956–964.
  4. 4. Daniel JM, Spring CM, Crawford HC, Reynolds AB, Baig A (2002) The p120(ctn)-binding partner Kaiso is a bi-modal DNA-binding protein that recognizes both a sequence-specific consensus and methylated CpG dinucleotides. Nucleic Acids Res 30: 2911–2919.
  5. 5. Prokhortchouk A, Hendrich B, Jorgensen H, Ruzov A, Wilm M, et al. (2001) The p120 catenin partner Kaiso is a DNA methylation-dependent transcriptional repressor. Genes Dev 15: 1613–1618.
  6. 6. Ruzov A, Hackett JA, Prokhortchouk A, Reddington JP, Madej MJ, et al. (2009) The interaction of xKaiso with xTcf3: a revised model for integration of epigenetic and Wnt signalling pathways. Development 136: 723–727.
  7. 7. Ruzov A, Savitskaya E, Hackett JA, Reddington JP, Prokhortchouk A, et al. (2009) The non-methylated DNA-binding function of Kaiso is not required in early Xenopus laevis development. Development 136: 729–738.
  8. 8. Rodova M, Kelly KF, VanSaun M, Daniel JM, Werle MJ (2004) Regulation of the rapsyn promoter by kaiso and delta-catenin. Mol Cell Biol 24: 7188–7196.
  9. 9. Kelly KF, Spring CM, Otchere AA, Daniel JM (2004) NLS-dependent nuclear localization of p120ctn is necessary to relieve Kaiso-mediated transcriptional repression. J Cell Sci 117: 2675–2686.
  10. 10. Kim SW, Park JI, Spring CM, Sater AK, Ji H, et al. (2004) Non-canonical Wnt signals are modulated by the Kaiso transcriptional repressor and p120-catenin. Nat Cell Biol 6: 1212–1220.
  11. 11. Del Valle-Perez B, Casagolda D, Lugilde E, Valls G, Codina M, et al. (2011) Wnt controls the transcriptional activity of Kaiso through CK1epsilon-dependent phosphorylation of p120-catenin. J Cell Sci 124: 2298–2309.
  12. 12. Park JI, Kim SW, Lyons JP, Ji H, Nguyen TT, et al. (2005) Kaiso/p120-catenin and TCF/beta-catenin complexes coordinately regulate canonical Wnt gene targets. Dev Cell 8: 843–854.
  13. 13. Ruzov A, Dunican DS, Prokhortchouk A, Pennings S, Stancheva I, et al. (2004) Kaiso is a genome-wide repressor of transcription that is essential for amphibian development. Development 131: 6185–6194.
  14. 14. Soubry A, van Hengel J, Parthoens E, Colpaert C, Van Marck E, et al. (2005) Expression and nuclear location of the transcriptional repressor Kaiso is regulated by the tumor microenvironment. Cancer Res 65: 2224–2233.
  15. 15. Donaldson NS, Nordgaard CL, Pierre CC, Kelly KF, Robinson SC, et al. (2010) Kaiso regulates Znf131-mediated transcriptional activation. Exp Cell Res 316: 1692–1705.
  16. 16. Prokhortchouk A, Sansom O, Selfridge J, Caballero IM, Salozhin S, et al. (2006) Kaiso-deficient mice show resistance to intestinal cancer. Mol Cell Biol 26: 199–208.
  17. 17. Berx G, van Roy F (2009) Involvement of members of the cadherin superfamily in cancer. Cold Spring Harb Perspect Biol 1: a003129.
  18. 18. Derksen PW, Braumuller TM, van der Burg E, Hornsveld M, Mesman E, et al. (2011) Mammary-specific inactivation of E-cadherin and p53 impairs functional gland development and leads to pleomorphic invasive lobular carcinoma in mice. Dis Model Mech 4: 347–358.
  19. 19. Derksen PW, Liu X, Saridin F, van der Gulden H, Zevenhoven J, et al. (2006) Somatic inactivation of E-cadherin and p53 in mice leads to metastatic lobular mammary carcinoma through induction of anoikis resistance and angiogenesis. Cancer Cell 10: 437–449.
  20. 20. Schackmann RC, van Amersfoort M, Haarhuis JH, Vlug EJ, Halim VA, et al. (2011) Cytosolic p120-catenin regulates growth of metastatic lobular carcinoma through Rock1-mediated anoikis resistance. J Clin Invest 121: 3176–3188.
  21. 21. van de Wetering M, Barker N, Harkes IC, van der Heyden M, Dijk NJ, et al. (2001) Mutant E-cadherin breast cancer cells do not display constitutive Wnt signaling. Cancer Res 61: 278–284.
  22. 22. Dabbs DJ, Bhargava R, Chivukula M (2007) Lobular versus ductal breast neoplasms: the diagnostic utility of p120 catenin. Am J Surg Pathol 31: 427–437.
  23. 23. Sarrio D, Perez-Mies B, Hardisson D, Moreno-Bueno G, Suarez A, et al. (2004) Cytoplasmic localization of p120ctn and E-cadherin loss characterize lobular breast carcinoma from preinvasive to metastatic lesions. Oncogene 23: 3272–3283.
  24. 24. Shibata T, Kokubu A, Sekine S, Kanai Y, Hirohashi S (2004) Cytoplasmic p120ctn regulates the invasive phenotypes of E-cadherin-deficient breast cancer. Am J Pathol 164: 2269–2278.
  25. 25. van der Groep P, Bouter A, van der Zanden R, Menko FH, Buerger H, et al. (2004) Re: Germline BRCA1 mutations and a basal epithelial phenotype in breast cancer. J Natl Cancer Inst 96: 712–713; author reply 714:
  26. 26. van der Groep P, Bouter A, van der Zanden R, Siccama I, Menko FH, et al. (2006) Distinction between hereditary and sporadic breast cancer on the basis of clinicopathological data. J Clin Pathol 59: 611–617.
  27. 27. Packeisen J, Korsching E, Herbst H, Boecker W, Buerger H (2003) Demystified…tissue microarray technology. Mol Pathol 56: 198–204.
  28. 28. van Diest PJ (2002) No consent should be needed for using leftover body material for scientific purposes. For. BMJ 325: 648–651.
  29. 29. Daniel JM, Ireton RC, Reynolds AB (2001) Monoclonal antibodies to Kaiso: a novel transcription factor and p120ctn-binding protein. Hybridoma 20: 159–166.
  30. 30. Kornegoor R, Verschuur-Maes AH, Buerger H, Hogenes MC, de Bruin PC, et al. (2012) Molecular subtyping of male breast cancer by immunohistochemistry. Mod Pathol 25: 398–404.
  31. 31. Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, et al. (2001) Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 98: 10869–10874.
  32. 32. Christgen M, Bruchhardt H, Hadamitzky C, Rudolph C, Steinemann D, et al. (2009) Comprehensive genetic and functional characterization of IPH-926: a novel CDH1-null tumour cell line from human lobular breast cancer. J Pathol 217: 620–632.
  33. 33. Dai SD, Wang Y, Miao Y, Zhao Y, Zhang Y, et al. (2009) Cytoplasmic Kaiso is associated with poor prognosis in non-small cell lung cancer. BMC Cancer 9: 178.
  34. 34. Siziopikou KP, Cobleigh M (2007) The basal subtype of breast carcinomas may represent the group of breast tumors that could benefit from EGFR-targeted therapies. Breast 16: 104–107.
  35. 35. Livasy CA, Karaca G, Nanda R, Tretiakova MS, Olopade OI, et al. (2006) Phenotypic evaluation of the basal-like subtype of invasive breast carcinoma. Mod Pathol 19: 264–271.
  36. 36. Takahashi K, Suzuki K, Tsukatani Y (1997) Induction of tyrosine phosphorylation and association of beta-catenin with EGF receptor upon tryptic digestion of quiescent cells at confluence. Oncogene 15: 71–78.
  37. 37. Mariner DJ, Davis MA, Reynolds AB (2004) EGFR signaling to p120-catenin through phosphorylation at Y228. J Cell Sci 117: 1339–1350.
  38. 38. Singh R, Lei P, Andreadis ST (2009) PKC-delta binds to E-cadherin and mediates EGF-induced cell scattering. Exp Cell Res 315: 2899–2913.
  39. 39. van Hengel J, Vanhoenacker P, Staes K, van Roy F (1999) Nuclear localization of the p120(ctn) Armadillo-like catenin is counteracted by a nuclear export signal and by E-cadherin expression. Proc Natl Acad Sci U S A 96: 7980–7985.
  40. 40. Schonborn I, Zschiesche W, Behrens J, Herrenknecht K, Birchmeier W (1997) Expression of E-cadherin/catenin complexes in breast cancer. Int J Oncol 11: 1327–1334.
  41. 41. Qureshi HS, Linden MD, Divine G, Raju UB (2006) E-cadherin status in breast cancer correlates with histologic type but does not correlate with established prognostic parameters. Am J Clin Pathol 125: 377–385.
  42. 42. Gonzalez-Angulo AM, Sahin A, Krishnamurthy S, Yang Y, Kau SW, et al. (2006) Biologic markers in axillary node-negative breast cancer: differential expression in invasive ductal carcinoma versus invasive lobular carcinoma. Clin Breast Cancer 7: 396–400.
  43. 43. Kelly KF, Daniel JM (2006) POZ for effect–POZ-ZF transcription factors in cancer and development. Trends Cell Biol 16: 578–587.
  44. 44. Costoya JA, Pandolfi PP (2001) The role of promyelocytic leukemia zinc finger and promyelocytic leukemia in leukemogenesis and development. Curr Opin Hematol 8: 212–217.
  45. 45. Reljic R, Wagner SD, Peakman LJ, Fearon DT (2000) Suppression of signal transducer and activator of transcription 3-dependent B lymphocyte terminal differentiation by BCL-6. J Exp Med 192: 1841–1848.
  46. 46. Wagner SD, Ahearne M, Ferrigno PK (2011) The role of BCL6 in lymphomas and routes to therapy. Br J Haematol 152: 3–12.
  47. 47. Chen WY, Zeng X, Carter MG, Morrell CN, Chiu Yen RW, et al. (2003) Heterozygous disruption of Hic1 predisposes mice to a gender-dependent spectrum of malignant tumors. Nat Genet 33: 197–202.
  48. 48. Lopes EC, Valls E, Figueroa ME, Mazur A, Meng FG, et al. (2008) Kaiso contributes to DNA methylation-dependent silencing of tumor suppressor genes in colon cancer cell lines. Cancer Res 68: 7258–7263.
  49. 49. Park JI, Ji H, Jun S, Gu D, Hikasa H, et al. (2006) Frodo links Dishevelled to the p120-catenin/Kaiso pathway: distinct catenin subfamilies promote Wnt signals. Dev Cell 11: 683–695.
  50. 50. Peek RM, Ogden SR, Wroblewski LE, Weydig C, Romero-Gallo J, et al. (2008) p120 and Kaiso Regulate Helicobacter pylori-induced Expression of Matrix Metalloproteinase-7. Molecular Biology of the Cell 19: 4110–4121.
  51. 51. Iioka H, Doerner SK, Tamai K (2009) Kaiso is a bimodal modulator for Wnt/beta-catenin signaling. FEBS Lett 583: 627–632.
  52. 52. Zhang PX, Wang Y, Liu Y, Jiang GY, Li QC, et al. (2011) p120-catenin isoform 3 regulates subcellular localization of Kaiso and promotes invasion in lung cancer cells via a phosphorylation-dependent mechanism. Int J Oncol 38: 1625–1635.
  53. 53. Perez-Moreno M, Davis MA, Wong E, Pasolli HA, Reynolds AB, et al. (2006) p120-catenin mediates inflammatory responses in the skin. Cell 124: 631–644.
  54. 54. Stairs DB, Bayne LJ, Rhoades B, Vega ME, Waldron TJ, et al. (2011) Deletion of p120-catenin results in a tumor microenvironment with inflammation and cancer that establishes it as a tumor suppressor gene. Cancer Cell 19: 470–483.
  55. 55. Smalley-Freed WG, Efimov A, Short SP, Jia P, Zhao Z, et al. (2011) Adenoma formation following limited ablation of p120-catenin in the mouse intestine. PLoS One 6: e19880.
  56. 56. Davis MA, Reynolds AB (2006) Blocked acinar development, E-cadherin reduction, and intraepithelial neoplasia upon ablation of p120-catenin in the mouse salivary gland. Dev Cell 10: 21–31.
  57. 57. Dai SD, Wang Y, Zhang JY, Zhang D, Zhang PX, et al. (2011) Upregulation of delta-catenin is associated with poor prognosis and enhances transcriptional activity through Kaiso in non-small-cell lung cancer. Cancer Sci 102: 95–103.
  58. 58. Kelly KF, Otchere AA, Graham M, Daniel JM (2004) Nuclear import of the BTB/POZ transcriptional regulator Kaiso. J Cell Sci 117: 6143–6152.
  59. 59. Korkola JE, DeVries S, Fridlyand J, Hwang ES, Estep AL, et al. (2003) Differentiation of lobular versus ductal breast carcinomas by expression microarray analysis. Cancer Res 63: 7167–7175.
  60. 60. Zhao H, Langerod A, Ji Y, Nowels KW, Nesland JM, et al. (2004) Different gene expression patterns in invasive lobular and ductal carcinomas of the breast. Mol Biol Cell 15: 2523–2536.
  61. 61. de Leeuw WJ, Berx G, Vos CB, Peterse JL, Van de Vijver MJ, et al. (1997) Simultaneous loss of E-cadherin and catenins in invasive lobular breast cancer and lobular carcinoma in situ. J Pathol 183: 404–411.