throbber
Cadherin 6 Is a New RUNX2 Target in TGF-β Signalling
`Pathway
`Valentina Sancisi1, Greta Gandolfi1, Moira Ragazzi2, Davide Nicoli1, Ione Tamagnini2, Simonetta Piana2,
`Alessia Ciarrocchi1*
`
`1 Laboratory of Molecular Biology, Department of Oncology and Advanced Technologies, Azienda Ospedaliera Arcispedale S. Maria Nuova-IRCCS, Reggio
`Emilia, Italy, 2 Pathology Unit, Department of Oncology and Advanced Technologies, Azienda Ospedaliera Arcispedale S. Maria Nuova-IRCCS, Reggio Emilia,
`Italy
`
`Abstract
`
`Modifications in adhesion molecules profile may change the way tumor cells interact with the surrounding
`microenvironment. The Cadherin family is a large group of transmembrane proteins that dictate the specificity of the
`cellular interactions. The Cadherin switch that takes place during epithelial-mesenchymal transition (EMT) contributes
`to loosening the rigid organization of epithelial tissues and to enhancing motility and invasiveness of tumor cells.
`Recently, we found Cadherin-6 (CDH6, also known as K-CAD) highly expressed in thyroid tumor cells that display
`mesenchymal features and aggressive phenotype, following the overexpression of the transcriptional regulator Id1. In
`this work, we explored the possibility that CDH6 is part of the EMT program in thyroid tumors. We demonstrate that
`CDH6 is a new transforming growth factor-β (TGF-β) target and that its expression is modulated similarly to other
`EMT mesenchymal markers, both in vitro and in thyroid tumor patients. We show for the first time that CDH6 is
`expressed in human thyroid carcinomas and that its expression is enhanced at the invasive front of the tumor.
`Finally, we show that CDH6 is under the control of the transcription factor RUNX2, which we previously described as
`a crucial mediator of the Id1 pro-invasive function in thyroid tumor cells. Overall, these observations provide novel
`information on the mechanism of the EMT program in tumor progression and indicate CDH6 as a potential regulator
`of invasiveness in thyroid tumors.
`
`Citation: Sancisi V, Gandolfi G, Ragazzi M, Nicoli D, Tamagnini I, et al. (2013) Cadherin 6 Is a New RUNX2 Target in TGF-β Signalling Pathway. PLoS
`ONE 8(9): e75489. doi:10.1371/journal.pone.0075489
`Editor: Jun Li, Sun Yat-sen University Medical School, China
`Received April 12, 2013; Accepted August 15, 2013; Published September 12, 2013
`Copyright: © 2013 Sancisi et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
`unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
`Funding: This work was supported by grants from the Italian Association for Cancer Research (AIRC, MFAG:10745)(http://www.airc.it/) and the Guido
`Berlucchi Foundation (http://www.fondazioneberlucchi.com/). The funders had no role in study design, data collection and analysis, decision to publish, or
`preparation of the manuscript.
`Competing interests: The authors have declared that no competing interests exist.
`* E-mail: Alessia.Ciarrocchi@asmn.re.it
`
`Introduction
`
`The transdifferentiation of epithelial tumor cells towards a
`mesenchymal condition is a complex process that allows tumor
`cells to leave their original site and to invade adjacent tissues.
`During this transition (also known as epithelial-mesenchymal
`transition - EMT), the epithelial cells shed their differentiated
`characteristics, including cell-cell adhesion, polarity, and lack of
`motility, and acquire instead mesenchymal features, including
`motility,
`invasiveness, and resistance
`to apoptosis. The
`importance of the EMT program in mediating epithelial cancer
`progression is supported by a massive amount of evidence that
`has been published on this topic over the last 15 years [1-5].
`While the relevance of this process to the biology of tumors is
`well established and fully accepted, the complexity of the
`molecular events and regulatory pathways at the basis of EMT
`is far from being fully understood. Furthermore, some of the
`
`molecular players involved in this process remain unknown.
`The first functional consequence of EMT program activation is
`the alteration of the epithelial tumor cell interactions with the
`surrounding microenvironment. The epithelial adhesion
`molecules, in particular the E-Cadherin, are displaced by the
`multiprotein complexes at the adherent junctions and are
`substituted by mesenchymal Cadherins (such as N-Cadherin).
`This alteration signals within
`the cells
`triggering
`the
`complicated cytoskeleton rearrangement that it is necessary to
`support cell motility [6]. The E-Cadherin and N-Cadherin are
`the prototypes and by far the most studied members of the
`large Cadherin family, which includes over 50 proteins in
`vertebrates and non-vertebrate organisms. Some evidence
`suggests that different Cadherins play non-redundant roles in
`cells and it is commonly believed that such large variability
`originates from the need of complex organisms to specifically
`differentiate intercellular interactions [7]. Despite this, the
`
`PLOS ONE | www.plosone.org
`
`1
`
`September 2013 | Volume 8 | Issue 9 | e75489
`
`1 of 16
`
`OnCusp
`Ex. 1027
`
`

`

`potential role of Cadherins other than the E- and N-Cadherin in
`cancer development and progression has being
`rarely
`investigated. Recently, we have shown that the aggressive
`phenotype induced by the transcription regulator Id1 in thyroid
`tumor cells is accompanied by acquisition of mesenchymal
`features and by deregulation of over 400 genes, most of which
`are known to be deregulated or partake in the EMT program.
`Among the Id1 target genes, the Cadherin-6 (CDH6, also
`known as K-Cadherin) was strongly induced by Id1 in thyroid
`tumor cells [8]. CDH6 is a class II Cadherin, which is expressed
`mainly in kidney and central nervous system [9-11]. CDH6 is
`highly expressed in renal tissue during embryogenesis in which
`it drives the mesenchymal-epithelial differentiation that is
`necessary for kidney morphogenesis [9,12,13]. In spite of its
`role
`in
`promoting
`the
`epithelial
`phenotype
`during
`embryogenesis, CDH6 has been described as strongly
`expressed in ovarian cancer and renal carcinoma [14,15]. In
`the latter, CDH6 expression has been shown to strongly
`correlate with aggressive tumor behavior and poorer patient
`outcome [16,17]. To date, nothing is known regarding the
`involvement of CDH6 in the EMT program during epithelial
`tumor development and progression.
`In
`this work, we
`investigated the ability of normal and tumor thyroid cells to
`activate the EMT program in response to transforming growth
`factor-β (TGF-β) and we explored the possibility that CDH6 is a
`TGF-β target during EMT in thyroid tumors. Intriguingly, we
`found that thyroid tumor cells constitutively display markers of
`an active EMT process as compared to normal thyrocytes, both
`in vitro and in human patients. We also showed that CDH6 is
`strongly induced by TGF-β treatment both in normal and tumor
`thyroid cells, and that its expression accompanies invasiveness
`in human thyroid tumor patients.
`
`Materials and Methods
`
`Cell cultures and treatments
`B-CPAP, TPC1, and WRO human cell lines were obtained
`from Dr. Massimo Santoro, University of Naples (Naples, Italy)
`[18,19,20,21]. Nthy.ori 3.1 and FTC133 cell
`lines were
`purchased from Sigma-Aldrich (Milan, Italy). All cell lines were
`grown at 37°C/5% CO2. B-CPAP, TPC1, and WRO were grown
`in DMEM supplemented with 10% fetal bovine serum. Nthy.ori
`3.1 were grown in RPMI supplemented with 10% fetal bovine
`serum. FTC133 were grown in DMEM:Ham’s F12 (1:1)
`supplemented with 10% fetal bovine serum. Id1- and RUNX2-
`overexpressing B-CPAP clones and control clones were grown
`in presence of 400 µg/ml geneticin (Life Technologies, Monza,
`Italy).
`Cell lines were starved with culture medium containing 1%
`fetal bovine serum 16-18 hours before TGF-β treatment. TGF-β
`(Peprotech, Rocky Hill, NJ) was added in starvation medium at
`concentrations of 5 ng/ml and 100 ng/ml for the indicated
`periods of time.
`Italy) and
`(Sigma-Aldrich, Milan,
`For Actinomycin D
`Cycloheximide (Sigma-Aldrich, Milan, Italy) treatments, cells
`were starved with 1% fetal bovine serum, were treated after
`16-18 hours with Actinomycin D 5 mg/ml or cycloheximide 50
`
`CDH6 in TGF-β Mediated EMT Program
`
`mg/ml for 4 hours, then TGF-β was added at 100 ng/ml for 24
`hours.
`For TGF-β inhibitor experiments, BCPAP and TPC1 cell lines
`were starved over night and treated with 10mM of SB-431542
`(Abcam, Cambridge, UK) for 24h or 48h. For cells treated for
`48h, fresh inhibitor was added after 24h by replacing the
`medium.
`
`RNA extraction from formalin-fixed paraffin-embedded
`tissues
`Total RNA was collected from 5 slices, 5 µm thick, of
`formalin-fixed and paraffin-embedded PTCs using the High
`Pure RNA paraffin kit (Roche, Milan, Italy). All samples
`(normal, primary
`tumor, and metastasis) were manually
`dissected under microscopic guidance by two pathologists (MR
`and SP). In order to minimize the biases that a different tissue
`processing may introduce in RNA quality, we performed gene
`expression analysis comparing normal tissue, primary tumor,
`and metastasis from the same patient. Primary tumor and
`normal tissues were collected from the same slide. Quantity
`and purity of the total RNA were checked using the NanoDrop
`2000 spectrophotometer (Thermo Scientific, Waltham, MA).
`Primers for qRT-PCR were designed in order to obtain
`amplicons less than 100 bp in length.
`
`RNA extraction and quantitative real time-PCR (qRT-
`PCR)
`Total RNA purification from cells was performed with
`RNAeasy Mini kit (Qiagen, Milan, Italy). 500 ng of total RNA
`was retrotranscribed using the iScript cDNA kit (Biorad,
`Segrate, Italy). qRT-PCR was conducted using Sso Fast
`EvaGreen Super Mix (BioRad, Segrate, Italy) in the CFX96
`Real Time PCR Detection System (BioRad, Segrate, Italy).
`Relative expression of target genes was calculated using the
`ΔΔCt method by normalizing to the geometric mean of three
`reference genes expression: Glyceraldehyde 3-phosphate
`dehydrogenase (GAPDH), Cyclophilin A (CYPA) and Beta-D-
`Glucuronidase (GUSB) unless otherwise specified. Sequences
`of primers are listed in Table S1.
`
`Western blot
`For Western blot analysis, cells were lysed in RIPA buffer.
`Equal amounts of protein extracts were analyzed by SDS-
`PAGE using
`the Bio-Rad
`(Segrate,
`Italy) Mini-Protean
`apparatus. Staining was performed with the ECL Western blot
`detection reagent (GE Healthcare, Milan,
`Italy). Primary
`antibodies were mouse anti-E-CAD
`(BD Biosciences,
`Buccinasco,
`Italy), mouse anti-N-CAD
`(BD Biosciences,
`Buccinasco,
`Italy), rabbit anti-FN 1 (H-300, Santa Cruz
`Biotechnology, Santa Cruz, CA, USA), mouse anti-pERK (E-4,
`Santa Cruz Biotechnology, Santa Cruz, CA, USA), rabbit anti-
`pAKT (Santa Cruz Biotechnology, Santa Cruz, CA, USA), and
`mouse anti-Actin (AC-15, Sigma-Aldrich, Milan, Italy). For
`CDH6 we tried several antibodies from several dealers (Abcam
`ab64917 and ab71434, Santa Cruz sc-59974, Epitomics
`T0233). None of these antibodies gave reliable results in either
`western blot or immunohistochemistry. Secondary antibodies
`
`PLOS ONE | www.plosone.org
`
`2
`
`September 2013 | Volume 8 | Issue 9 | e75489
`
`2 of 16
`
`OnCusp
`Ex. 1027
`
`

`

`CDH6 in TGF-β Mediated EMT Program
`
`were HRP-conjugated anti-rabbit (GE Healthcare, Milan, Italy)
`and anti-mouse (GE Healthcare, Milan, Italy).
`
`Additional materials and methods are provided as supporting
`information.
`
`Patient samples and immunohistochemistry
`All PTC samples (N=15) and matched LNMs (N=7) were
`retrieved from the archive of the Pathology Unit of Arcispedale
`S. Maria Nuova. Description of the clinicopathological features
`of the analyzed PTC patients is provided in Table S2. Tissue
`specimens were fixed in 10% formalin, paraffin embedded, and
`cut in 4 µm thick sections. For the immunohistochemistry
`analysis, we used the mouse monoclonal anti-CDH6 antibody
`(HPA007047) from Sigma-Aldrich (Milan, Italy), which resulted
`in a strong membrane staining. As control of the specificity of
`the staining, we tested the antibody on renal carcinomas. No
`specific staining was observed in 3 mammary carcinoma
`samples. As indicated in the datasheet, this antibody does not
`work in western blot. Images were captured using a Nikon
`Eclipse E80 microscope (Nikon Instruments, Calenzano, Italy).
`
`Ethics statement
`This project was approved by the “Comitato etico provinciale
`di Reggio Emilia” (local ethics committee of Reggio Emilia).
`Written informed consent was obtained by all the patients
`involved in this project. The local ethics committee approved
`the entire procedure of informed consent collection and
`patients data managing. This study has been conducted
`according to the Declaration of Helsinki.
`
`Immunofluorescence
`Cells were grown in 4 well Lab-Tek Chamber slides
`(NunThermo Scientific, Waltham, MA, USA). After appropriate
`treatments, cells were fixed in 4% PFA in PBS 1X for 15
`minutes at room temperature, permeabilized with 0.1% Triton
`in PBS 1X for 2 minutes, blocked with 20% FBS and 2% BSA
`in PBS 1X for 1 hour, then incubated with a mouse anti-
`Smad2/3 antibody (C-8, Santa Cruz Biotechnology, Santa
`Cruz, CA, USA) in a humidified chamber for 1 hour. Antibody
`binding was revealed with a secondary anti-mouse Alexa 488
`conjugated antibody (Life Technologies, Monza, Italy). Cell
`nuclei were stained with DAPI (Life Technologies, Monza,
`Italy). Slides were mounted using the SlowFade mounting
`medium (Life Technologies, Monza, Italy) and observed using
`an Axiophot fluorescent microscope (Zeiss, Arese, Italy).
`
`Small interfering RNA (siRNA) transfection
`Stealth RNA interference oligos against RUNX2 and control
`oligos were purchased from Life Technologies (Monza, Italy),
`and 30 nM of either anti-RUNX2 or control siRNA was
`transfected using the RNAiMax Lipofectamine reagent (Life
`Technologies, Monza, Italy) using the reverse transfection
`protocol.
`
`Statistical analysis
`Statistical analysis was performed using GraphPad Prism
`Software (GraphPad, San Diego, California, USA). When
`statistical analyses were performed, the specific tests used are
`indicated in the figure legend.
`
`Results
`
`CDH6 is a new TGF-β target gene
`In order to investigate the effect of the TGF-β in thyroid
`tumor progression and the possibility that CDH6 is one of the
`TGF-β targets, five different thyroid-derived cell lines were
`selected and used as a model. The origin and specific features
`of the cell lines used are summarized in Table 1. Two different
`doses of TGF-β (5 ng/ml and 100 ng/ml) were given to cells for
`24h, after which changes
`in
`the expression
`levels of
`established EMT markers and CDH6 were measured by means
`of qRT-PCR (Figure 1). Two epithelial markers - E-CAD and
`Cadherin-16 (CDH16) - and four mesenchymal markers - N-
`CAD, Tenascin C (TNC), Vimentin (VIM) and Fibronectin 1
`(FN1) - were analyzed to monitor the activation of the EMT
`program [8,22-24]. All cell lines responded to TGF-β to some
`extent (Figure 1 A-E). However, the strength of the response
`was different between the Nthy.ori 3.1 thyrocytes and the tumor
`cell lines. In the Nthy. ori3.1 cells, E-CAD was significantly
`repressed, by the TGF-β treatment, while N-CAD, TNC, VIM,
`and FN 1 expression were considerably increased (Figure 1E).
`In contrast, TGF-β treatment induced only a modest effect on
`the four tumor-derived cell lines analyzed (Figure 1A-D). No E-
`CAD expression could be detected in any condition in B-CPAP,
`TPC1, and WRO cells, whereas CDH16 expression was
`detected in all cell lines and was significantly repressed only in
`TPC1 cells. Western Blot analysis of some of these markers in
`TGF-β treated Nthy.ori 3.1, B-CPAP and TPC1 cells confirmed
`the validity of the qRT-PCR results (Figure 1F). TGF-β
`treatment on the metastatic FTC133 cells resulted in increased
`E-CAD expression. This may be consistent with the need of
`metastatic cells to reverse the EMT phenotype necessary for
`metastatic site colonization [1]. Noticeably, CDH6 was strongly
`induced in all cell lines analyzed, with the exception of the
`WRO cells. While in tumor-derived cells CDH6 expression
`increased two-threefold after TGF-β treatment, in Nthy.ori 3.1
`CDH6 induction was much more pronounced (eight-fifteen
`fold), similar to the one observed for TNC and superior to the
`induction observed for N-CAD and FN 1. These results
`demonstrate that CDH6 is a TGF-β target in thyroid cells and
`that its expression is modulated similarly to other mesenchymal
`markers. With the intent to understand whether this could be a
`thyroid specific mechanism, we have extended this analysis to
`other types of tumor. To this purpose, A549 lung cancer, A375
`melanoma and MDA-MB-231 breast cancer cell lines were
`treated with TGF-β and expression of CDH6 was monitored by
`qRT-PCR. We were not able to detect CDH6 expression in
`either A375 or MDA-MB-231 cell lines (data not shown). In
`A549 lung cancer cell line CDH6 was expressed and induced
`upon TGF-β treatment at levels comparable to the thyroid
`tumor cell lines (Figure S1). These results suggest that CDH6
`expression is tissue specific and the ability of TGF-β to target
`this gene is common to other types of tumor.
`All together, our observations suggested that tumor-derived
`cells are less responsive to the TGF-β than normal thyrocytes.
`
`PLOS ONE | www.plosone.org
`
`3
`
`September 2013 | Volume 8 | Issue 9 | e75489
`
`3 of 16
`
`OnCusp
`Ex. 1027
`
`

`

`Table 1. Origin and mutational status of thyroid-derived cell
`lines.
`
`FTC133
`B-CPAP TPC1 WRO
`Nthy.ori 3.1
`Cell line
`FTC-LNM
`PTC
`PTC FTC
`Normal thyroid
`Origin
`wt
`V600E
`wt
`wt/V600E*
`nd
`BRAF
`wt
`wt
`wt
`wt
`nd
`NRAS
`wt
`wt
`wt
`wt
`nd
`HRAS
`wt
`wt
`wt
`wt
`nd
`KRAS
`wt
`wt
`wt
`wt
`nd
`PI3KCA
`R130stop
`wt
`wt
`wt
`nd
`PTEN
`R273H
`D259Y
`wt
`wt
`nd
`TP53
`-
`-
`+
`-
`nd
`RET/PTC1
`-
`-
`-
`-
`nd
`RET/PTC3
`-
`-
`-
`-
`nd
`PAX8/PPARg
`PTC, Papillary Thyroid Carcinoma; FTC, Follicular Thyroid Carcinoma; FTC-LNM,
`Lymph Node Metastasis from Follicular Thyroid Carcinoma; nd, not determined; wt,
`wild-type; * BRAF V600E mutation in WRO cell line is reported only by some
`authors; + presence of rearrangement; - absence of rearrangement (modified from
`Saiselet et al. 2012) [21].
`doi: 10.1371/journal.pone.0075489.t001
`
`A lower expression of the TGF-β receptors in thyroid tumor
`cells could result in a minor capacity to transduce the TGF-β
`signal. To test this hypothesis, we compared the expression
`levels of the TGF-β receptors 1 and 2 in the five cell lines by
`qRT-PCR analysis. As shown in Figure 1G and H, all cell lines
`displayed comparable levels of the TGFR1, while a more
`heterogeneous expression of the TGFR2 was observed.
`However, in contrast with the extent of the TGF-β response, all
`tumor-derived cells, with the exception of the FTC133 cells,
`had higher levels of the TGFR2 than the Nthy.ori 3.1 cells.
`
`The EMT program is constitutively active in tumor-
`derived cells.
`We investigated the possibility that tumor-derived cells have
`already undergone an EMT transformation by comparing the
`expression
`levels of
`the above-mentioned EMT markers
`between normal Nthy.ori 3.1 thyrocytes and tumor-derived
`cells. Noticeably, the expression of all mesenchymal markers
`analyzed (N-CAD, TNC, VIM and FN 1) was significantly higher
`in the tumor cells than in the Nthy.ori 3.1 thyrocytes. By
`contrast, E-CAD expression could be detected only in the
`Nthy.ori 3.1 and FTC133 cells, while no expression of this
`epithelial marker was detected in the other tumor-derived cells
`(Figure 2A). CDH16 expression was higher in TPC1 and
`FTC133 than in the rest of the cell lines. Noticeably, CDH6
`expression was greatly enhanced in all thyroid tumor-derived
`cells as compared to the normal thyrocytes (Figure 2B).
`Western blot analysis was in accordance with the gene
`expression data (Figure 2C). Then we tested whether the EMT-
`like phenotype displayed by
`thyroid
`tumor cells was
`accompanied by a constitutive activation of
`the TGF-β
`downstream pathways. TGF-β signals within the cells through
`three major pathways: nuclear translocation of the SMAD2/3
`proteins, the activation of the MAPK, and of PI3K cascade [25].
`
`CDH6 in TGF-β Mediated EMT Program
`
`Since our interest was mainly on the mechanism controlling the
`biology of papillary thyroid tumors, we limited the analysis to
`papillary-derived tumor cells. Nthy.ori 3.1, B-CPAP, and TPC1
`cells were treated with TGF-β and changes in the activation of
`the three signaling pathways were monitored. In the Nthy.ori
`3.1 thyrocytes, the levels of both pERK and pAKT rose early
`after the addition of TGF-β. In B-CPAP cells, pERK levels did
`not change in response to TGF-β, while AKT phosphorylation
`was significantly induced. In TPC1 cells, phosphorylation levels
`of both ERK and AKT did not change in response to TGF-β
`(Figure 2D). Furthermore, in untreated Nthy. ori.3.1 cells
`SMAD2/3 were localized in the cytoplasm, to be strongly
`translocated to nuclei early after TGF-β exposure (Figure 2E
`upper panels). In untreated B-CPAP cells SMAD2/3 were
`localized mainly in the nuclei and their localization did not
`change significantly in the presence of TGF-β (Figure 2E
`middle panels). In untreated TPC1 cells, the SMAD2/3 staining
`was visible in both nucleus and cytoplasm. However, after
`TGF-β treatment, the cytoplasmic staining was no longer
`detectable, indicating that all the protein was translocated to
`the nuclei (Figure 2E lower panels). Overall, these data
`suggest
`that TGF-β-dependent signaling cascades are
`constitutively active in thyroid tumor cells. These results may
`be partially explained by the fact that tumor cells, including the
`B-CPAP and TPC1, often harbor somatic mutations
`in
`components of these signaling pathways (see Table 1).
`However, tumor cells retain a partial ability to respond to the
`TGF-β signal by further activating some of these pathways, like
`AKT in the B-CPAP and SMAD2/3 in the TPC1. The EMT
`program is controlled by a number of transcription factors,
`which are known targets of TGF-β [5,26]. Both Id1 and RUNX2
`have been shown to be TGF-β targets and have been
`proposed as crucial regulators of the EMT program [8,27-29].
`We
`investigated how
`the expression of EMT-related
`transcription factors changed in response to TGF-β in thyroid
`cell lines. SNAI1, SNAI2, Id1, and RUNX2 were induced by
`TGF-β in both Nthy.ori 3.1 and TPC1 cells. In accordance with
`what was observed for the EMT markers (Figure 1), the
`activation of SNAI1, SNAI2, and Id1 was lower in TPC1 than in
`Nthy.ori 3.1 (Figure 2F-H). In B-CPAP cells slight change in
`SNAI1, SNAI2, ZEB1 and TWIST expression was observed
`after 24h (Figure 2G). ZEB1 and TWIST levels were not
`affected in Nthy.ori 3.1 and TPC1 cells. It is worth pointing out
`that the induction of Id1 and RUNX2 preceded the activation of
`the other factors, suggesting that these factors are early targets
`of the TGF-β-dependent EMT program.
`
`CDH6 is overexpressed in human PTCs and mainly
`localized at the invasive front of the tumors.
`To confirm in vivo that thyroid tumor cells display features of
`a constitutively active EMT-program, we analyzed
`the
`expression of EMT markers
`in human papillary
`thyroid
`carcinoma
`(PTC) samples and matched
`lymph node
`metastases (LNMs). We collected total RNA from tumor and
`surrounding normal tissue for 15 PTC patients and we
`analyzed EMT markers by means of qRT-PCR (Figure 3A).
`Noticeably, the expression of mesenchymal markers (N-CAD,
`TNC, and FN 1) was significantly higher in the vast majority of
`
`PLOS ONE | www.plosone.org
`
`4
`
`September 2013 | Volume 8 | Issue 9 | e75489
`
`4 of 16
`
`OnCusp
`Ex. 1027
`
`

`

`CDH6 in TGF-β Mediated EMT Program
`
`Figure 1. Nthy.ori 3.1 thyrocytes are more responsive to the TGF-β-mediated EMT program than tumor-derived cells. A-E)
`qRT-PCR analysis of EMT markers (E-CAD; N-CAD; CDH16; TNC; VIM; FN 1) and CDH6 in non-treated (NT; black bars) or TGF-β
`treated (5 ng/ml grey bars; 100 ng/ml white bars) thyroid-derived cell lines. The bars represent the average fold change of indicated
`genes in TGF-β treated cells as compared to non-treated cells, normalized to the geometric mean of levels of three reference
`genes: GAPDH, CYPA, GUSB. F) Western Blot analysis of E-CAD, N-CAD, FN 1, and Actin in Nthy.ori 3.1 cells, B-CPAP and TPC1
`cells non-treated (NT) or treated with 100 ng/ml of TGF-β for 6h and 24h. G, H) qRT-PCR analysis of TGFR1 (G) and TGFR2 (H) in
`non-treated thyroid-derived cell lines. The bars represent the average fold change of TGFR1 and TGFR2 in tumor cells (B-CPAP,
`TPC1, WRO, and FTC-133) as compared to thyrocytes (Nthy.ori 3.1), normalized to the to the geometric mean of GAPDH, CYPA,
`GUSB levels. Error bars represent s.e.m. (n=3). p-value was calculated by two-tailed Student’s t-test. *** p≤ 0.001; ** p≤ 0.01; * p≤
`0.05. Each experiment has been replicated a minimum of two times with comparable results.
`doi: 10.1371/journal.pone.0075489.g001
`
`PLOS ONE | www.plosone.org
`
`5
`
`September 2013 | Volume 8 | Issue 9 | e75489
`
`5 of 16
`
`OnCusp
`Ex. 1027
`
`

`

`CDH6 in TGF-β Mediated EMT Program
`
`Figure 2. Tumor-derived cell lines display a constitutive EMT-like phenotype. qRT-PCR analysis of EMT markers (A) (E-
`CAD; N-CAD; CDH16; TNC; VIM; FN 1) and CDH6 (B) in non-treated thyroid-derived cell lines. The bars represent the average fold
`change of the indicated genes in tumor cells (B-CPAP, TPC1, WRO, and FTC-133) as compared to thyrocytes (Nthy.ori 3.1),
`normalized to the geometric mean of levels of three reference genes: GAPDH, CYPA, GUSB. C) Western Blot analysis of E-CAD,
`N-CAD, FN1 and Actin in non-treated Nthy.ori 3.1; B-CPAP and TPC1 cells. D) Western Blot analysis of phosphorylated ERK,
`phosphorylated AKT, and Actin in Nthy.ori 3.1 (Left panels); B-CPAP (middle panels) and TPC1 (right panels) cells untreated (NT)
`or after TGF-β exposure for the indicated times. E) Immunofluorescence staining of SMAD2/3 proteins (green) in Nthy.ori 3.1 (upper
`panels); B-CPAP (middle panels) and TPC1 (lower panels), non-treated (NT) or after TGF-β exposure for the indicated times. DAPI
`(Blue) stains the nuclei. Magnification 200X. F-H) qRT-PCR analysis of transcription factors known to partake in the EMT program
`(SNAI1, SNAI2, ZEB1, TWIST, Id1, and RUNX2) in Nthy.ori 3.1 (E), B-CPAP (F) and TPC1 (G) cells, non-treated (NT) or treated
`with TGF-β for the indicated times. The bars represent the fold change of the indicated genes in TGF-β treated cells as compared to
`the non-treated cell levels, normalized to the geometric mean of GAPDH, CYPA, GUSB levels. p-value was calculated by two-tailed
`Student’s t-test. *** p≤ 0.001; ** p≤ 0.01; * p≤ 0.05. Error bars represent s.e.m. (n=3).
`doi: 10.1371/journal.pone.0075489.g002
`
`PLOS ONE | www.plosone.org
`
`6
`
`September 2013 | Volume 8 | Issue 9 | e75489
`
`6 of 16
`
`OnCusp
`Ex. 1027
`
`

`

`tumors (black circles) than in normal tissues (baseline). By
`contrast, CDH16, marker of the epithelial phenotype, was
`significantly less expressed in tumor cells than in normal
`thyrocytes. E-CAD expression was not significantly different in
`tumor sample as compared with the normal tissue. LNMs were
`available for 7 of the analyzed PTC samples. The expression
`pattern of EMT markers in LNM (black and white circles)
`reflected the trend observed in the primary lesions, confirming
`that active EMT features are a constitutive hallmark of tumor
`transformation in thyroid tumors. To confirm that CDH6 is a
`marker of the mesenchymal phenotype in thyroid tumors, we
`investigated the expression of CDH6 in the aforementioned
`PTC samples by qRT-PCR (Figure 3B). As for the other
`mesenchymal markers, CDH6 expression was higher in tumor
`tissue (black circles) and LNMs
`than
`in normal
`tissue
`(baseline). The CDH6 expression levels seemed to decrease in
`the transition from primary to metastatic site, suggesting that
`this factor may be required particularly in the early phases of
`the metastasization process. Next, we analyzed the CDH6
`protein expression by immunohistochemistry in the same PTC
`samples previously analyzed by qRT-PCR. As expected, CDH6
`expression was clearly higher in tumor cells than in normal
`tissue (Figure 3C and Figure S2A-F). However, the expression
`of CDH6 was not uniform within the tumor but was restricted to
`groups of cells. Intriguingly, CDH6-positive cells were localized
`preferentially at the invasion front of the tumor, strongly
`supporting the hypothesis that CDH6 is important in controlling
`cell motility and invasiveness in PTCs (Figure 3C and Figure
`S2A and B).
`
`CDH6 splicing variants display a different profile in
`thyroid tumor-derived cells
`The CDH6 gene codes for two different splicing isoforms,
`starting from a common promoter. The long isoform comprises
`both an extracellular domain (containing 4 ectodomain modules
`–EC– and a membrane proximal extracellular domain–MPE)
`and a cytoplasmic domain (containing the membrane proximal
`conserved domain–MPC- and the conserved catenin binding
`site–CBS) [7] (Figure S2G). Instead, the short isoform displays
`only the extracellular domain but lacks the cytoplasmic domain.
`Due to the ability of transmembrane proteins to transduce the
`signals within the cell through their cytoplasmic domains, it is
`reasonable to suppose that the two variants may play different
`roles. To address this issue, we analyzed the expression of the
`two CDH6 isoforms by qRT-PCR using specific primers (Figure
`S2G). Both isoforms were expressed in all cell lines (Figure 4A)
`and more strongly expressed in tumor-derived cells than in
`Nthy.ori 3.1 (Figure 4B), as observed for the total CDH6
`(Figure 2B). Intriguingly, the expression change in tumor-
`derived cells (B-CPAP and TPC1) as compared to the Nthy.ori
`3.1 was more pronounced for the CDH6-long isoform (CDH6-L)
`than for the short isoform (CDH6-S). Further, we analyzed
`changes in the expression levels of CDH6-L and CDH6-S
`isoforms by TGF-β treatment. Both isoforms were similarly
`induced by TGF-β in all cell lines (Figure 4C, D, E). This
`suggests
`that TGF-β controls CDH6 expression at
`the
`transcriptional level. To confirm this hypothesis we investigated
`the effect of TGF-β treatment on CDH6 expression in the
`
`CDH6 in TGF-β Mediated EMT Program
`
`absence of active transcription (with Actinomycin D) or in the
`absence of active protein synthesis (with Cycloheximide). As
`shown in Figure 5A, the addition of Actinomycin D completely
`abolished the TGF-β-mediated CDH6 induction, confirming that
`the regulatory effect of TGF-β requires an active transcription.
`Furthermore, the addition of Cycloheximide also resulted in a
`complete suppression of TGF-β induction, demonstrating that
`the effect on the CDH6 expression is indirect and requires the
`previous synthesis of an activator factor.
`
`RUNX2 mediates the TGF-β effect in thyroid cells
`In a recent microarray analysis, we found CDH6 as a target
`of Id1 in aggressive thyroid tumor cells [8]. Further, we showed
`that the transcription factor RUNX2, also a target of Id1, is a
`crucial mediator of the Id1 pro-invasive function in thyroid
`tumor cells [30]. Both RUNX2 and Id1 have been proposed to
`control EMT in epithelial tumors. We reasoned that RUNX2
`could be responsible for the TGF-β-dependent CDH6 induction.
`To test this hypothesis, Nthy.ori 3.1 cells were transfected with
`siRNA against RUNX2 or control siRNA and treated with TGF-
`β for 24h. As shown in Figure 5B, the siRNA-mediated RUNX2
`depletion profoundly impaired the TGF-β effect on CDH6
`expression. These experiments confirm that CDH6 is a target
`of RUNX2 in the TGF-β pathway. However, siRNA mediated
`RUNX2 ablation does not completely abolish the TGF-β effect
`on CDH6 expression suggesting that other TGF-β–dependent
`factors beside RUNX2 may be involved in controlling this gene
`in thyroid tumor cells.
`RUNX2 has been shown to control the expression of SNAI
`factors [29]. After TGF-β treatment, RUNX2 induction occurs
`earlier that SNAI1 and SNAI2 activation. Thus, it is possible
`that the effect of RUNX2 on CDH6 expression could be indirect
`and mediated by SNAI factors. However, RUNX2 silencing
`does not affect TGF-β- dependent SNAI1 or SNAI2 induction
`suggesting that these transcription factors are not involved in
`this regulation (Figure S2H-I).
`We showed that TGF-β signaling pathway is constitutively
`activated in thyroid tumor cells and suggested that RUNX2 and
`CDH6 is part of this pathway. If this hypothesis is correct
`blocking the TGF-β signaling in thyroid cancer cells should
`affect RUNX2 and CDH6 expression levels. To address this
`issue we treated B-CPAP and TPC1 cells with SB-431542, a
`small molecule known to block kinase activity of the TGF-β
`receptors family [30]. Noticeably, exposure to SB-431542
`results in a strong repression of CDH6 and RUNX2 in both B-
`CPAP and TPC1 cells, which was clearly visible after 24h and
`increased after 48h from the beginning of the treatment (Figure
`5C,D). The inhibitory effect of th

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket