Stemness and epithelial–mesenchymal transition (EMT) are two fundamental characteristics of metastasis that are controlled by diverse regulatory factors, including transcription factors. Compared with other subtypes of breast cancer, basal-type or triple-negative breast cancer (TNBC) has high frequencies of tumor relapse. However, the role of alpha-globin transcription factor CP2 (TFCP2) has not been reported as an oncogenic driver in those breast cancers. Here, we show that TFCP2 is a potent factor essential for EMT, stemness, and metastasis in breast cancer. TFCP2 directly bound promoters of EGF and TGFα to regulate their expression and stimulate autocrine signaling via EGFR. These findings indicate that TFCP2 is a new antimetastatic target and reveal a novel regulatory mechanism in which a positive feedback loop comprising EGF/TGFα and AKT can control malignant breast cancer progression.

Significance:

TFCP2 is a new antimetastatic target that controls TNBC progression via a positive feedback loop between EGF/TGFα and the AKT signaling axis.

Tumor metastasis is a multistep process that occurs through local invasion, intravasation, transport, extravasation, and colonization, resulting in the dissemination of tumor cells from their primary site to a distant site where secondary tumors are formed. In the tumor metastasis process, epithelial–mesenchymal transition (EMT), and cancer stem cell (CSC)-like characteristics make primary contributions to drive tumor initiation and development (1). During the EMT process, epithelial cells shift toward a mesenchymal phenotype, with the loss of cell–cell contacts and adhesions and an increased ability for migration and invasion during morphogenesis (2). After the EMT program is activated, cancer cells can also exhibit high plasticity by acquiring stem-like traits that endow them with the potential for tumor initiation and metastasis, all of which promote cancer progression. Hence, tumor cells that undergo EMT and possess CSC-like features have greater metastatic potential and result in poorer outcomes in patients with cancer.

TFCP2 is a member of the CP2/Grainyhead family of transcription factors that are conserved throughout metazoans and fungi. In mammals, the CP2/Grainyhead family consists of two distinct subfamilies: the first includes three Grainyhead-like factors currently termed GRHL1–3, and the other subfamily consists of three factors known as TFCP2 (CP2c), TFCP2L1 (CRTR1), and UBP1 (CP2a and CP2b; refs. 3, 4). Among these, TFCP2, also known as LSF, was first identified as a transcriptional activator factor of the Simian virus 40 late promoter in HeLa cells (5) and of the murine α-globin promoter (6). As mentioned earlier, TFCP2 regulates a diverse range of cellular and viral promoters (7, 8), is expressed in all mammalian cell types, and plays an important role in cell-cycle regulation (8). It facilitates entry into G1–S-phase of the cell cycle, promotes DNA synthesis, and functions as an antiapoptotic factor (9). Overexpression of TFCP2 may promote transformation and cancer cell survival. Recent studies suggest that TFCP2 may also play a role in the pathogenesis of colon, lung, and hepatocellular carcinoma (10–12). Moreover, TFCP2 has been shown to activate osteopontin and matrix metalloproteinase-9 expression to regulate invasion, metastasis, and angiogenesis in hepatocellular carcinoma (HCC; refs. 13, 14). Although the expression and regulatory roles of TFCP2 have been reported individually in some types of cancer, there is no evidence of TFCP2 involvement in metastatic triple-negative breast cancer (TNBC) progression.

The EGFR is a major oncogene identified in a variety of human cancers, including breast cancer (15–17). These receptors are activated by ligand binding and consequent receptor homo- and heterodimerization, which leads to activation of the kinase domain, auto- and transphosphorylation of their intracellular domains, and the initiation of signaling (18). Many different ligands, including EGF-like molecules, TGFα, and neuregulins, activate the receptor by binding to the extracellular domain and inducing the formation of receptor homodimers or heterodimers (19). Genes in the EGF signaling pathway are among the most frequently activated oncogenes in breast cancer (20). Clinical studies have shown that TNBC is more aggressive than other breast cancers and patients with TNBC have a worse prognosis than those with other breast cancer subtypes (21). More than 60% of TNBCs express EGFR, which may serve as a prognostic marker for TNBC outcomes (22). Although EGFR has been widely studied in breast cancer tumorigenesis, the mechanism underlying TFCP2-induced malignancy remains unknown. The goal of this study was to determine the role of TFCP2 in breast cancer progression, with a focus on EMT, stemness, and metastasis. Using basal-like breast cancer cell lines and NOD/SCID gamma (NSG, 5–6 weeks old) mouse models, we investigated the mechanisms by which TFCP2 affects the EGFR signaling axis positively or negatively in TNBCs.

Materials and reagents

All breast cell lines (MCF10A, MCF7, SKBR3, T47D, BT474, MDA-MB-453, MDA-MB-231, BT549, Hs578T, and MDA-MB-361) were purchased from ATCC, cultured in the indicated media, and incubated at 37°C with 5% CO2 according to standard protocols. They were routinely tested for Mycoplasma contamination using PCR methods and cultured cell lines were often treated with Plasmocin treatment (InvivoGen) to remain contamination free. Cells were used at least less than 20 passages number for 3 months, but were not independently authenticated. After each thawing, cells were confirmed as Mycoplasma negative prior to experiments. The p-EF1α (control vector), p-EF1α-CP2 [TFCP2 wild-type (WT) vector], TFCP2 D153A, and LSFdn vectors were received from Department of Life Science, Hanyang University (Seoul, South Korea), and SNAI1 (pBabe puro Snail) and pGL3-basic vectors were purchased from Addgene. EGF (#AFL236) and TGFα (#239-A) were purchased from R&D Systems, Inc. Antibodies and inhibitors' information are listed in Supplementary Table S1.

Transfection

p-EF1α, p-EF1α-CP2, TFCP2 D153A, LSFdn, and SNAI1 vectors or siRNAs were transfected into the appropriate cells using Lipofectamine Reagents (Invitrogen) according to the manufacturer's instructions. Cells were harvested 48 hours after transfection for subsequent experiments. All siRNAs were purchased from Genolution Pharmaceuticals, Inc.

Scratch, soft agar, morphology (collagen coating), and transwell assays

For the wound-healing (scratch) assay, cells were seeded in 35-mm cell culture dishes and cultured until 80%–90% confluent. Afterwards, a 200-μL pipet tip was used to make a scratch wound across the middle of the cell monolayer. Images were taken with Olympus IX71 Fluorescence Microscope (Olympus) immediately after and 24 hours after the scratch was made. The rate of cell migration from at least three independent experiments was calculated with ImageJ. To examine anchorage-independent growth, cells were suspended in 0.4% agar in growth medium and analyzed as described previously (23). For morphologic analysis, cells were seeded at medium confluency directly into Corning BioCoat Collagen I-Coated Plates (Corning Inc.) and photographed 24 hours later. Migration assays were performed using Boyden Chambers (Corning Inc.). Cells (2 × 104) in 200 μL of serum-free medium were seeded into the top chamber, and 800 μL of medium with 10% FBS was added to the bottom chamber as a chemoattractant. Migratory cells were stained using a Diff-Quick Kit (Thermo Fisher Scientific) then imaged and counted. The invasion assays were carried out in accordance with the migration assays except that each transwell chamber was coated with growth factor–reduced Matrigel (BD Biosciences). All experiments were performed in triplicate.

Spheroid assays

For the sphere formation assays, sphere size was determined using Motic Images Plus 2.0 Software (Motic) in three randomly chosen visual fields each day until day 4 after the cells were seeded. For colony formation assays, single-cell suspensions containing breast cells were plated in 96-well cell culture plates and colony formation was observed at different time points for 2 weeks. Colonies were photographed and their diameter was measured (24).

IHC analysis

For IHC experiments, paraffin-embedded tissue sections were deparaffinized in xylene and then rehydrated in a graded series of ethanol (95%, 90%, 80%, and 70%), followed by PBS treatment. Epitopes were retrieved with 20 mg/mL proteinase K in PBS containing 0.1% Triton X-100. Sections were incubated overnight with appropriate primary antibodies at 4°C and then processed as described previously (25).

RNA preparation and qPCR

Total RNA was prepared using TRIzol Reagent (Invitrogen), and RNA quality was measured using a NanoDrop Spectrophotometer (ND1000, NanoDrop Technologies). qRT-PCR was performed using a KAPA SYBR FAST qPCR Kit (KAPA Biosystems) according to the manufacturer's procedures. Reactions were performed in a Rotor Gene Q Instrument (Qiagen), and the results were expressed as the fold-change relative to the control sample calculated using the ΔΔCt method. β-actin served as an internal normalization control. All primers were purchased from DNA Macrogen. All primers used in this study are listed in Supplementary Tables S2 and S3.

Western blotting

Total cellular protein was extracted in cold lysis buffer [Tris–HCl (40 mmol/L, pH 8.0), NaCl (120 mmol/L), and Nonidet-P40 (0.1%)] enriched with protease inhibitors and was quantified using a BSA assay. Protein lysates were separated by SDS-PAGE and transferred onto nitrocellulose membranes (Amersham). The membranes were then blocked in PBST containing 5% milk and probed with the indicated primary antibody followed by the corresponding horseradish peroxidase–conjugated secondary antibody. Finally, the protein bands were detected using chemiluminescence (Amersham) according to the manufacturer's instructions.

Flow cytometry analysis

To assess cell death, cells were incubated in the listed conditions for the desired time, after which, they were labeled with propidium iodide (Sigma, 50 ng/mL), incubated for 20 minutes at 4°C, and analyzed immediately. To detect the cancer stemness marker CD44 and CD24, 1 × 106 of TFCP2 knockdown cells were harvested by trypsin digestion, washed, and resuspended in PBS. An R-phycoerythrin–conjugated anti-CD44 mAb and FITC-conjugated anti-CD24 antibody (Miltenyi Biotec Inc.) were used for detection. All data were analyzed using CellQuest Software (BD Biosciences) and repeated three times.

ELISA

Cellular protein was prepared as Western blotting. The concentrations of total and phosphorylated EGFR were measured using human EGFR (pY1068) and total EGFR ELISA Kits (Abcam), respectively, according to the manufacturer's instructions.

Chromatin immunoprecipitation assays

Prior to performing chromatin immunoprecipitation (ChIP) experiments, cells were cross-linked with 4% paraformaldehyde. ChIP assays were performed using an EZ-ChIP Kit (EMD Millipore) according to the manufacturer's instructions. Immunoprecipitation was performed using an anti-TFCP2 antibody or a rabbit isotype control IgG (Upstate Biotechnology). PCR was performed using primers specific to the EGF and TGFα gene promoter regions shown in Supplementary Table S3.

Construction of luciferase reporter plasmids

The human EGF and TGFα promoter regions were obtained by PCR, using genomic DNA from MDA-MB-231 cells as a template. The promoters of EGF and TGFα were generated by PCR using primers shown in Supplementary Table S3. They were subcloned into XhoI and HindIII sites of pGL3-basic vector (Addgene). All constructs were verified by sequencing.

Luciferase reporter assay

HEK293T cells were seeded in a 24-well plate, at 60%–70% confluence and cells were cotransfected using Lipofectamine 2000 according to the manufacturer's manual. In brief, each well was transfected with 300 ng of reporter constructs and 300 ng of pRL-CMV-Renilla plasmid (Promega) for 48 hours. Luciferase activity was measured using a Dual-Luciferase Reporter Assay System (Promega) according to the manufacturer's instructions and normalized to Renilla luciferase activity. All experiments were performed in triplicate.

In vivo xenografts and metastasis assays

All animal procedures were performed according to the guidelines of the Institutional Animal Care and Use Committee of Academia Sinica. NSG mice (5–6 weeks old) were obtained from Orient Bio. For these experiments, 40 μL of metastatic MDA-MB-231-LM1 breast cancer cells (1 × 106), which were derived from lung lesions after colonization (shCtrl-LM1 and shTFCP2-LM1), were injected into the fourth mammary fat pad of NSG mice. Twice per week, the mice were weighed and tumor size was determined using a digital caliper. Tumor volumes were assessed by measuring the length (l) and width (w) and calculated using the following formula: (shortest diameter2) × (longest diameter/2). Mice were sacrificed 8–12 weeks after injection, and the lungs were removed and fixed in 9% paraformaldehyde. Detectable tumor nodules on the surface of the entire lung were counted to calculate the metastatic index. Tumor tissues were homogenized, and the expression of target genes was analyzed by Western blotting and qRT-PCR.

Immunofluorescence

For immunofluorescence staining, cells were plated onto glass coverslips, fixed with 4% paraformaldehyde, and permeabilized with 0.1% Triton in PBS. Cells then incubated overnight at 4°C with the appropriate primary antibody. The following day, Alexa Fluor 488–conjugated anti-rabbit or anti-mouse and Alexa Fluor 546–conjugated anti-rabbit or anti-mouse (Molecular Probes) secondary antibodies were used to visualize the proteins. Cell nuclei were counterstained with 4′,6-diamidino-2-phenylindole (Sigma). Immunostained cells were observed using an IX71 Fluorescence Microscope (Olympus).

Human tissue microarrays

Breast tissue microarray samples were obtained from US Biomax (BR1101, BR20814, and BR1509). Healthy specimens were also included in each of the array blocks. Samples were reviewed by a pathologist to confirm the diagnosis of breast carcinoma, histologic grade, and tumor purity. TFCP2, EGF, and TGFα levels were graded as 0, 1, 2, or 3 according to the intensity scores.

Gene set enrichment analysis, dataset evaluation, and Kaplan–Meier analysis

Gene set enrichment analysis (GSEA) was performed on diverse gene signatures by comparing gene sets from either the Molecular Signature Database (MSigDB) or published gene signatures. To analyze the expression of TFCP2, TFCP2L1, and UBP1 in breast invasive carcinomas, previously published microarray data under accession codes GSE41313, GSE1456, GSE20713, GSE7513, GSE2603, and GSE25055 were reanalyzed. To examine the prognostic value of TFCP2, EGF, and TGFα, patient samples were divided into two groups (low and high expression) for each gene, which were analyzed using the Kaplan–Meier plot program (http://kmplot.com/analysis/) as described previously (26).

Statistical analysis

All experimental data are presented as the mean ± SD of at least three independent experiments. Statistical analyses were performed using an unpaired two-tailed parametric Student t test. Multiple group comparisons were made by ANOVA using PRISM 8.0 Software (GraphPad). Variances were confirmed to be similar between groups that were being statistically compared, and P < 0.05 was considered significant. No samples were excluded from the analysis. The investigators were not blinded to allocation during experiments and outcome assessments.

TFCP2 is upregulated and associated with poor survival in patients with breast cancer

To explore the association of CP2 family transcription factors in breast cancer, we utilized an online database screening system to investigate their expression in normal and breast cancer tissues using gene expression profiling interactive analysis (27). Assessment of the dataset showed that TFCP2 and UBP1 expression is comparatively higher in breast tumors than in normal tissues, but not TFCP2L1 (Fig. 1A). To verify further, additionally, GSEA was performed with data from the MSigDB, which showed that TFCP2 expression is well-correlated with aggressive basal subtypes of breast cancer compared with the luminal type (Fig. 1B; Supplementary Fig. S1) while UBP1 does not seem so, suggesting the possible involvement of TFCP2 in basal type breast tumors. Consistent data were also observed in basal and luminal breast cancer subtypes using a Gene Expression Omnibus (GEO) dataset (28) and Gene Expression-Based Outcome for Breast Cancer Online (29) datasets (Fig. 1C and D). In agreement with the online database findings, quantification of TFCP2, TFCP2L1, and UBP1 mRNA expression shows that the basal-type cell lines expressed high levels of TFCP2 compared with the luminal cell lines (Fig. 1E). After validating the specificity of TFCP2 levels in malignant breast cancer, we confirmed its expression in human tissue samples using tissue microarray analysis. Notably, increased TFCP2 expression was observed in basal and high-grade breast carcinomas (Fig. 1F and G). Furthermore, Kaplan–Meier survival analysis (30) determined that high expression of TFCP2 correlates with poor survival in patients with breast cancer independently among basal, luminal, and HER2 subtypes (Fig. 1H). These observations strongly suggest that TFCP2 is primarily associated with malignant breast cancer.

Figure 1.

High TFCP2 expression was associated with invasive metastatic breast carcinoma. A, TFCP2, TFCP2L1, and UBP1 expression data in normal breast tissue (n = 291) and breast cancer tissue (n = 1,085) from patients with breast cancer were obtained from the GEPIA database (http://gepia.cancer-pku.cn/index.html). B, Heatmap analyses showed that TFCP2 is upregulated (red) in basal-type (n = 61) versus luminal-type (n = 50) breast cancers in the GSEA datasets (GSE41313). Blue, downregulation. C and D, TFCP2, TFCP2L1, and UBP1 expression data in basal (n = 61) and luminal (n = 80) human breast cancer types were obtained from the GEO database (human patients; GSE41313; https://www.ncbi.nlm.nih.gov/geo) and the GOBO database (human cell lines; http://co.bmc.lu.se/gobo/). E, qRT-PCR was performed to detect the expression of TFCP2 subfamily members in various basal- and luminal-type breast cancer cell lines (n = 3). F, Tissue microarray analysis of TFCP2 expression in basal (n = 87) and luminal (n = 95) type of breast carcinoma tissues. G, Analysis of TFCP2 expression in breast cancer through gradewise. High TFCP2 protein expression was correlated with higher tumor grade breast carcinoma. Scale bar, 100 μm. H, Kaplan–Meier survival analysis for TFCP2 in breast cancer cohorts (basal, HER2, and luminal) based on low and high expression (GSE1456, n = 159). Values in the graph represent the means ± SD (n = 3). ***, P < 0.001; ns, not significant; determined by two-tailed Student t test (95% confidence interval.

Figure 1.

High TFCP2 expression was associated with invasive metastatic breast carcinoma. A, TFCP2, TFCP2L1, and UBP1 expression data in normal breast tissue (n = 291) and breast cancer tissue (n = 1,085) from patients with breast cancer were obtained from the GEPIA database (http://gepia.cancer-pku.cn/index.html). B, Heatmap analyses showed that TFCP2 is upregulated (red) in basal-type (n = 61) versus luminal-type (n = 50) breast cancers in the GSEA datasets (GSE41313). Blue, downregulation. C and D, TFCP2, TFCP2L1, and UBP1 expression data in basal (n = 61) and luminal (n = 80) human breast cancer types were obtained from the GEO database (human patients; GSE41313; https://www.ncbi.nlm.nih.gov/geo) and the GOBO database (human cell lines; http://co.bmc.lu.se/gobo/). E, qRT-PCR was performed to detect the expression of TFCP2 subfamily members in various basal- and luminal-type breast cancer cell lines (n = 3). F, Tissue microarray analysis of TFCP2 expression in basal (n = 87) and luminal (n = 95) type of breast carcinoma tissues. G, Analysis of TFCP2 expression in breast cancer through gradewise. High TFCP2 protein expression was correlated with higher tumor grade breast carcinoma. Scale bar, 100 μm. H, Kaplan–Meier survival analysis for TFCP2 in breast cancer cohorts (basal, HER2, and luminal) based on low and high expression (GSE1456, n = 159). Values in the graph represent the means ± SD (n = 3). ***, P < 0.001; ns, not significant; determined by two-tailed Student t test (95% confidence interval.

Close modal

TFCP2 enhances EMT, metastasis, and stemness in breast cancer

To discover the pathologic mechanism of TFCP2 upregulation in breast cancer, we screened hallmarks of cancer with the GSEA database in breast cancer. Gene ontology enrichment analysis showed that TFCP2 is positively correlated with signature gene sets relating to EMT and cancer stemness (Fig. 2A). When we ectopically introduced TFCP2 in luminal MCF7 and SKBR3 breast cancer cells to examine EMT, MCF7 and SKBR3 cells showed a reduction in epithelial features and an increase in mesenchymal features as evidenced by the elongation of cells on the collagen-coated surface, enhanced cell invasion and migration abilities and the induction of fibronectin (FN), N-cadherin (CDH2), and vimentin (VIM) expression (Fig. 2BE; Supplementary Fig. S2A–S2D). Similar results were also observed in MCF10A normal breast epithelial cells with TFCP2 overexpression (Supplementary Fig. S2E–S2H). Prior to the TFCP2 overexpression experiments in MCF7, additional experiments with TFCP2 family members TFCP2L1 and UBP1 were performed, which confirmed that silencing of these genes did not affect the migration and invasion capabilities of Hs578T and MDA-MB-453 cells, which expresses high level of TFCP2L1 and UBP1, respectively (Supplementary Fig. S3A and S3B). In addition, TFCP2 knockdown (with siRNA#1 and siRNA#2) mitigated these effects in both MDA-MB-231 and BT549 basal-type breast cancer cells (Supplementary Fig. S3C–S3H). Recent studies have documented that the acquisition of CSC traits occurs with the EMT program (31, 32) and the induction of EMT can induce many of the defining characteristics of stem cells, including self-renewal (33). In accordance with these studies and our GSEA analysis data (Fig. 2A), we tried to determine the involvement of TFCP2 in the acquisition of stemness in breast cancer cells. The sphere-forming or clonal efficiency of breast cancer cells was dramatically reduced after TFCP2 knockdown in both EGF-treated (a well-known EMT inducer) MCF7 and SKBR3 cells in a sphere-permissive medium (Fig. 2FI). The inhibitory effect of TFCP2 on stem-like breast cells was observed until several passages, as evidenced by limiting dilution sphere-forming assays with MCF7 cells (Supplementary Fig. S3I). We also screened several CSC-related transcription factors, such as SOX2, NANOG, and OCT4. qRT-PCR and immunofluorescence data showed that OCT4 was prominently affected along with CD44 after TFCP2 knockdown in spheres formed from MCF7 or SKBR3 cells (Supplementary Fig. S3J and S3K). EMT-related transcription factors have been reported to boost the CD44+/CD24 subpopulation, as observed in breast CSCs (34). In agreement, flow cytometry analysis revealed that the percentage of CD44+ and CD24 cells were decreased in both MCF7 and SKBR3 cells after TFCP2 knockdown (Fig. 2J). In addition, we also performed sphere formation assay and checked percentage of CD44+/CD24 by FACS analysis in TFCP2 overexpression system using MCF7 cells. We found that overexpressed TFCP2 can induce cancer stemness in those MCF7 cells (Supplementary Fig. S3L and S3M). On the other hand, interestingly, silencing of TFCP2 did not affect breast cancer cell proliferation, as confirmed by soft agar–independent cell growth and cell death assays (Supplementary Fig. S3N–S3P). These results showed that TFCP2 has the potential to regulate EMT and the CSC phenotype rather than cell proliferation and death in breast cancer cells.

Figure 2.

TFCP2 potentiated EMT, metastasis, and the CSC phenotype in breast cancer cell lines. A, GSEA (GSE41313) analysis was performed in TFCP2-positive breast cancers for hallmarks of cancer progression, including signature genes of the EMT and CSC phenotypes. GSEA analyses indicated that TFCP2 significantly upregulated genes involved in the EMT and CSC processes. ES, enrichment score; NES, normalized enrichment score. B, Representative images of the morphology of MCF7 breast cancer cells overexpressing TFCP2. TFCP2 overexpression was performed 48 hours prior to experiments. Scale bar, 10 μm. C, Migration and invasion assays were performed using MCF7 cells overexpressing TFCP2. D and E, qRT-PCR and Western blot analysis of EMT regulators (ZEB1, SNAI1, and SNAI2) and EMT markers (FN, CDH2, and VIM) in TFCP2-overexpressing MCF7 cells after 48 hours of overexpression. F, Sphere formation assays in EGF (100 ng/μL)-treated MCF7 and SKBR3 cells with TFCP2 silencing (right). Cell lines were treated with EGF for 6 hours after starvation overnight prior to TFCP2 silencing and TFCP2 knockdown efficiency was analyzed by Western blot analysis (left) after 48 hours of silencing. Scale bar, 10 μm. G, The total number of spheres in three independent fields was counted and plotted in a graph. H, Sphere formation of single-cell suspensions at different time points under similar treatment conditions. I, The average sphere size was measured after 14 days in indicated panels using Motic Images Plus 2.0 software. Scale bar, 100 μm. J, Flow cytometry analysis of the CSC markers of CD44+/CD24 in EGF-treated MCF7 and SKBR3 cells with TFCP2 knockdown. EGF treatment and TFCP2 knockdown was performed similarly in these cells as mentioned above. β-actin was used as a control (Ctrl) for normalization. All these experiments were performed in triplicates and values are presented as SD. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant; determined by two-tailed Student t test (95% confidence interval).

Figure 2.

TFCP2 potentiated EMT, metastasis, and the CSC phenotype in breast cancer cell lines. A, GSEA (GSE41313) analysis was performed in TFCP2-positive breast cancers for hallmarks of cancer progression, including signature genes of the EMT and CSC phenotypes. GSEA analyses indicated that TFCP2 significantly upregulated genes involved in the EMT and CSC processes. ES, enrichment score; NES, normalized enrichment score. B, Representative images of the morphology of MCF7 breast cancer cells overexpressing TFCP2. TFCP2 overexpression was performed 48 hours prior to experiments. Scale bar, 10 μm. C, Migration and invasion assays were performed using MCF7 cells overexpressing TFCP2. D and E, qRT-PCR and Western blot analysis of EMT regulators (ZEB1, SNAI1, and SNAI2) and EMT markers (FN, CDH2, and VIM) in TFCP2-overexpressing MCF7 cells after 48 hours of overexpression. F, Sphere formation assays in EGF (100 ng/μL)-treated MCF7 and SKBR3 cells with TFCP2 silencing (right). Cell lines were treated with EGF for 6 hours after starvation overnight prior to TFCP2 silencing and TFCP2 knockdown efficiency was analyzed by Western blot analysis (left) after 48 hours of silencing. Scale bar, 10 μm. G, The total number of spheres in three independent fields was counted and plotted in a graph. H, Sphere formation of single-cell suspensions at different time points under similar treatment conditions. I, The average sphere size was measured after 14 days in indicated panels using Motic Images Plus 2.0 software. Scale bar, 100 μm. J, Flow cytometry analysis of the CSC markers of CD44+/CD24 in EGF-treated MCF7 and SKBR3 cells with TFCP2 knockdown. EGF treatment and TFCP2 knockdown was performed similarly in these cells as mentioned above. β-actin was used as a control (Ctrl) for normalization. All these experiments were performed in triplicates and values are presented as SD. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant; determined by two-tailed Student t test (95% confidence interval).

Close modal

Silencing TFCP2 expression blocks EMT and stemness in vivo

We further investigated the effect of TFCP2 knockdown on metastasis in a breast cancer mouse models by injecting LM1-MDA-MB-231 cells into the fat pad (Fig. 3A). As shown in the knockdown experiments, diminished TFCP2 expression reduced the formation of lung metastatic foci with inhibition of EMT- and CSC-related transcription factors and markers such as FN, CDH2, VIM, OCT4, and CD44 (Fig. 3BJ). However, tumor growth remained unaffected in these xenografts after TFCP2 knockdown (Supplementary Fig. S4A–S4C). Measurement of Ki67 and cleaved caspase 3 in shTFCP2-injected mouse tissues confirmed that TFCP2 did not induce any cell death in breast tumors (Supplementary Fig. S4D and S4E). These data further revealed a critical role of TFCP2 in maintaining mesenchymal features to sustain EMT and metastasis with stemness in basal-type breast cancer without affecting growth or cell death.

Figure 3.

Depletion of TFCP2 inhibited EMT and CSC progression in vivo. A, LM1 cells stably transfected with shCtrl or shTFCP2 (1 × 106/40 μL per mouse, 5 weeks) were injected into the fourth mammary fat pad of NSG mice (n = 5 per group). The silencing efficiency of shTFCP2 in MDA-MB-231 LM1 cells was confirmed by Western blotting. B, Images of lung sections in the shCtrl and shTFCP2 NSG mice groups. The graph shows the numbers of lung metastatic foci in the respective groups. C and D, qRT-PCR and Western blot analysis of the expression levels of EMT regulators and markers in tumor tissues from control and TFCP2 knockdown mice groups. E, IHC staining of SNAI1, FN, VIM, CDH2, and CDH1 expression in tumor tissues from shCtrl- and shTFCP2-injected mice. F, Representative graph showing the IHC staining scores in both groups. G and H, qRT-PCR and Western blot analysis of CSC marker and regulators (CD44, SOX2, NANOG, and OCT4) in shCtrl- and shTFCP2-treated mouse tissues. I and J, IHC staining assays and intensity score analysis of CSC markers and regulators in control and TFCP2 knockdown mouse tissues. Scale bar, 100 μm. β-actin was used as a control for normalization. Data are presented as mean of three independent experiments (SD). *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant; determined by two-tailed Student t test (95% confidence interval).

Figure 3.

Depletion of TFCP2 inhibited EMT and CSC progression in vivo. A, LM1 cells stably transfected with shCtrl or shTFCP2 (1 × 106/40 μL per mouse, 5 weeks) were injected into the fourth mammary fat pad of NSG mice (n = 5 per group). The silencing efficiency of shTFCP2 in MDA-MB-231 LM1 cells was confirmed by Western blotting. B, Images of lung sections in the shCtrl and shTFCP2 NSG mice groups. The graph shows the numbers of lung metastatic foci in the respective groups. C and D, qRT-PCR and Western blot analysis of the expression levels of EMT regulators and markers in tumor tissues from control and TFCP2 knockdown mice groups. E, IHC staining of SNAI1, FN, VIM, CDH2, and CDH1 expression in tumor tissues from shCtrl- and shTFCP2-injected mice. F, Representative graph showing the IHC staining scores in both groups. G and H, qRT-PCR and Western blot analysis of CSC marker and regulators (CD44, SOX2, NANOG, and OCT4) in shCtrl- and shTFCP2-treated mouse tissues. I and J, IHC staining assays and intensity score analysis of CSC markers and regulators in control and TFCP2 knockdown mouse tissues. Scale bar, 100 μm. β-actin was used as a control for normalization. Data are presented as mean of three independent experiments (SD). *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant; determined by two-tailed Student t test (95% confidence interval).

Close modal

Because TFCP2 knockdown affects SNAI1 expression more effectively than the other EMT transcription factors tested, we hypothesized that TFCP2 mediated metastatic activity through the SNAI1 pathway. We knocked down TFCP2 with siRNA in SNAI1-overexpressing MDA-MB-231 basal-type breast cancer cells. Consistent with previous experiments, exogenous SNAI1 expression recovered TFCP2-mediated inhibition of migration, invasion, and expression of the mesenchymal markers FN, CDH2, and VIM in those cells (Supplementary Fig. S4F–S4H). Similar effects were observed when MCF7 cells in spheroids were treated with similar conditions. When overexpressing SNAI1, MCF7 cells with TFCP2 knockdown regained the ability to form spheres and exhibited a stemness phenotype along with CD44+ cells (Supplementary Fig. S4I–S4L). Thus, we suggest that SNAI1 drives TFCP2-induced EMT and stemness, thereby sustaining metastatic programming in basal-type breast cancer cells.

TFCP2 regulates EGFR signaling activation in breast cancer

To achieve mechanism selectivity, GSEA analysis was performed to investigate which signaling pathways are involved in the TFCP2-induced prometastatic effects in basal-type breast cancer. We found that there is a highly positive correlation between TFCP2 expression and EGFR signaling along with its downstream signaling pathway (Fig. 4A). Next, we performed ELISA and Western blot assays to determine whether TFCP2 regulates EGFR activity. Both assays showed that TFCP2 enhanced EGFR protein level activity based on the TFCP2 silencing and overexpression experiments; however, the EGFR mRNA expression levels did not change (Fig. 4BD; Supplementary Fig. S5A and S5B). EGFR is widely involved in a variety of cellular processes, including proliferation, motility, and survival, and can be activated by a variety of polypeptide ligands, such as EGF and HBEGF. Because TFCP2 regulates EGFR activity, we assessed the expression level of several EGFR ligands, including EGF, TGFα, epiregulin (EREG), amphiregulin (AREG), epithelial mitogen (EPGN), heparin-binding EGF (HBEGF), and betacellulin (BTC), all of which can activate EGFR. Silencing and overexpressing TFCP2 revealed that TFCP2 more profoundly affects EGF and TGFα than all the other ligands tested in MDA-MB-231 and MCF7 cells, respectively (Fig. 4E; Supplementary Fig. S5C). Subsequently, we hypothesized that if there is any correlation between TFCP2 and both the EGF and TGFα ligands. We screened breast cancer cohorts through GEO databases and found a positive correlation of TFCP2 with both EGF and TGFα (Fig. 4F). Luciferase assays confirmed the binding of TFCP2 on EGF and TGFα promoters, suggesting their role in TFCP2-induced EGFR signaling (Fig. 4G). To further confirm this correlation, we predicted TFCP2-binding sites to EGF and TGFα promoters using the JASPAR online tool (http://jaspar.genereg.net) and performed ChIP assays using MDA-MB-231 and Hs578T cell lines. We found that TFCP2 can directly bind to the F2 fragment site of the EGF promoter and F1-4 fragment sites of the TGFα promoter in both cells (Fig. 4H and I). In addition, when TFCP2 D153A (which mutated aspartate (D) to alanine (A) at the TFCP2 DNA binding site), LSF dominant negative (LSFdn, double amino acid substitution mutant of LSF that is unable to bind DNA, initially named 234QL/236KE; ref. 9), and TFCP2 WT constructs were overexpressed in MCF7 cells, we observed that only TFCP2 WT significantly upregulated EGF and TGFα mRNA expression along with EGFR protein activity; however, there was no change observed in cells overexpressing TFCP2 D153A or LSFdn, which was similar to the levels in the control cells (Supplementary Fig. S5D and S5E). ChIP assay also confirmed that neither TFCP2 D153A nor LSFdn can bind to EGF or TGFα promoters (Supplementary Fig. S5F). It is worth mentioning that inhibiting EGFR signaling in TFCP2-overexpressing MCF7 cells suppressed signature genes related to EMT and CSC, which further suggests that TFCP2-induced EGFR activation regulates both phenotypes in breast cancer (Supplementary Fig. S5G).

Figure 4.

TFCP2 induced EGF and TGFα expression to activate EGFR signaling in breast cancer. A, Major functional pathways modulated by TFCP2 in breast cancer cells based on transcriptome analysis using GSEA (GSE7513) analysis. NES, normalized enrichment score. Among the identified pathways, EGFR signaling showed a positive correlation with high expression levels of TFCP2 based on a dataset from the GEO database (https://www.ncbi.nlm.nih.gov/geo/). Enrichment plots are shown in various panels and are stratified by high versus low expression of TFCP2. Quantification of expression data is shown in the graph. B and C, EGFR and AKT activation was determined by Western blot analysis; TFCP2 and EGFR mRNA analysis by qPCR both in TFCP2-silenced MDA-MB-231 and Hs578T cell lines after 48 hours. D, ELISA was used to measure total and phosphorylated EGFR levels in TFCP2 knockdown MDA-MB-231 cells after 48 hours. E, qRT-PCR analysis for screening the expression of EGFR ligands in MDA-MB-231 cells with silenced TFCP2 after 48 hours. F, A positive correlation between TFCP2 expression and EGF/TGFα was obtained from the GEO database (GSE2603, n = 121; GSE25055, n = 310). G, Luciferase reporter assays for TFCP2 directly binding to the EGF and TGFα promoters. H, Schematic figure shows predicted TFCP2 binding sites of the EGF and TGFα promoter regions. I, The ChIP assay showed that TFCP2 directly binds to the EGF and TGFα promoters at specific sites in basal type cell lines MDA-MB-231 and Hs578T. J, Rescued experiments were performed using Western blot analysis in TFCP2-silenced MDA-MB-231 and Hs578T cells with recombinant EGF or TGFα after 48 hours. β-Actin was used as a control for normalization. Data are presented as mean of three independent experiments (SD). *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant; determined by two-tailed Student t test (95% confidence interval).

Figure 4.

TFCP2 induced EGF and TGFα expression to activate EGFR signaling in breast cancer. A, Major functional pathways modulated by TFCP2 in breast cancer cells based on transcriptome analysis using GSEA (GSE7513) analysis. NES, normalized enrichment score. Among the identified pathways, EGFR signaling showed a positive correlation with high expression levels of TFCP2 based on a dataset from the GEO database (https://www.ncbi.nlm.nih.gov/geo/). Enrichment plots are shown in various panels and are stratified by high versus low expression of TFCP2. Quantification of expression data is shown in the graph. B and C, EGFR and AKT activation was determined by Western blot analysis; TFCP2 and EGFR mRNA analysis by qPCR both in TFCP2-silenced MDA-MB-231 and Hs578T cell lines after 48 hours. D, ELISA was used to measure total and phosphorylated EGFR levels in TFCP2 knockdown MDA-MB-231 cells after 48 hours. E, qRT-PCR analysis for screening the expression of EGFR ligands in MDA-MB-231 cells with silenced TFCP2 after 48 hours. F, A positive correlation between TFCP2 expression and EGF/TGFα was obtained from the GEO database (GSE2603, n = 121; GSE25055, n = 310). G, Luciferase reporter assays for TFCP2 directly binding to the EGF and TGFα promoters. H, Schematic figure shows predicted TFCP2 binding sites of the EGF and TGFα promoter regions. I, The ChIP assay showed that TFCP2 directly binds to the EGF and TGFα promoters at specific sites in basal type cell lines MDA-MB-231 and Hs578T. J, Rescued experiments were performed using Western blot analysis in TFCP2-silenced MDA-MB-231 and Hs578T cells with recombinant EGF or TGFα after 48 hours. β-Actin was used as a control for normalization. Data are presented as mean of three independent experiments (SD). *, P < 0.05; **, P < 0.01; ***, P < 0.001; ns, not significant; determined by two-tailed Student t test (95% confidence interval).

Close modal

Rescue experiments were performed in both MDA-MB-231 and Hs578T cell lines with silenced TFCP2 in the presence or absence of recombinant EGF/TGFα. Interestingly, EMT-related SNAI1, CDH2, and VIM protein expression; migration; and invasion were decreased upon silencing TFCP2; these decreases were rescued again with the treatment of EGF or TGFα in MDA-MB-231 and Hs578T cells (Fig. 4J; Supplementary Fig. S5H). Similar effects were observed in TFCP2-overexpressing MCF7 cells with or without EGF/TGFα silencing (Supplementary Fig. S5I). Taken together, these results indicated that TFCP2 can directly upregulate EGF and TGFα expression to activate the EGFR signaling pathway in metastatic breast cancer cells.

A positive feedback loop exists between TFCP2 and the EGFR/PI3K/AKT axis

Because TFCP2 could regulate EGF/TGFα expression to activate EGFR activation, we investigated the upstream regulator of TFCP2. To this end, we examined TFCP2 expression in the presence of various signaling pathway inhibitors, such as U0126 (a MEK1/2 inhibitor), a JNK inhibitor, SB203580 (a p38 MAP kinase), a JAK inhibitor, WP1066 (a STAT3 inhibitor), LY294002 (a PI3 kinase/AKT inhibitory), and PP2 (a SRC inhibitor). Prior to this experiment, the efficacy of the inhibitors was confirmed by Western blot analysis (Supplementary Fig. S6A). These analyses revealed that inhibiting the AKT pathway downregulates TFCP2 mRNA levels to a greater extent than inhibition of other pathways in MDA-MB-231 cells (Fig. 5A). To further confirm that the AKT pathway was blocked in the same cells with AKT-targeted siRNA or LY294002, the TFCP2 protein level was measured and shown to be dramatically reduced as observed by Western blot analysis (Fig. 5B; Supplementary Fig. S6B). As we found above, EGFR can be activated through TFCP2, and we next asked whether AKT could regulate EGFR through TFCP2. Knockdown or pharmacologic blockade of AKT attenuated EGFR and TFCP2 activity in MDA-MB-231 cells, while this attenuation was rescued in the presence of TFCP2 overexpression. The expression of SNAI1, FN, CDH2, CD44, and OCT4 in MDA-MB-231 cells was also affected by LY294002 treatment, which was rescued with TFCP2 overexpression (Fig. 5C and D; Supplementary Fig. S6C). This result supported the view that AKT serves an essential upstream regulator of TFCP2, thereby inducing EGFR activation to promote metastatic activity. Earlier, we observed that concomitant treatment with recombinant EGF/TGFα increases TFCP2 expression in TFCP2-silenced MDA-MB-231 cells compared with that in cells with TFCP2 knockdown alone (Fig. 4J). Hence, we hypothesized whether there is any positive feedback loop such as EGFR activation can also regulate TFCP2 expression in breast cancer. To verify this, we analyzed TFCP2 expression in the presence of either si-EGFR or an EGFR inhibitor (AG1478). Both approaches decreased the protein activity and mRNA expression of TFCP2 in MDA-MB-231 cells (Fig. 5E; Supplementary Fig. S6D and S6E), suggesting potential involvement of EGFR in TFCP2 induction. Similar observations were also made in BT549 cells by inhibiting EGFR on TFCP2 levels (Supplementary Fig. S6F). To evaluate whether the EGFR/PI3K/AKT axis is a true upstream candidate of TFCP2 feedback regulation, we treated MCF7 cells with recombinant EGF/TGFα in the presence or absence of LY294002. TFCP2 expression was greatly enhanced in the presence of EGF/TGFα alone, but this effect was reduced when LY294002 was added to the cells (Fig. 5F). These data indicated that TFCP2 expression is upregulated by the EGFR/PI3K/AKT axis in metastatic breast cancer cells through a positive feedback loop.

Figure 5.

EGFR increased TFCP2 expression in breast cancer via a positive feedback loop. A, Detection of TFCP2 mRNA expression in MDA-MB-231 cells after treatment with U0126 (10 μmol/L, ERK inhibitor), a JNK inhibitor (10 μmol/L), SB203580 (25 μmol/L, P38 inhibitor), a JAK inhibitor (10 μmol/L), a STAT3 inhibitor (10 μmol/L), LY294002 (10 μmol/L, PI3K inhibitor), and PP2 (10 μmol/L, SRC inhibitor). TFCP2 mRNA expression was analyzed after 24 hours of inhibitors treatment. B, Western blot analysis of TFCP2 levels in MDA-MB-231 cells after 24 hours of treatment with LY294002. C and D, Western blot and immunofluorescence analysis for p-EGFR, TFCP2, SNAI1, FN, CDH2, OCT4, and CD44 protein levels in MDA-MB-231 cells overexpressing TFCP2 and treated with si-AKT. si-AKT was treated together with TFCP2 overexpression before 48 hours of experiment. Scale bar, 10 μm. E, Analysis of TFCP2 protein and mRNA expression in MDA-MB-231 cells at 24 hours after treatment with the EGFR inhibitor AG1478 (10 μmol/L) using Western blotting and qRT-PCR, respectively. F, Rescue experiments for the detection of TFCP2 mRNA expression in MCF7 cells after 6 hours treatment with EGF and TGFα (100 ng/μL) in the absence or presence of LY294002. Scale bar, 100 μm. β-Actin was used as a control (Ctrl) for normalization. Data are presented as mean of three independent experiments (SD). **, P < 0.01; ***, P < 0.001; ns, not significant; determined by two-tailed Student t test (95% confidence interval).

Figure 5.

EGFR increased TFCP2 expression in breast cancer via a positive feedback loop. A, Detection of TFCP2 mRNA expression in MDA-MB-231 cells after treatment with U0126 (10 μmol/L, ERK inhibitor), a JNK inhibitor (10 μmol/L), SB203580 (25 μmol/L, P38 inhibitor), a JAK inhibitor (10 μmol/L), a STAT3 inhibitor (10 μmol/L), LY294002 (10 μmol/L, PI3K inhibitor), and PP2 (10 μmol/L, SRC inhibitor). TFCP2 mRNA expression was analyzed after 24 hours of inhibitors treatment. B, Western blot analysis of TFCP2 levels in MDA-MB-231 cells after 24 hours of treatment with LY294002. C and D, Western blot and immunofluorescence analysis for p-EGFR, TFCP2, SNAI1, FN, CDH2, OCT4, and CD44 protein levels in MDA-MB-231 cells overexpressing TFCP2 and treated with si-AKT. si-AKT was treated together with TFCP2 overexpression before 48 hours of experiment. Scale bar, 10 μm. E, Analysis of TFCP2 protein and mRNA expression in MDA-MB-231 cells at 24 hours after treatment with the EGFR inhibitor AG1478 (10 μmol/L) using Western blotting and qRT-PCR, respectively. F, Rescue experiments for the detection of TFCP2 mRNA expression in MCF7 cells after 6 hours treatment with EGF and TGFα (100 ng/μL) in the absence or presence of LY294002. Scale bar, 100 μm. β-Actin was used as a control (Ctrl) for normalization. Data are presented as mean of three independent experiments (SD). **, P < 0.01; ***, P < 0.001; ns, not significant; determined by two-tailed Student t test (95% confidence interval).

Close modal

Correlation between TFCP2 and EGF/TGFα expression in patient samples

To evaluate whether our in vitro findings are correlated with human breast cancer and metastasis formation, we assessed the prognostic value of TFCP2 expression with EGF and TGFα in breast cancer patient samples. To this end, we performed IHC analysis to examine the coexpression of TFCP2, EGF, and TGFα in tumors derived from patients with breast carcinoma (n = 150). Tissue array data showed that expression of TFCP2, EGF, and TGFα was significantly higher in tumor tissues than in matched normal tissues (Fig. 6A). The increased TFCP2 expression correlated well with the augmented expression of EGF and TGFα (Fig. 6B). Furthermore, Kaplan–Meier survival analysis revealed that the survival time of patients with high TFCP2 and either EGF or TGFα expression was shorter than those with low expression of these genes (Fig. 6C and D). In addition, high expression of TFCP2, EGF, and TGFα was found to be significantly associated with poor outcomes in patients with breast cancer, as shown in Fig. 6E. Overall, clinical dataset analysis indicates that TFCP2 levels are positively correlated with EGFR signaling in breast tumors, and higher TFCP2 levels are associated with reduced metastasis-free survival in breast tumors and an increased probability of poor overall survival in EGF/TGFα-high tumors.

Figure 6.

Correlation between TFCP2 expression and EGF/TGFα in patients with breast cancer. A, Representative IHC images of TFCP2, EGF, and TGFα in breast cancer and corresponding normal tissues. Scale bar, 100 μm. Distribution of the TFCP2, EGF, and TGFα staining intensity in breast cancer tissues. B, The association between TFCP2 and both EGF and TGFα expression in breast cancer tissues. The number of cases and the percentage of positive staining in the corresponding groups as well as the statistical significance based on Student t tests and Pearson correlations of expression are shown in the table. C–E, Kaplan–Meier survival analysis showed that high levels of TFCP2 expression along with high levels of either EGF or TGFα expression or high levels of all three were associated with lower survival rates of patients with breast cancer. F, Schematic representation of the TFCP2/EGFR/PI3K/AKT axis mechanism in metastatic breast cancer.

Figure 6.

Correlation between TFCP2 expression and EGF/TGFα in patients with breast cancer. A, Representative IHC images of TFCP2, EGF, and TGFα in breast cancer and corresponding normal tissues. Scale bar, 100 μm. Distribution of the TFCP2, EGF, and TGFα staining intensity in breast cancer tissues. B, The association between TFCP2 and both EGF and TGFα expression in breast cancer tissues. The number of cases and the percentage of positive staining in the corresponding groups as well as the statistical significance based on Student t tests and Pearson correlations of expression are shown in the table. C–E, Kaplan–Meier survival analysis showed that high levels of TFCP2 expression along with high levels of either EGF or TGFα expression or high levels of all three were associated with lower survival rates of patients with breast cancer. F, Schematic representation of the TFCP2/EGFR/PI3K/AKT axis mechanism in metastatic breast cancer.

Close modal

Here, we demonstrated that TFCP2 functions as an activator of prometastatic transcription factors by directly regulating the expression of the EGFR ligands EGF and TGFα, resulting in EGFR activation; this signaling cascade is a critical determinant of oncogenic EGFR signaling, leading to poor long-term survival in patients with breast cancer. Although various studies have shown that EMT affects the migratory potential of metastatic breast cancer cells and subsequent metastasis, many critical factors regarding how these tumor cells access this fundamental cellular event remain unknown. In this study, we found that the transcription factor, TFCP2, promotes breast cancer cells to acquire increased migration/invasion abilities and CSC traits via the EGFR signaling pathway, but has no effect on tumor growth and apoptosis. As we know, EGFR overexpression is frequently observed in TNBC (16, 35–37), the aggressive behavior of TNBC and the lack of established clinical treatment targets create a major challenge in treating these patients. In this article, we showed that high levels of TFCP2 expression in TNBC cells increased the activation of the EGFR signaling pathway. In this pathway, ligands play several significant roles at different levels to promote invasion and metastasis (38). Furthermore, our ChIP assay data suggested that TFCP2 directly binds to the promoters of the EGFR ligands EGF and TGFα (Fig. 4H and I). Dysregulation of the EGFR pathway via overexpression or constitutive activation can promote processes related to tumor progression, such EMT and stemness, and is associated with poor prognosis in breast malignancies (39–41). Hence, inhibiting TFCP2 expression, which can suppress EGF and TGFα expression, may be effective at preventing EGFR activation in TNBC.

In a previous study, TFCP2 was shown to be overexpressed in HCC and to target FN1 and TJP1 to regulate HCC metastasis (42). Moreover, TFCP2 can affect pancreatic cancer cell growth, invasion, and migration (43). However, in these studies, the mechanism by which TFCP2 expression is regulated was not fully addressed. In this study, we found that TFCP2 expression was mediated by the EGFR/PI3K/AKT axis in TNBC, which caused a positive feedback loop (Fig. 5). However, there are some limitations to this study. First, we identified a signaling pathway that can regulate TFCP2 expression, but the specific regulators of TFCP2 expression remain unclear. Second, although TFCP2 induced both EGF and TGFα expression to activate EGFR, we could not distinguish which ligand was more important for EGFR signaling. Third, TFCP2 regulated the EGF/TGFα/EGFR axis in breast cancer; however, we do not know whether this also occurs in other human malignancies. Further studies are required to determine the role of TFCP2 in other cancer types.

In summary, we describe the functions of TFCP2 as an oncogenic driver in TNBC. In addition, we identified the underlying mechanisms whereby TFCP2 regulates EMT and CSC activity in breast cancer through the EGFR/AKT axis. In turn, TFCP2 expression is also regulated by the EGFR/AKT axis (Fig. 6F). Enhanced expression of TFCP2 was associated with poor patient survival and therefore lends itself as a potential target for metastatic breast cancer treatment.

No potential conflicts of interest were disclosed.

Conception and design: Y. Zhao, C.G. Kim, S.-J. Lee

Development of methodology: Y. Zhao, N. Uddin, S.-J. Lee

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Y. Zhao, J.-H. Kang, N.K. Kaushik, S.-J. Lee

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): Y. Zhao, N.K. Kaushik, S.H. Son, S.-J. Lee

Writing, review, and/or revision of the manuscript: Y. Zhao, N. Kaushik, S.H. Son, S.-J. Lee

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): Y. Zhao, J.-H. Kang, M.-J. Kim, S.-J. Lee

Study supervision: C.G. Kim, S.-J. Lee

We are thankful to all the research participants who participated in this study. This study was supported by the Bio & Medical Technology Development Program of the National Research Foundation funded by the Korean government (MSIT; 2019M3E5D1A01069361 to S.-J. Lee).

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

1.
Zhou
P
,
Li
B
,
Liu
F
,
Zhang
M
,
Wang
Q
,
Liu
Y
, et al
The epithelial to mesenchymal transition (EMT) and cancer stem cells: implication for treatment resistance in pancreatic cancer
.
Mol Cancer
2017
;
16
:
52
.
2.
Tsai
JH
,
Yang
J
. 
Epithelial-mesenchymal plasticity in carcinoma metastasis
.
Genes Dev
2013
;
27
:
2192
206
.
3.
Traylor-Knowles
N
,
Hansen
U
,
Dubuc
TQ
,
Martindale
MQ
,
Kaufman
L
,
Finnerty
JR
. 
The evolutionary diversification of LSF and Grainyhead transcription factors preceded the radiation of basal animal lineages
.
BMC Evol Biol
2010
;
10
:
101
.
4.
Kang
HC
,
Chae
JH
,
Lee
YH
,
Park
MA
,
Shin
JH
,
Kim
SH
, et al
Erythroid cell-specific alpha-globin gene regulation by the CP2 transcription factor family
.
Mol Cell Biol
2005
;
25
:
6005
20
.
5.
Kim
CH
,
Heath
C
,
Bertuch
A
,
Hansen
U
. 
Specific stimulation of simian virus 40 late transcription in vitro by a cellular factor binding the simian virus 40 21-base-pair repeat promoter element
.
Proc Natl Acad Sci U S A
1987
;
84
:
6025
9
.
6.
Kim
CG
,
Barnhart
KM
,
Sheffery
M
. 
Purification of multiple erythroid cell proteins that bind the promoter of the alpha-globin gene
.
Mol Cell Biol
1988
;
8
:
4270
81
.
7.
Veljkovic
J
,
Hansen
U
. 
Lineage-specific and ubiquitous biological roles of the mammalian transcription factor LSF
.
Gene
2004
;
343
:
23
40
.
8.
Hansen
U
,
Owens
L
,
Saxena
UH
. 
Transcription factors LSF and E2Fs: tandem cyclists driving G0 to S?
Cell Cycle
2009
;
8
:
2146
51
.
9.
Powell
CM
,
Rudge
TL
,
Zhu
Q
,
Johnson
LF
,
Hansen
U
. 
Inhibition of the mammalian transcription factor LSF induces S-phase-dependent apoptosis by downregulating thymidylate synthase expression
.
EMBO J
2000
;
19
:
4665
75
.
10.
Jiang
H
,
Du
J
,
Jin
J
,
Qi
X
,
Pu
Y
,
Fei
B
. 
LSF expression and its prognostic implication in colorectal cancer
.
Int J Clin Exp Pathol
2014
;
7
:
6024
31
.
11.
Santhekadur
PK
,
Rajasekaran
D
,
Siddiq
A
,
Gredler
R
,
Chen
D
,
Schaus
SE
, et al
The transcription factor LSF: a novel oncogene for hepatocellular carcinoma
.
Am J Cancer Res
2012
;
2
:
269
85
.
12.
Zhang
X
,
Sun
F
,
Qiao
Y
,
Zheng
W
,
Liu
Y
,
Chen
Y
, et al
TFCP2 is required for YAP-dependent transcription to stimulate liver malignancy
.
Cell Rep
2017
;
21
:
1227
39
.
13.
Yoo
BK
,
Gredler
R
,
Chen
D
,
Santhekadur
PK
,
Fisher
PB
,
Sarkar
D
. 
c-Met activation through a novel pathway involving osteopontin mediates oncogenesis by the transcription factor LSF
.
J Hepatol
2011
;
55
:
1317
24
.
14.
Santhekadur
PK
,
Gredler
R
,
Chen
D
,
Siddiq
A
,
Shen
XN
,
Das
SK
, et al
Late SV40 factor (LSF) enhances angiogenesis by transcriptionally up-regulating matrix metalloproteinase-9 (MMP-9)
.
J Biol Chem
2012
;
287
:
3425
32
.
15.
Camidge
DR
,
Pao
W
,
Sequist
LV
. 
Acquired resistance to TKIs in solid tumours: learning from lung cancer
.
Nat Rev Clin Oncol
2014
;
11
:
473
81
.
16.
Masuda
H
,
Zhang
D
,
Bartholomeusz
C
,
Doihara
H
,
Hortobagyi
GN
,
Ueno
NT
. 
Role of epidermal growth factor receptor in breast cancer
.
Breast Cancer Res Treat
2012
;
136
:
331
45
.
17.
Hynes
NE
,
Lane
HA
. 
ERBB receptors and cancer: the complexity of targeted inhibitors
.
Nat Rev Cancer
2005
;
5
:
341
54
.
18.
Yarden
Y
,
Sliwkowski
MX
. 
Untangling the ErbB signalling network
.
Nat Rev Mol Cell Biol
2001
;
2
:
127
37
.
19.
Yarden
Y
. 
The EGFR family and its ligands in human cancer. Signalling mechanisms and therapeutic opportunities
.
Eur J Cancer
2001
;
37
:
S3
8
.
20.
Lane
A
,
Segura-Cabrera
A
,
Komurov
K
. 
A comparative survey of functional footprints of EGFR pathway mutations in human cancers
.
Oncogene
2014
;
33
:
5078
89
.
21.
Dent
R
,
Trudeau
M
,
Pritchard
KI
,
Hanna
WM
,
Kahn
HK
,
Sawka
CA
, et al
Triple-negative breast cancer: clinical features and patterns of recurrence
.
Clin Cancer Res
2007
;
13
:
4429
34
.
22.
Arnes
JB
,
Begin
LR
,
Stefansson
I
,
Brunet
JS
,
Nielsen
TO
,
Foulkes
WD
, et al
Expression of epidermal growth factor receptor in relation to BRCA1 status, basal-like markers and prognosis in breast cancer
.
J Clin Pathol
2009
;
62
:
139
46
.
23.
Kim
RK
,
Kim
MJ
,
Seong
KM
,
Kaushik
N
,
Suh
Y
,
Yoo
KC
, et al
Beneficial effects of low dose radiation in response to the oncogenic KRAS induced cellular transformation
.
Sci Rep
2015
;
5
:
15809
.
24.
Eom
H
,
Kaushik
N
,
Yoo
KC
,
Shim
JK
,
Kwon
M
,
Choi
MY
, et al
MerTK mediates STAT3-KRAS/SRC-signaling axis for glioma stem cell maintenance
.
Artif Cells Nanomed Biotechnol
2018
;
46
:
87
95
.
25.
Cui
YH
,
Kim
H
,
Lee
M
,
Yi
JM
,
Kim
RK
,
Uddin
N
, et al
FBXL14 abolishes breast cancer progression by targeting CDCP1 for proteasomal degradation
.
Oncogene
2018
;
37
:
5794
809
.
26.
Labidi-Galy
SI
,
Clauss
A
,
Ng
V
,
Duraisamy
S
,
Elias
KM
,
Piao
HY
, et al
Elafin drives poor outcome in high-grade serous ovarian cancers and basal-like breast tumors
.
Oncogene
2015
;
34
:
373
83
.
27.
Tang
Z
,
Li
C
,
Kang
B
,
Gao
G
,
Li
C
,
Zhang
Z
. 
GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses
.
Nucleic Acids Res
2017
;
45
:
W98
W102
.
28.
Edgar
R
,
Domrachev
M
,
Lash
AE
. 
Gene Expression Omnibus: NCBI gene expression and hybridization array data repository
.
Nucleic Acids Res
2002
;
30
:
207
10
.
29.
Ringner
M
,
Fredlund
E
,
Hakkinen
J
,
Borg
A
,
Staaf
J
. 
GOBO: gene expression-based outcome for breast cancer online
.
PLoS One
2011
;
6
:
e17911
.
30.
Gyorffy
B
,
Lanczky
A
,
Eklund
AC
,
Denkert
C
,
Budczies
J
,
Li
Q
, et al
An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients
.
Breast Cancer Res Treat
2010
;
123
:
725
31
.
31.
Singh
A
,
Settleman
J
. 
EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer
.
Oncogene
2010
;
29
:
4741
51
.
32.
Mani
SA
,
Guo
W
,
Liao
MJ
,
Eaton
EN
,
Ayyanan
A
,
Zhou
AY
, et al
The epithelial-mesenchymal transition generates cells with properties of stem cells
.
Cell
2008
;
133
:
704
15
.
33.
Brabletz
T
,
Jung
A
,
Spaderna
S
,
Hlubek
F
,
Kirchner
T
. 
Opinion: migrating cancer stem cells - an integrated concept of malignant tumour progression
.
Nat Rev Cancer
2005
;
5
:
744
9
.
34.
Kalluri
R
,
Weinberg
RA
. 
The basics of epithelial-mesenchymal transition
.
J Clin Invest
2009
;
119
:
1420
8
.
35.
Burness
ML
,
Grushko
TA
,
Olopade
OI
. 
Epidermal growth factor receptor in triple-negative and basal-like breast cancer: promising clinical target or only a marker?
Cancer J
2010
;
16
:
23
32
.
36.
Guerin
M
,
Gabillot
M
,
Mathieu
MC
,
Travagli
JP
,
Spielmann
M
,
Andrieu
N
, et al
Structure and expression of c-erbB-2 and EGF receptor genes in inflammatory and non-inflammatory breast cancer: prognostic significance
.
Int J Cancer
1989
;
43
:
201
8
.
37.
Nakai
K
,
Hung
MC
,
Yamaguchi
H
. 
A perspective on anti-EGFR therapies targeting triple-negative breast cancer
.
Am J Cancer Res
2016
;
6
:
1609
23
.
38.
Eccles
SA
. 
The epidermal growth factor receptor/Erb-B/HER family in normal and malignant breast biology
.
Int J Dev Biol
2011
;
55
:
685
96
.
39.
Hong
A
,
Dobbins
T
,
Lee
CS
,
Jones
D
,
Jackson
E
,
Clark
J
, et al
Relationships between epidermal growth factor receptor expression and human papillomavirus status as markers of prognosis in oropharyngeal cancer
.
Eur J Cancer
2010
;
46
:
2088
96
.
40.
Lurje
G
,
Lenz
HJ
. 
EGFR signaling and drug discovery
.
Oncology
2009
;
77
:
400
10
.
41.
Salomon
DS
,
Brandt
R
,
Ciardiello
F
,
Normanno
N
. 
Epidermal growth factor-related peptides and their receptors in human malignancies
.
Crit Rev Oncol Hematol
1995
;
19
:
183
232
.
42.
Xu
X
,
Liu
Z
,
Zhou
L
,
Xie
H
,
Cheng
J
,
Ling
Q
, et al
Characterization of genome-wide TFCP2 targets in hepatocellular carcinoma: implication of targets FN1 and TJP1 in metastasis
.
J Exp Clin Cancer Res
2015
;
34
:
6
.
43.
Yuedi
D
,
Yuankun
C
,
Jiaying
Z
,
Han
L
,
Yueqi
W
,
Houbao
L
, et al
TFCP2 activates beta-catenin/TCF signaling in the progression of pancreatic cancer
.
Oncotarget
2017
;
8
:
70538
49
.