Epithelial-to-mesenchymal transition (EMT)-inducing transcription factors (TF) are well known for their ability to induce mesenchymal states associated with increased migratory and invasive properties. Unexpectedly, nuclear expression of the EMT-TF ZEB2 in human primary melanoma has been shown to correlate with reduced invasion. We report here that ZEB2 is required for outgrowth for primary melanomas and metastases at secondary sites. Ablation of Zeb2 hampered outgrowth of primary melanomas in vivo, whereas ectopic expression enhanced proliferation and growth at both primary and secondary sites. Gain of Zeb2 expression in pulmonary-residing melanoma cells promoted the development of macroscopic lesions. In vivo fate mapping made clear that melanoma cells undergo a conversion in state where ZEB2 expression is replaced by ZEB1 expression associated with gain of an invasive phenotype. These findings suggest that reversible switching of the ZEB2/ZEB1 ratio enhances melanoma metastatic dissemination.

Significance:

ZEB2 function exerts opposing behaviors in melanoma by promoting proliferation and expansion and conversely inhibiting invasiveness, which could be of future clinical relevance.

Cutaneous melanoma is the most aggressive form of skin cancers and remains a major clinical challenge. Malignant transformation of melanocytes is often associated with constitutive MAPK signaling, mostly driven by activating mutations in BRAF or NRAS, and inactivation of tumor suppressors such as PTEN, CDKN2A, or TP53. Melanoma is a highly heterogeneous tumor, composed of phenotypically distinct subpopulations (1). While the mechanisms that contribute to intratumor heterogeneity remain poorly understood, it is without a doubt a major contributor to both intrinsic and acquired therapy resistance and metastatic dissemination. Identification of mediators of intratumor heterogeneity may therefore lead to new therapeutic avenues that exhibit longer-lasting antitumor responses, prevent metastatic dissemination and identification of prognostic markers for therapy stratification.

Melanoma progression and therapy resistance does not rely solely on mutation-driven mechanisms. There is increasing evidence that nongenetic mechanisms that modulate the epigenetic landscape and/or transcriptional and metabolic state, also contribute to progression and drug resistance (2). It is increasingly recognized that phenotypic heterogeneity stems, at least partly, from the ability of melanoma cells to switch back and forth between a proliferative cell state and a mesenchymal-like, invasive state. Both cell-intrinsic and -extrinsic (i.e., microenvironmental) factors such as oxygen, cytokines, and growth factors modulate this reversible phenotype switch. Over recent years, the reported reversible mechanisms of melanoma invasion have supported this model (3–7). Phenotype switching is reminiscent to epithelial-to-mesenchymal transition (EMT). This phenotype switching event was also shown to promote therapy resistance and is a key driver of metastatic spreading. In epithelial tissues, EMT-inducing transcription factors, such as SNAIL and ZEB family members, in addition to promote invasion and metastasis may also contribute to de novo carcinogenesis by bypassing cellular senescence and expanding the cancer stem cell pool. The functional role of EMT drivers has thus been expanded beyond their ability to induce a mesenchymal transcriptional program. The regulation and role of EMT-inducing transcription factors in nonepithelial contexts such as melanocytes and melanoma may even be more diverse and pleiotropic. We have, for instance, shown that ZEB2 is an important driver of normal melanocyte development and differentiation and established correlations between nuclear ZEB2 expression in the primary melanoma tumor and recurrence-free survival (8). We also showed that ZEB2 positively regulate MITF levels and/or activity during melanocyte homeostasis. These data raised the possibility that ZEB2 may contribute to melanoma plasticity and, as such, modulate melanoma growth and progression and metastatic dissemination in a manner that is unconventional for a EMT-TF. To test this possibility, we examined the contribution of ZEB2 to melanoma development and metastatic spreading. Our data establish a critical role for ZEB2 as a key modulator of melanoma heterogeneity and cell population dynamics in vivo.

Human tissues

Tissue samples were obtained from the Department of Dermatology, Universitair Ziekenhuis Gent (Ghent, Belgium), from the Department of Pathology, University Hospital Leuven, KU Leuven, Belgium and from archival paraffin-embedded patient samples from St. Vincent's University Hospital (Dublin, Ireland). All patient specimens were used in accordance with institutional and national policies at the respective locations, with appropriate approval provided by the relevant Ethics committees at the respective institutions. All patient-related information was anonymized.

Cell lines

M-series primary melanoma cell cultures have been established from surplus material from cutaneous melanoma metastases. Written informed consent was approved by the local institutional review board (IRB; EK647 and EK800). Melanoma cells were brought into culture as described previously (9). SK-MEL28 and 501MEL were obtained from ATCC. TGFβ treatment of cell lines M000921 and M010817 was performed with recombinant hTGFβ, 5 ng/mL (PeproTech, 100-21). All cell lines were screened periodically for Mycoplasma contamination.

Mice

Mice were kept in accordance with the institutional guidelines regarding the care and use of laboratory animals and all procedures and experiments were approved by the institutional review ethics committee. Mice with the following alleles have been described elsewhere: conditional Zeb2fl/fl, Rosa-YFP, Rosa-Zeb2TG/TG-IRES-GFP, Tyr::CreERT2 and Tyr:NRAS p53 (10–13). Conditionally, Rosa-Zeb2TG/TG-IRES-GFP–overexpressing mice were generated using G4 hybrid ES cells and the Gateway-compatible Rosa26 locus targeting vector, as described previously (14). For TyrCreERT2 induction, the dorsal hair of anesthetized mice was removed with a (50:50 w/w) mix of beeswax and gum rosin (Sigma-Aldrich, 243221 and 60895-250G). 4-hydroxytamoxifen (4-OHT; 70% Z-isomer, Sigma Sigma-Aldrich, H-6278) was dissolved at 50 mg/mL in DMSO at 37°C under gentle agitation. 4-OHT was further diluted at 5 mg/mL in ethanol and 5 μL of the 4-OHT solution was applied using a pipette on the depilated area directly. All Tyr::CreERT2 animals including control animals received 4-OHT treatment. Mice were examined at a regular basis and sacrificed at an endpoint defined by adverse clinical symptoms, such as weight loss (>15%), a hunched posture or multiple skin tumors (diameter > 5 mm). Quantification of the percentage of mice bearing macro-metastases in each organ was visually examined under the binocular at necropsy.

Histology and IHC

Tumors, organs, and skin were isolated and fixed overnight in 4% paraformaldehyde solution, dehydrated, embedded in paraffin and cut into 5-μm sections. For histology, samples were stained with hematoxylin and eosin. For IHC staining, antigen retrieval was done in citrate buffer and endogenous peroxidases were blocked with 3% H2O2 in methanol. The sections were incubated with primary antibodies and stained with biotin-conjugated secondary antibodies followed by Streptavidin-HRP based development (substrate development with DAB). When necessary, the signal was amplified using the Tyramide Signal Amplification (TSA) kit (Perkin Elmer). TMA microarray slides containing 178 primary melanoma samples were digitally scanned on an Aperio ScanScope scanner and analyzed using Aperio algorithms. Correlation between the IHC data was compared with clinical parameters using the two-tailed Fisher exact test and the χ2 test. Immunostaining against ZEB1 and ZEB2 on mouse sections was performed using in-house generated rabbit and mouse mAb, respectively, GFP was detected using a rabbit monoclonal antibody (clone D5.1 XP Rabbit mAb #2956, Cell Signaling Technology). Rabbit anti-ZEB2 (HPA003456, Sigma) and rabbit anti-ZEB1 (Clone H-102, sc-25388, Santa Cruz Biotechnology) was used for human sections.

Lentiviral transductions

For generation of melanoma cells overexpressing ZEB1, virus production was performed in HEK293T cells using calcium phosphate transfection with pMD2.G (envelope plasmid), psPAX2 (packaging plasmid), and pSIN-TRE-GW-3xHA ZEB1. Transduced cells were selected by puromycin (2 μg/mL for all cell lines except 501MEL and SK-MEL28: 1 μg/mL). The shRNA vectors are pLKO1.5-based from Sigma, Mission shRNA series.

siRNA and plasmid transfections

For RNAi, we used siRNA pools (Dharmacon) and a scrambled siRNA pool (Dharmacon) as a control. Transfections were performed with HiPerfect (Qiagen) according to the manufacturer's instructions (siZeb1: Dharmacon, M-051513-01-0005; siZeb2: M-059671-01-0005; siZEB1: M-006564-02-0005; siZEB2: M-006914-00-0005). Plasmids carrying myc-tagged ZEB2, HA-tagged MITF, or empty cDNA expression cassettes were transiently transfected. 501MEL cells were transfected using FugeneHD (Promega E-231): cells were seeded in a 6-well to reach 75% confluency prior to the transfection in 3 mL growth medium with a 4:1 ratio DNA:Lipofectant. Subsequently, for each well, 3.3 μg total DNA was diluted in 160 μL OptiMEM, 13 μL Fugene HD reagent was added, and vortexed briefly, and the solution was incubated at room temperature during 5 minutes, and 150 μL of the mixture was added dropwise on the cells (in 3 mL growth medium). MITF-VP16 chimera, a transcriptionally more active MITF-derivative, and MITF cDNA expression constructs were kindly provided by Prof. Vachtenheim (Department of Transcription and Cell Signaling, Charles University, Prague, Czech Republic).

Electric cell surface impedance sensing

8W10E array plates (Applied Biophysics) were washed and treated with 10 mmol/L cysteine for 30 minutes at room temperature. Next, for HUVEC assays, array plates were coated with laminin before cell seeding. HUVECs (Lonza) were seeded and challenged with melanoma cells once stable impedance (and visible confluency of the HUVECs) was achieved. As a control, heat-inactivated cells (10 minutes at 55°C) in conditioned medium were included. Next, arrays were mounted onto the array holder connected to the electric cell surface impedance sensing (ECIS) module and placed in a standard incubator at 37°C and 5% CO2 throughout the experiment. All impedance measurements were performed in the multifrequency measurement mode.

RNA isolation

RNA was extracted from cell cultures by using RNeasy extraction columns (Qiagen) or TRIsure lysis buffer (BioLine) according to the manufacturer's guidelines. For tumors, after collecting, the tissue is subsequently disinfected in a povidone–iodine solution (iso-Betadine) and washed twice in 70% ethanol and three times in PBS. Skin preparations were carefully dissected, followed by removal of fat tissue using a scalpel. Tissue pieces were incubated in RNAlater (Ambion, AM7021; BioLine, BIO-38032) at 4°C overnight. Subsequently, the pieces were put in a petridish in 1 mL TRISure on ice and cut into small pieces. Subsequently, the pieces were homogenized using a mechanical mixer followed by passing the sample five times through a 21-gauge needle. To reduce the carryover of RNases during further processing, the homogenized sample is centrifuged for 15 minutes, 20,000 × g, 4°C to eliminate debris. The supernatant is transferred to a new RNAse-free Eppendorf tube and incubated for 5 minutes at room temperature. After phase separation by adding 200 μL chloroform and vigorous shaking, 250 μL from the aqueous upper phase was added to 500 μL cold RNase-free ethanol. Samples were snap-frozen in liquid nitrogen and stored at −80°C.

qRT-PCR

RNA was treated with 1 U of RNAse-free DNase RQ1 (Promega) per μg RNA for 30 minutes at 37°C in appropriate buffer. DNAse was inactivated by incubation in Promega stop solution for 10 minutes at 65°C. Bulk Mg2+ was removed by using Amicon ultra 0.5-mL centrifugal filters (Millipore, UFC510096) in two consecutive diluting washes. cDNA synthesis was performed with iScript advanced cDNA synthesis kit (Bio-Rad, 172-5038) following the manufacturer's instructions. Quantitative PCR was done using the SYBR Master Mix Kit SensiFast SYBR No-Rox Kit (Bioline, CSA-01190) for the genes of interest and reference genes. Plates were run on the LightCycler 480 (Roche). The average threshold cycle of triplicate reactions was used for all subsequent calculations using the ΔCt method, calculated in qBasePLUS (BioGazelle). The following reference genes were used for a first read-out: Calm2, Matr3, Hmbs, Rpl13a, Oaz1, Gapdh, Ywhaz, Tbp, Cox4i1, and Eef1a for mouse, EEF1A1, DCAF6, OAZZ1, CALM2, MATR3, COX4I1, RPL37A, FTL, SH3KBP1, TPT1 for human. Subsequently, the most stable reference (housekeeping) genes from this set were determined via qBase+ (geNorm) prior to repeating the experiment.

RNA sequencing

Melanoma cell cultures were established from patient biopsies. Patients gave their informed consent and was approved by the local IRB (EK647 and EK800). RNA from the melanoma cell cultures were isolated with the QIAGEN RNeasy Kit. RNA capture was performed with TruSeq RNA Library Prep Kit v2 (Illumina) and sequenced on a HiSeq4000. RNA counts were quantified from single-end reads using STAR aligner (15). Normalization of RNA counts was performed with edgeR (16). RNA sequencing (RNA-seq) data of TGFβ treatment human melanoma cell lines available at GEO database (GSE148767).

Single-cell RNA-seq analyses

ZEB1 and ZEB2 expression values (TPMs) were inferred from single-cell RNA-sequencing analyses (scRNA-seq) data (GSE115978) from human malignant melanoma cells and log-normalized expression values represented for n = 1,951 cells.

Seurat R package (version 3.1.3) was used to log normalize the data, perform dimensionality reduction analysis, clustering of the cells and performing differential gene expression. We extracted the melanoma cells that were annotated by Jerby-Arnon and colleagues (17). However, we deleted a small cluster of contaminating CD3E+ CD45+ T cells that were initially annotated as melanoma cells in the original dataset. 526 of 1,951 cells were double negative for ZEB1 and ZEB2. Remaining cells came from 23 patients. 1,373 cells showed ZEB2 and 238 cells ZEB1 expression. ZEB1 and ZEB2 expression per single cells are significantly anticorrelated, as shown by Spearman correlation (ρ = −0.28; P < 2.2E-16).

SRB proliferation assay

The proliferative capacity of melanoma cells was analyzed by performing the Sulforhodamine B (SRB) proliferation assay. Briefly, cells were seeded at 3,000 to 7,000 cells per 96-well and at day 0 and 4, the cells were fixed with 30% (wt/vol) trichloroacetic acid for 1 hour, and stained for 30 minutes at room temperature with 0.4% SRB in 1% acetic acid. Excessive dye was washed off with 1% acetic acid and dissolved in 10 mmol/L Tris buffer (pH 10.5). The plates were measured with a microplate reader (Bio-Rad, Eke, Belgium) at 570 nm. Day 0 was used as a reference sample.

Western blot analysis

Tissues were mechanically separated from the dermis and ground into small pieces prior to lysis, culture cells were immediately lysed in Laemmli-lysis buffer (50 mmol/LTris-HCl pH 6.8, 10% glycerol, 2% SDS) using standard practices. After sonicating and centrifuging the samples, 20 μg of protein was separated on a acrylamide gel and transferred to a polyvinylidene difluoride membrane. Membranes were incubated with primary antibodies and appropriate HRP-labeled secondary antibodies (GE Healthcare). Primary antibodies were diluted in 3% milk. Detection was performed with the Western Lightning Chemiluminescence Reagent Plus Kit (PerkinElmer) or the Immobilion Western HRP Substrate (Millipore).

Colony formation in soft agar

Petri dishes (60-mm diameter) were coated with 0.5% Bacto agar (Difco) in growth medium. A suspension of 1,104 cells in growth medium containing 0.35% Noble agar (Difco) was seeded on top of the agar coating and then covered with growth medium. Fresh medium was applied weekly. Cell colonies were allowed to form over a time period of 55 days, after which, they were stained for 1 hour with 0.005% crystal violet (Sigma) in PBS and photographed.

Invasion and migration assay

An QCM ECMatrix 24-well (8 μm) Fluorimetric Cell Invasion Assay kit (Chemicon; Sigma-Aldrich) was used according to the manufacturer's protocol. An insert polycarbonate membrane with a pore size of 8 μm) was coated with a thin layer of ECMatrix. Cells were seeded in the insert (top chamber) at a density of 25 × 103 cells/well in serum-free RPMI. RPMI medium supplemented with 10% FCS was added to the bottom chamber. Following 24-hour incubation, invading cell numbers were determined via fluorescent read-out. For migration assays, similar inserts were not coated with ECMatrix.

Statistical analyses

Results were evaluated using the statistical tests and indicated with P values in the figure legends. P < 0.05 was considered statistically significant. Survival curves and metastasis-free survival were analyzed by Kaplan–Meier analysis, which were compared by log-rank (Mantel–Cox–Kaplan Meyer Plot). GraphPad Prism 7 was used to perform log-rank (Mantel–Cox), Student t test, two-way ANOVA followed by post-hoc Tukey HSD (SRB assays). R (http://www.cran.r-project.org) was used to perform Fisher exact test and the χ2 test (for TMA staining), Benjamini–Hochberg Multiple Testing Correction (adjusted) P value (for bulk RNA-seq data). Differential gene expression during single-cell analysis was performed via a nonparameteric Wilcoxon rank sum test in Seurat.

Zeb2 is required for melanoma growth and development

ZEB2 expression in human melanoma is heterogenous (Fig. 1A). Such a heterogeneous pattern was also observed in lesions from the previously established NRASQ61K-driven mouse model of melanoma (Fig. 1B). Note that NRAS is consistently altered in 20% of melanomas and is the second most common oncogenic driver mutation (18). Constitutive melanocyte-specific expression of one copy of the activated form of human NRAS (NRASQ61K) triggers melanoma development in a small fraction of Tyr::NRASQ61K mice. The mouse skin is hyperpigmented and reminiscent of patients with a giant congenital nevus (GCN). These nevi are large pigmented lesions that frequently progress to melanoma. On average 15%–20% of Tyr::NRASQ61K mice develop melanomas within 1 year in the absence of secondary genetic alterations (11). Concomitant loss of p53 or p16INK4A functionally increases the incidence to nearly 100% and shortens the latency significantly (11). The biological significance of the varying ZEB2 levels remains unclear. We hypothesized that graded levels of ZEB2 may associate with distinct melanoma phenotypic properties. To test this possibility, we engineered a conditional ZEB2 allelic series onto the NRASQ61K-driven mouse melanoma background (Fig. 1C). The Tyr::NRASQ61K;TyrCreERT2;Trp53lox/lox mice (henceforth NRASQ61K p53−/− mice) were intercrossed with mice carrying a Zeb2 conditional transgenic allele, Rosa26-(loxP-STOP-loxP)-Zeb2. Note that the Zeb2 open reading frame is followed by an IRES-EGFP reporter sequence, GFP expression can therefore be used to fate map transgenic cells. Mice carrying two copies of this allele are thereafter referred to as Rosa-Zeb2. In addition, the Tyr::NRASQ61K;TyrCreERT2;Trp53lox/lox mice have also been intercrossed with mice carrying a conditional Zeb2 knockout allele (Zeb2fl/fl; refs. 10, 14). To modulate ZEB2 expression the compound mice were exposed to topical application of 4-hydroxytamoxifen (4-OHT) onto their back skin. Nevus formation and melanoma development was monitored weekly.

Figure 1.

ZEB2 is heterogeneously expressed in melanoma. A, ZEB2-stained human melanoma samples exhibit heterogeneity in ZEB2 expression. Top and bottom selected rectangles depict areas with respectively strong and weak nuclear ZEB2 expression. Micrograph images were taken with ×4/0.1 and ×20/0.4 objective. B, ZEB2-stained sections from Tyr::NRASQ61K and Tyr::NRASQ61K p53fl/fl mouse melanoma models. C, Overview of mouse models with graded Zeb2 levels on a Tyr::NRASQ61K p53fl/fl-targeted background.

Figure 1.

ZEB2 is heterogeneously expressed in melanoma. A, ZEB2-stained human melanoma samples exhibit heterogeneity in ZEB2 expression. Top and bottom selected rectangles depict areas with respectively strong and weak nuclear ZEB2 expression. Micrograph images were taken with ×4/0.1 and ×20/0.4 objective. B, ZEB2-stained sections from Tyr::NRASQ61K and Tyr::NRASQ61K p53fl/fl mouse melanoma models. C, Overview of mouse models with graded Zeb2 levels on a Tyr::NRASQ61K p53fl/fl-targeted background.

Close modal

Strikingly, Zeb2 ablation dramatically attenuated nevus and melanoma outgrowth (Fig. 2A and B). Moreover, targeting Zeb2 in a NRASQ61K p53−/− background delayed melanoma initiation (Fig. 2C). Interestingly, all analyzed pigmented tumors that arose in NRASQ61K p53−/− Zeb2fl/fl mice contained only Zeb2 wild-type melanoma cells, indicating that these lesions originated from melanoma cells that escaped Zeb2 ablation (Supplementary Fig. S1A and S1B). As such, no genuine Zeb2-knockout melanoma tumors are formed. These results indicate that ZEB2 is required for the growth and expansion of primary NRAS-driven melanoma. In contrast, ectopic ZEB2 overexpression accelerated melanoma initiation, decreased the latency and increased melanoma penetrance. ZEB2 expression did not specifically affect metastatic spread on a p53-null–mutated background.

Figure 2.

ZEB2 is required for melanoma development and growth on a p53-null background. A, Morphologic appearance of skins of mice with 32 weeks of age. B, H&E staining of skin section at defined time points posttreatment. C, Kaplan–Meier tumor-free survival curves for Tyr::NRASQ61K p53fl/fl Rosa26-Zeb2 (n = 13) or Tyr::NRASQ61K p53fl/fl Zeb2fl/fl mice (n = 15) and their wild-type controls (respectively, n = 17 and n = 21). Tumor-free percentages were compared using the log-rank (Mantel–Cox) test.

Figure 2.

ZEB2 is required for melanoma development and growth on a p53-null background. A, Morphologic appearance of skins of mice with 32 weeks of age. B, H&E staining of skin section at defined time points posttreatment. C, Kaplan–Meier tumor-free survival curves for Tyr::NRASQ61K p53fl/fl Rosa26-Zeb2 (n = 13) or Tyr::NRASQ61K p53fl/fl Zeb2fl/fl mice (n = 15) and their wild-type controls (respectively, n = 17 and n = 21). Tumor-free percentages were compared using the log-rank (Mantel–Cox) test.

Close modal

Zeb2 drives a melanoma “proliferative” transcriptional program

To assess whether ZEB2 can promote melanomagenesis in the presence of functional p53, we conditionally overexpressed ZEB2 in a Tyr::NRASQ61K (p53 wild-type) background. Ectopic expression of transgenic ZEB2 and GFP was confirmed via immunoblot analysis (Supplementary Fig. S2A). Forced expression of ZEB2 in Tyr::NRASQ61K Rosa26-Zeb2 mice (NRASQ61K Rosa-Zeb2) did not significantly affect tumor latency or incidence (Supplementary Fig. S2B). Ectopic ZEB2 expression is therefore not sufficient to transform NRASQ61K–expressing melanocytes. Molecular analysis of NRASQ61K and NRASQ61K Rosa-Zeb2 tumor cells showed an increased expression in genes that belong to the previously described melanoma “proliferative” gene signature including Mitf, Mc1r, and Sox10 (19, 20). This increase occurred concomitantly to a decrease in expression of Zeb1 expression (Fig. 3A). RNAi-mediated Zeb2 knockdown in melanoma cell lines Mel3 and Mel6 derived from the Rosa-Zeb2 melanoma mouse model attenuated and reversed this gene signature, as illustrated by a decrease in Mitf levels (Fig. 3B). This was accompanied by an increase in Zeb1 levels. As soon as cells regained Zeb2 expression (6 days after transfection of the siRNA molecules), the “proliferative” gene signature reemerged, highlighting the reversibility and dynamic nature of the ZEB2-driven transcriptional program (Fig. 3C). Importantly, Zeb2 depletion led to a decrease in the proliferation potential of primary melanoma cell lines derived from 5 different NRASQ61K Rosa-Zeb2 mice and the B16 melanoma cell line (Supplementary Fig. S3A) and induces a melanoma “invasive/mesenchymal-like” gene expression in B16 cells (Supplementary Fig. S3B; refs. 19, 20). Zeb2 depletion, Mitf depletion or ectopic Zeb1 expression in B16 cells promoted an invasive and migratory phenotype in vitro (Supplementary Fig. S3C and S3D). A transendothelial migration assay as described by Tarantola and colleagues (21) and Keese and colleagues (22) showed how ZEB1 promotes melanoma cells to attach to and cause retraction of the endothelial monolayer (Supplementary Fig. S3E). We validated our conclusions in a panel of human melanoma cell lines. Lu1205 melanoma cells show increased or attenuated invasive capacity upon respectively enhanced or reduced ZEB1 expression (Supplementary Fig. S3F). Upon overexpression of ZEB1 in the human 501MEL melanoma cell line, we readily observed a dramatic growth attenuation (Supplementary Fig. S3G). Three days following doxycycline administration, the 501MEL iZEB1 cell line altered predominantly genes associated with the proliferative phenotype, whereas the invasive gene repertoire is not induced yet to the fullest. ZEB1 induction shifts the cells toward an invasive gene signature after 6 days, similar to ZEB2 depletion (Supplementary Fig. S3G). ZEB1-mediated depletion of MITF protein levels and increased migratory capacities were confirmed in the differentiated melanoma cell lines 501MEL and SK-MEL28 (Supplementary Fig. S3H and S3I).

Figure 3.

ZEB2 promotes reversible expression of differentiation markers. A, Relative mRNA expression levels for Zeb2, GFP, Zeb1, Sox10, Mitf, and Mc1r in melanoma samples. Data were compared by using unpaired Student t test and are presented as means ± 95% SD. B, Relative mRNA expression of phenotype-specific genes 96 hours after siRNA treatment against Zeb2 in two cell lines generated from two different Tyr::NRASQ61K Rosa26-Zeb2 mouse melanomas. C, Relative mRNA expression of phenotype-specific genes in cell line Mel6 after siRNA withdrawal and a Zeb2 recovery phase of 6 days after knockdown. Data were compared by using Student t test and are presented as averages + SD. D, IHC analysis for GFP, ZEB2, and ZEB1 in primary and metastatic melanoma samples from Tyr::NRASQ61K Rosa26-Zeb2 mice to evaluate heterogeneity of endogenous and exogenous Zeb2 expression. *, P < 0.05; **, P < 0.005; ***, P < 0.0005; ns, nonsignificant.

Figure 3.

ZEB2 promotes reversible expression of differentiation markers. A, Relative mRNA expression levels for Zeb2, GFP, Zeb1, Sox10, Mitf, and Mc1r in melanoma samples. Data were compared by using unpaired Student t test and are presented as means ± 95% SD. B, Relative mRNA expression of phenotype-specific genes 96 hours after siRNA treatment against Zeb2 in two cell lines generated from two different Tyr::NRASQ61K Rosa26-Zeb2 mouse melanomas. C, Relative mRNA expression of phenotype-specific genes in cell line Mel6 after siRNA withdrawal and a Zeb2 recovery phase of 6 days after knockdown. Data were compared by using Student t test and are presented as averages + SD. D, IHC analysis for GFP, ZEB2, and ZEB1 in primary and metastatic melanoma samples from Tyr::NRASQ61K Rosa26-Zeb2 mice to evaluate heterogeneity of endogenous and exogenous Zeb2 expression. *, P < 0.05; **, P < 0.005; ***, P < 0.0005; ns, nonsignificant.

Close modal

We next assessed whether the ectopically induced Zeb2 transcriptional program could be spontaneously reversed in the in vivo setting. We took advantage of the GFP-reporter gene expression to fate map the Zeb2-transgenic cells in primary NRASQ61K Rosa-Zeb2 melanoma (Fig. 3D). Remarkably, an attenuation of ZEB2 transgenic protein levels was observed in a fraction of the melanoma cells thus recapitulating the naturally occurring cellular heterogeneity observed in the NRASQ61K wild-type Zeb2 melanoma lesions. Because, the Zeb2 transgene does not contain untranslated regions, we concluded that silencing of its expression is likely due to a (post)translational regulatory event. Interestingly, tumor areas that had lost Zeb2-transgenic expression became strongly positive for Zeb1. This was in keeping with the above in vitro and with our previous observation that Zeb2-deficient melanocytes acquire a dedifferentiated stem cell phenotype exhibiting elevated Zeb1 levels (8).

Zeb2 promotes the growth from micro- to macrometastasis

Complete necropsy of NRASQ61K and NRASQ61K Rosa26-Zeb2 tumor-bearing mice revealed the presence of large pigmented pulmonary metastases that had colonized the lung parenchyma, the liver, and muscle tissue in a large fraction of Zeb2 transgenic mice (n = 26), whereas no macrometastases could be observed in the control animals (n = 19; Fig. 4A). In the lung parenchyma of NRASQ61K control animals, we did observe small clusters of melanoma cells, only visible through microscopic analysis of hematoxylin and eosin (H&E) sections. Although such pulmonary micrometastases were detected in 88% of the control animals, none of them was able to succesfully grow to a visible metastatic lesion seen with the naked eye (Fig. 4B). Such micrometastases were not observed in other organs. From the NRASQ61K Rosa26-Zeb2 mice, 46% of the mice did have many pulmonary macrometastases (>50 lesions), in the remaining 54%, only micrometastases were detected through the analyses of H&E sections (Fig. 4C). Collectively, these findings indicated that ectopic Zeb2 expression facilitates the outgrowth of “dormant” disseminated melanoma cells and promote the formation of successful metastases.

Figure 4.

Forced Zeb2 expression enhances metastatic outgrowth on a Tyr::NRASQ61K–targeted background. A, Frequency of metastases (one or more) detected in Tyr::NRASQ61K p53fl/fl Zeb2fl/fl mice and their wild-type controls. B, Photographs of liver, lung, and muscle tissue of control mice and metastasis-bearing Tyr::NRASQ61K p53fl/fl Zeb2fl/fl mice. C, Micrograph of lungs of Tyr::NRASQ61K p53fl/fl Zeb2fl/fl mice and their wild-type controls (left) and H&E sections (right).

Figure 4.

Forced Zeb2 expression enhances metastatic outgrowth on a Tyr::NRASQ61K–targeted background. A, Frequency of metastases (one or more) detected in Tyr::NRASQ61K p53fl/fl Zeb2fl/fl mice and their wild-type controls. B, Photographs of liver, lung, and muscle tissue of control mice and metastasis-bearing Tyr::NRASQ61K p53fl/fl Zeb2fl/fl mice. C, Micrograph of lungs of Tyr::NRASQ61K p53fl/fl Zeb2fl/fl mice and their wild-type controls (left) and H&E sections (right).

Close modal

ZEB2 modulates the proliferative–invasive melanoma phenotypic switching

RNA-seq analysis of a large cohort of human primary melanoma cultures revealed a strong inverse correlation between expression levels of ZEB2, MITF, and the MITF-target gene TYR, and an inverse correlation between MITF and ZEB1 expression. ZEB1 expression was correlated with AXL, WNT5A, TCF4, CDH2, and FN1 expression (Fig. 5A). These correlations were confirmed via qRT-PCR on a selection of cultures and a panel of human melanoma cell lines (Fig. 5B). ZEB2-positive melanomas exhibit a differentiated and proliferative gene signature, whereas ZEB1 melanomas express the invasive gene signature, indicating that melanoma switch between a proliferative MITFhighZEB2highZEB1low state and an invasive and metastatic ZEB1highZEB2lowMITFlow state. Upon RNAi-mediated silencing of ZEB2, using two independent shRNAs (sh1 and sh2 ZEB2), SK-MEL28 melanoma cells and 501MEL cells loose MITF expression and exhibit, a severe growth retardation, while gaining expression of invasive markers including ZEB1 (Fig. 5C and D; Supplementary Fig. S4A and S4B). ZEB2 depletion also hampers the ability of these melanoma cells to form colonies in vitro (Fig. 5E; Supplementary Fig. S4C). Upon restoration of Zeb2 expression following doxycycline withdrawal, the cells recovered a proliferative phenotype, further confirming the reversibility of the phenotype switch (Fig. 5E). RNAi-mediated depletion of ZEB2 or MITF in 501MEL cells, results in a strong proliferation defect (Supplementary Fig. S4D). The decreased proliferation in siZEB2-treated 501MEL cells, but not siMITF-treated cells, can partially be rescued by siZEB1 treatment (Supplementary Fig. S4D and S4E). Reexpression of the MITF-VP16 chimera, a transcriptionally more active MITF-derivative is sufficient to restore growth in ZEB2/MITF-depleted cells. In contrast, ZEB2 reexpression is not able to rescue growth inhibition upon MITF depletion. This supports the model that ZEB2 is required for MITF functionality and suppression of the alternative ZEB1 state but is unable to compensate for MITF loss, while ZEB1 may act downstream of the ZEB2/MITF interplay and contributes to the phenotype switch when ZEB2/MITF functionality is compromised. Taken together, our data identified ZEB2 as a key modulator of the proliferative to invasive phenotype switch both in mouse and human melanoma cells. To further substantiate these findings, we examined the correlation between ZEB2 expression and Breslow Depth of Invasion index. This is used as a standard prognostic factor in melanoma pathology measuring how deep the tumor cells have invaded the dermis and the surrounding tissue in human melanoma biopsies. A robust association between intense nuclear ZEB2 immunoreactivity and lower Breslow score was identified, thus confirming that loss of ZEB2 in primary melanoma is associated with increased invasiveness in human patients (Fig. 6A and B). Moreover, IHC expression analyses on human melanoma samples made clear that ZEB2 is attenuated and ZEB1 nuclear expression is enhanced in the invasive front at the deepest margins of the tumor, strongly associating ZEB1 with the invasive behavior of human primary melanoma cells (Fig. 6C). To assess whether the ZEB1- and ZEB2-expressing cells are distinct subpopulations in human melanomas, we analyzed single-cell expression data of human melanoma (GSE115978; ref. 17). This analysis made clear that the majority of the melanoma cells (1,951 cells coming from 33 patients analysed by scRNA-seq) are ZEB2high ZEB1low and that ZEB2 and ZEB1 are clearly anticorrelated as shown by Spearman correlation (ρ = −0.28, P < 2.2E-16; Fig. 6D). Interestingly, ZEB2low ZEB1high cells have a clear loss of proliferation/differentation markers such as MITF, TYR, PMEL, and SOX10 and upregulation of invasion markers such as COL1A2, TPM2, ALDH1A3, and TCF4 (Fig. 6E).

Figure 5.

ZEB2 affects the phenotypical switch between proliferation and invasion in human melanoma A, Log2 (fold change) expression values determined via RNA-seq analysis of a large cohort of human primary melanoma cultures. B, Log2 (fold change) expression values of phenotype-specific genes in short-term melanoma cultures (passage < 5; left) or melanoma cell lines (right). Differential expression determined via qRT-PCR and normalized to the median of the expression levels and represented in a heatmap. C, Immunoblot analysis of ZEB2, MITF, and ZEB1 in SK-MEL28 cells transduced with lentiviral vectors expressing doxycycline-inducible shRNA directed against ZEB2. D, Relative proliferation after ZEB2 knockdown in SK-MEL28 melanoma cells measured via SRB assay. Data are represented as mean + SEM and compared using two-way ANOVA, post hoc Tukey HSD. E, Left, images of colony formation in doxycycline-induced SK-MEL28 cells. Cells were untreated (−DOX) or pretreated (+DOX) with doxycycline and grown in soft agar in the absence (−DOX) or presence (+DOX) of doxycycline. After 15 days, cell colonies were imaged using an inverted microscope. Right, quantification of colony formation of cells pretreated with doxycycline for 7 or 14 days, grown in soft agar in the presence (on DOX) of absence (withdrawal) of doxycycline in the culture medium. Data were compared by using unpaired Student t test and are presented as means ± 95% SD. *, P < 0.05; **, P < 0.005; ***, P < 0.0005; ns, not significant.

Figure 5.

ZEB2 affects the phenotypical switch between proliferation and invasion in human melanoma A, Log2 (fold change) expression values determined via RNA-seq analysis of a large cohort of human primary melanoma cultures. B, Log2 (fold change) expression values of phenotype-specific genes in short-term melanoma cultures (passage < 5; left) or melanoma cell lines (right). Differential expression determined via qRT-PCR and normalized to the median of the expression levels and represented in a heatmap. C, Immunoblot analysis of ZEB2, MITF, and ZEB1 in SK-MEL28 cells transduced with lentiviral vectors expressing doxycycline-inducible shRNA directed against ZEB2. D, Relative proliferation after ZEB2 knockdown in SK-MEL28 melanoma cells measured via SRB assay. Data are represented as mean + SEM and compared using two-way ANOVA, post hoc Tukey HSD. E, Left, images of colony formation in doxycycline-induced SK-MEL28 cells. Cells were untreated (−DOX) or pretreated (+DOX) with doxycycline and grown in soft agar in the absence (−DOX) or presence (+DOX) of doxycycline. After 15 days, cell colonies were imaged using an inverted microscope. Right, quantification of colony formation of cells pretreated with doxycycline for 7 or 14 days, grown in soft agar in the presence (on DOX) of absence (withdrawal) of doxycycline in the culture medium. Data were compared by using unpaired Student t test and are presented as means ± 95% SD. *, P < 0.05; **, P < 0.005; ***, P < 0.0005; ns, not significant.

Close modal
Figure 6.

High ZEB2 levels are associated with a recurrence-free survival and lower Breslow index of invasion in human melanoma. A, ZEB2-stained tissue microarray slides containing 178 primary melanoma samples were digitally scanned on an Aperio ScanScope scanner. Digital slides were analysed using Aperio algorithms. ZEB2 protein expression values were determined using an automated image analysis approach (IHC-MARK, Oncomark) designed to quantify IHC stained slides. The correlation between the IHC data (ZEB2 expression below or above average expression) was compared with the Breslow category using the two-tailed Fisher exact test and the χ2 test. B, Representative ZEB2-stained human melanoma samples. Micrograph images were taken with a ×10/0.25 objective. C, ZEB2- and ZEB1-stained human melanoma samples with histologic assessment of the invasive regions and the deepest margin of the tumor. D, ZEB1 and ZEB2 expression values (TPMs) were inferred from scRNA-seq data (GSE115978) from human malignant melanoma cells and log-normalized expression values are represented for n = 1,951 cells. Five-hundred and twenty-six of 1,951 cells were double negative for ZEB1 and ZEB2. Remaining cells came from 23 patients. The 1,373 cells showed ZEB2 expression and 238 cells showed ZEB1 expression. ZEB1 and ZEB2 expression per single cells were significantly anticorrelated, as shown by Spearman correlation (ρ = −0.28), P < 2.2E-16. The histogram next to the axis reflects the number of cells in specific ranges of expression levels for ZEB2 (left) or ZEB1 (bottom). E, Heatmap representing top 100 differentially expressed genes between Zeb1high and Zeb2high cells, performed via a nonparametric Wilcoxon rank sum test.

Figure 6.

High ZEB2 levels are associated with a recurrence-free survival and lower Breslow index of invasion in human melanoma. A, ZEB2-stained tissue microarray slides containing 178 primary melanoma samples were digitally scanned on an Aperio ScanScope scanner. Digital slides were analysed using Aperio algorithms. ZEB2 protein expression values were determined using an automated image analysis approach (IHC-MARK, Oncomark) designed to quantify IHC stained slides. The correlation between the IHC data (ZEB2 expression below or above average expression) was compared with the Breslow category using the two-tailed Fisher exact test and the χ2 test. B, Representative ZEB2-stained human melanoma samples. Micrograph images were taken with a ×10/0.25 objective. C, ZEB2- and ZEB1-stained human melanoma samples with histologic assessment of the invasive regions and the deepest margin of the tumor. D, ZEB1 and ZEB2 expression values (TPMs) were inferred from scRNA-seq data (GSE115978) from human malignant melanoma cells and log-normalized expression values are represented for n = 1,951 cells. Five-hundred and twenty-six of 1,951 cells were double negative for ZEB1 and ZEB2. Remaining cells came from 23 patients. The 1,373 cells showed ZEB2 expression and 238 cells showed ZEB1 expression. ZEB1 and ZEB2 expression per single cells were significantly anticorrelated, as shown by Spearman correlation (ρ = −0.28), P < 2.2E-16. The histogram next to the axis reflects the number of cells in specific ranges of expression levels for ZEB2 (left) or ZEB1 (bottom). E, Heatmap representing top 100 differentially expressed genes between Zeb1high and Zeb2high cells, performed via a nonparametric Wilcoxon rank sum test.

Close modal

TGFβ promotes ZEB switching

TGFβ1 has been shown to promote melanoma switching from a proliferative to an invasive state (23, 24). Accordingly, exposure of two early-passage melanoma cultures (M010817 and M000921), expressing a high ZEB2/ZEB1 ratio to TGFβ1 increased ZEB1 and fibronectin expression, while decreasing MITF levels, both at protein and RNA levels (Fig. 7A and B). This TGFβ1 treatment shifted the gene expression profiles of these cells toward an invasive/mesenchymal-like phenotype, as illustrated by the upregulation of genes such as AXL and WNT5A (Fig. 7A; Supplementary Fig. S5). Interestingly, while ZEB2 mRNA levels were not affected by this treatment in the M010817 cells 48 hours after TGFβ1 exposure, a clear drop in ZEB2 protein levels was observed, suggesting the involvement of a posttranslational mechanism in ZEB2 downregulation. Withdrawal of TGFβ1 after 96 hours, in combination with inhibitor SB431542 (inhibiting kinase activity of the TGFβ1 receptors ALK4, -5 and -7) to block all positive autocrine feedback loops, was able to revert this phenotype after 7 days. These data show that well-established phenotype switching inducer, TGFβ1, is sufficient to revert the ZEB2/ZEB1 ratio, an event that is likely to directly contribute to phenotype switching.

Figure 7.

TGFβ treatment mediates the switch from ZEB2 to ZEB1 and induces an invasive gene signature. A, Relationship of log2 (fold change) RNA-seq expression (base mean) and statistical significance [Benjamini–Hochberg multiple testing correction (adjusted) P value] of differently expressed genes after 96-hour TGFβ treatment with three biological replicates of short-term melanoma cultures from two independent cell lines (M010817 and M000921), represented in volcano plots. B, Immunoblot analysis of several markers involved in ZEB-switching in M010817 and M000921 cells after addition of TGFβ1 to the culture medium and reversion of the phenotype by administration of the TGFβ inhibitor SB431542, inhibiting kinase activity of TGFβ receptors ALK4, -5, and -7.

Figure 7.

TGFβ treatment mediates the switch from ZEB2 to ZEB1 and induces an invasive gene signature. A, Relationship of log2 (fold change) RNA-seq expression (base mean) and statistical significance [Benjamini–Hochberg multiple testing correction (adjusted) P value] of differently expressed genes after 96-hour TGFβ treatment with three biological replicates of short-term melanoma cultures from two independent cell lines (M010817 and M000921), represented in volcano plots. B, Immunoblot analysis of several markers involved in ZEB-switching in M010817 and M000921 cells after addition of TGFβ1 to the culture medium and reversion of the phenotype by administration of the TGFβ inhibitor SB431542, inhibiting kinase activity of TGFβ receptors ALK4, -5, and -7.

Close modal

We previously showed that ZEB2 is a gatekeeper of melanocyte development and differentiation (8). Here, we provide evidence that ZEB2 promotes the growth of both primary and metastatic melanoma lesions, while suppressing ZEB1 expression and thereby an invasive/mesenchymal-like transcriptional program. Genetic ablation of Zeb2 indeed hampered outgrowth of primary melanoma in mice, whereas ectopic expression enhanced melanoma proliferation and the growth of melanoma at both primary and secondary sites. Therefore, Zeb2 cannot be regarded as a genuine tumor suppressor, as suggested previously (25). In support of our findings, ZEB2 was instead recently identified as an “Achilles heel” of melanoma growth in a genome-wide RNAi-based loss-of-function screen aiming at establishing a “cancer dependency map” (26). Here, we show how strong nuclear ZEB2 expression in human primary melanoma was associated with lower Breslow index and increased metastasis-free survival. An increased metastatic burden in mice ectopically expressing Zeb2 has, at first, been difficult to reconcile with these observations. However, we noticed that although presence of increased Zeb2 mRNA levels, ZEB2 protein levels were not elevated in all GFP-tagged transgenic cells, indicating that specific microenvironmental cues were able to destabilize ZEB2 protein expression in a fraction of the cells within the primary tumor.

We hypothesize that the transgenic cells that lose ZEB2 expression are the cells that leave the primary lesions and eventually contribute to the seeding of metastasis-initiating cells in various distant organs. Interestingly, in control Tyr::NRASQ61K mice, only microscopic clusters of tumor cells were observed at distant sites. In contrast, multiple macroscopic colonies could be detected in Zeb2 transgenic mice. We therefore propose that increased Zeb2 expression facilitates the awakening of otherwise dormant ZEB2-negative cells. As such, the observed oscillation of both endogenous and transgenic ZEB2 levels within the primary tumor suggests that the increased metastatic burden may be due to enhanced outgrowth at distant sites, rather than the promotion of primary tumor invasion and metastasis. The ZEB2-mediated growth effect at distant sites is reminiscent of the enhancement of melanoma cell growth at primary cutaneous sites. When melanoma cells reach growth-permissive microenvironments, Zeb2 expression is reinstated to enable expansion. Melanoma cells that acquire elevated Zeb2 expression have a growth advantage outcompete Zeb2-negative cells and contribute to the development of macrometastases, an event that is generally considered as one of the severe rate-limiting steps during metastasis and that is likely more efficiently bypassed via the ZEB2 transgenic expression (27). Although ZEB2 reinforces a differentiated and proliferative phenotype, forced expression of Zeb2 did not necessarily eliminate phenotype-switching abilities in our mouse model of melanoma. Indeed, to achieve local invasion, tumor cells may temporarily abolish ZEB2 protein levels, which results in an oscillation from ZEB2 toward ZEB1, even in tumors with transgenic ZEB2 expression. Phenotype switching involving a dual role of ZEB2 in melanoma development shows striking parallels with the need for reversibility of EMT in metastasis of carcinomas (28, 29). Metastases of the most common human carcinomas often exhibit a redifferentiation. Reexpression of differentiation markers such as E-cadherin, induced by microenvironmental signals, confers a selective advantage and enhances communication with the neighboring parenchyme (30). In regards to this, it is generally accepted that organ colonization by circulating tumor cells is the most complex and rate-limiting step in the metastatic cascade. Our data also establish antagonistic roles for ZEB2 and ZEB1, two members of one EMT TF family. We provide evidence that the ZEB2/ZEB1 ratio is a critical determinant of the melanoma phenotypic state. A high ratio is associated with proliferation, whereas a low ratio favors invasion and migration. The strict dichotomy between proliferation and invasion can be nuanced, as it has been reported that proliferation and invasion are not always mutually exclusive states in melanoma and that several intermediate states exist (31, 32). Also, our single-cell analyses indicate that ZEB1/ZEB2 are coexpressed in a subset of melanoma cells. As elevated ZEB1 levels have been associated with drug resistance of many cancer types and increased survival of circulating tumor cells after extravasation (33), it is tempting to speculate that coexpression of ZEB2 may favor the growth of drug-resistant ZEB1high melanoma cells or the survival of ZEB1high-invasive circulating tumor cells. While we hypothesize that dormant, disseminated melanoma cells need to restore ZEB2 expression to a certain minimal level to regain proliferation potential, survival of circulating tumor cells prior to extravasation might also be enhanced by ZEB2 levels, contributing to the successful organ colonization of ZEB2-positive melanoma cells. While the role of oncogene signaling to direct the reversible reprogramming of EMT-inducing transcription factors in melanoma was characterized thoroughly by Caramel and colleagues, the microenvironmental signals that contribute to this ZEB1/ZEB2 switch in melanoma are largely unknown (25). Changes in the microenvironment are presumed to direct melanocyte plasticity and melanoma phenotype switching (2). In line with this, phenotype switching can initially be triggered by genetic instability that sensitizes the cells toward microenvironmental cues. TGFβ, which is abundant in the tumor microenvironment, is known to regulate phenotype switching (24). The extensive interplay between ZEB transcription factors and members of the TGFβ family in the context of epithelial cells is well established, but has been poorly examined in melanocytes and melanoma (34, 35). Our findings indicate that TGFβ may affect melanoma phenotype switching, at least by partly modulating the ZEB2/ZEB1 ratio. This antagonstic effect on ZEB1/ZEB2 levels is in contrast with the classic view that TGFβ activates both EMT inducers in epithelium-derived malignancies. However, because the role of ZEB2 in carcinoma progression is unrelated to the functions of ZEB2 in neural crest–derived malignancies, the regulation and interplay with TGFβ is also likely to be different. From a therapeutic standpoint, our data indicate that approaches aiming at targeting ZEB2 expression/function may be beneficial to limit the growth of metastatic lesions. However, such an approach is expected, in the same time, to promote the transition towards an invasive state and as such, favor metastatic spreading. Such an approach should therefore only be considered very carefully, in meticulously selected cases, or in combination with drugs that compromise the viability of invasive melanoma cells as well. Our data also call for the careful (re-)examination of the individual biological and pathologic functions of the EMT TFs as the repertoire of their activities keeps expanding beyond what was previously anticipated.

W.M. Gallagher has a paid consulting position and ownership interest with OncoMark Limited, and also has received commercial research funding and has an advisory relationship with Carrick Therapeutics. No potential conflicts of interest were disclosed by the other authors.

Conception and design: N. Vandamme, G. Denecker, G. Berx

Development of methodology: N. Vandamme, G. Denecker, G. Blancke, Ö. Akay, G. Berx

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): N. Vandamme, G. Denecker, K. Bruneel, G. Blancke, Ö. Akay, J. Taminau, E. De Smedt, N. Skrypek, W. Van Loocke, J. Wouters, D. Nittner, C. Köhler, P.F. Cheng, M.I.G. Raaijmakers, M.P. Levesque, F. Rambow, V. Andries, B. Balint, W.M. Gallagher, J.J. Haigh, P. Van Vlierberghe, S. Goossens, J.J. van den Oord, J.C. Marine, G. Berx

Analysis and interpretation of data (e.g. statistical analysis, biostatistics, computational analysis): N. Vandamme, J. De Coninck, F. Rambow, G. Berx

Writing, review, and/or revision of the manuscript: N. Vandamme, J.C. Marine, G. Berx

Administrative, technical, or material support (i.e. reporting or organizing data, constructing databases): U. Girish Mallya, M. Rafferty, B. Balint, W.M. Gallagher, D.S. Darling, L. Brochez, D. Huylebroeck, J.J. van den Oord

Study supervision: G. Berx

N. Vandamme was supported by a personal PhD fellowship at the Institute for the Promotion of Innovation by Science and Technology in Flanders (IWT) (2012-2016) and a personal PhD fellowship from Kom op tegen Kanker (2016-2017). K. Bruneel and E. de Smedt are predoctoral fellows with the FWO. G. Berx's laboratory is supported by the Fonds Wetenschappelijk Onderzoek (3G050217W), Strategic Basic Research (SBO; S008518N), the Geconcerteerde Onderzoeksacties Ghent University (GOA-01GB1013W), Vlaamse Liga tegen Kanker (365U8914U), and the Stichting tegen Kanker (FAF-F/2016/814).

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.
Miller
AJ
,
Mihm
MC
 Jr
. 
Melanoma
.
N Engl J Med
2006
;
355
:
51
65
.
2.
Vandamme
N
,
Berx
G
. 
Melanoma cells revive an embryonic transcriptional network to dictate phenotypic heterogeneity
.
Front Oncol
2014
;
4
:
352
.
3.
Ghislin
S
,
Deshayes
F
,
Middendorp
S
,
Boggetto
N
,
Alcaide-Loridan
C
. 
PHF19 and Akt control the switch between proliferative and invasive states in melanoma
.
Cell Cycle
2012
;
11
:
1634
45
.
4.
Roesch
A
,
Fukunaga-Kalabis
M
,
Schmidt
EC
,
Zabierowski
SE
,
Brafford
PA
,
Vultur
A
, et al
A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth
.
Cell
2010
;
141
:
583
94
.
5.
Eichhoff
OM
,
Weeraratna
A
,
Zipser
MC
,
Denat
L
,
Widmer
DS
,
Xu
M
, et al
Differential LEF1 and TCF4 expression is involved in melanoma cell phenotype switching
.
Pigment Cell Melanoma Res
2011
;
24
:
631
42
.
6.
Eichhoff
OM
,
Zipser
MC
,
Xu
M
,
Weeraratna
AT
,
Mihic
D
,
Dummer
R
, et al
The immunohistochemistry of invasive and proliferative phenotype switching in melanoma: a case report
.
Melanoma Res
2010
;
20
:
349
55
.
7.
Jeffs
AR
,
Glover
AC
,
Slobbe
LJ
,
Wang
L
,
He
S
,
Hazlett
JA
, et al
A gene expression signature of invasive potential in metastatic melanoma cells
.
PLoS One
2009
;
4
:
e8461
.
8.
Denecker
G
,
Vandamme
N
,
Akay
O
,
Koludrovic
D
,
Taminau
J
,
Lemeire
K
, et al
Identification of a ZEB2-MITF-ZEB1 transcriptional network that controls melanogenesis and melanoma progression
.
Cell Death Differ
2014
;
21
:
1250
61
.
9.
Geertsen
RC
,
Hofbauer
GF
,
Yue
FY
,
Manolio
S
,
Burg
G
,
Dummer
R
. 
Higher frequency of selective losses of HLA-A and -B allospecificities in metastasis than in primary melanoma lesions
.
J Invest Dermatol
1998
;
111
:
497
502
.
10.
Higashi
Y
,
Maruhashi
M
,
Nelles
L
,
Van de Putte
T
,
Verschueren
K
,
Miyoshi
T
, et al
Generation of the floxed allele of the SIP1 (Smad-interacting protein 1) gene for Cre-mediated conditional knockout in the mouse
.
Genesis
2002
;
32
:
82
4
.
11.
Gembarska
A
,
Luciani
F
,
Fedele
C
,
Russell
EA
,
Dewaele
M
,
Villar
S
, et al
MDM4 is a key therapeutic target in cutaneous melanoma
.
Nat Med
2012
;
18
:
1239
47
.
12.
Yajima
I
,
Belloir
E
,
Bourgeois
Y
,
Kumasaka
M
,
Delmas
V
,
Larue
L
. 
Spatiotemporal gene control by the Cre-ERT2 system in melanocytes
.
Genesis
2006
;
44
:
34
43
.
13.
Srinivas
S
,
Watanabe
T
,
Lin
CS
,
William
CM
,
Tanabe
Y
,
Jessell
TM
, et al
Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus
.
BMC Dev Biol
2001
;
1
:
4
.
14.
Tatari
MN
,
De Craene
B
,
Soen
B
,
Taminau
J
,
Vermassen
P
,
Goossens
S
, et al
ZEB2-transgene expression in the epidermis compromises the integrity of the epidermal barrier through the repression of different tight junction proteins
.
Cell Mol Life Sci
2014
;
71
:
3599
609
.
15.
Dobin
A
,
Davis
CA
,
Schlesinger
F
,
Drenkow
J
,
Zaleski
C
,
Jha
S
, et al
STAR: ultrafast universal RNA-seq aligner
.
Bioinformatics
2013
;
29
:
15
21
.
16.
Robinson
MD
,
McCarthy
DJ
,
Smyth
GK
. 
edgeR: a Bioconductor package for differential expression analysis of digital gene expression data
.
Bioinformatics
2010
;
26
:
139
40
.
17.
Jerby-Arnon
L
,
Shah
P
,
Cuoco
MS
,
Rodman
C
,
Su
MJ
,
Melms
JC
, et al
A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade
.
Cell
2018
;
175
:
984
97
e24.
18.
Johnson
DB
,
Puzanov
I
. 
Treatment of NRAS-mutant melanoma
.
Curr Treat Options Oncol
2015
;
16
:
15
.
19.
Verfaillie
A
,
Imrichova
H
,
Atak
ZK
,
Dewaele
M
,
Rambow
F
,
Hulselmans
G
, et al
Decoding the regulatory landscape of melanoma reveals TEADS as regulators of the invasive cell state
.
Nat Commun
2015
;
6
:
6683
.
20.
Hoek
KS
,
Eichhoff
OM
,
Schlegel
NC
,
Dobbeling
U
,
Kobert
N
,
Schaerer
L
, et al
In vivo switching of human melanoma cells between proliferative and invasive states
.
Cancer Res
2008
;
68
:
650
6
.
21.
Tarantola
M
,
Marel
AK
,
Sunnick
E
,
Adam
H
,
Wegener
J
,
Janshoff
A
. 
Dynamics of human cancer cell lines monitored by electrical and acoustic fluctuation analysis
.
Integr Biol
2010
;
2
:
139
50
.
22.
Keese
CR
,
Bhawe
K
,
Wegener
J
,
Giaever
I
. 
Real-time impedance assay to follow the invasive activities of metastatic cells in culture
.
BioTechniques
2002
;
33
:
842
4
, 46, 48–50.
23.
Perrot
CY
,
Gilbert
C
,
Marsaud
V
,
Postigo
A
,
Javelaud
D
,
Mauviel
A
. 
GLI2 cooperates with ZEB1 for transcriptional repression of CDH1 expression in human melanoma cells
.
Pigment Cell Melanoma Res
2013
;
26
:
861
73
.
24.
Javelaud
D
,
Alexaki
VI
,
Pierrat
MJ
,
Hoek
KS
,
Dennler
S
,
Van Kempen
L
, et al
GLI2 and M-MITF transcription factors control exclusive gene expression programs and inversely regulate invasion in human melanoma cells
.
Pigment Cell Melanoma Res
2011
;
24
:
932
43
.
25.
Caramel
J
,
Papadogeorgakis
E
,
Hill
L
,
Browne
GJ
,
Richard
G
,
Wierinckx
A
, et al
A switch in the expression of embryonic EMT-inducers drives the development of malignant melanoma
.
Cancer Cell
2013
;
24
:
466
80
.
26.
Tsherniak
A
,
Vazquez
F
,
Montgomery
PG
,
Weir
BA
,
Kryukov
G
,
Cowley
GS
, et al
Defining a cancer dependency map
.
Cell
2017
;
170
:
564
76
.
27.
Vanharanta
S
,
Massague
J
. 
Origins of metastatic traits
.
Cancer Cell
2013
;
24
:
410
21
.
28.
Brabletz
T
. 
EMT and MET in metastasis: where are the cancer stem cells?
Cancer Cell
2012
;
22
:
699
701
.
29.
Puisieux
A
,
Brabletz
T
,
Caramel
J
. 
Oncogenic roles of EMT-inducing transcription factors
.
Nat Cell Biol
2014
;
16
:
488
94
.
30.
Palen
K
,
Weber
J
,
Dwinell
MB
,
Johnson
BD
,
Ramchandran
R
,
Gershan
JA
. 
E-cadherin re-expression shows in vivo evidence for mesenchymal to epithelial transition in clonal metastatic breast tumor cells
.
Oncotarget
2016
;
7
:
43363
75
.
31.
Haass
NK
,
Beaumont
KA
,
Hill
DS
,
Anfosso
A
,
Mrass
P
,
Munoz
MA
, et al
Real-time cell cycle imaging during melanoma growth, invasion, and drug response
.
Pigment Cell Melanoma Res
2014
;
27
:
764
76
.
32.
Tuncer
E
,
Calcada
RR
,
Zingg
D
,
Varum
S
,
Cheng
P
,
Freiberger
SN
, et al
SMAD signaling promotes melanoma metastasis independently of phenotype switching
.
J Clin Invest
2019
;
129
:
2702
16
.
33.
Smit
MA
,
Peeper
DS
. 
Zeb1 is required for TrkB-induced epithelial-mesenchymal transition, anoikis resistance and metastasis
.
Oncogene
2011
;
30
:
3735
44
.
34.
Gregory
PA
,
Bracken
CP
,
Smith
E
,
Bert
AG
,
Wright
JA
,
Roslan
S
, et al
An autocrine TGF-beta/ZEB/miR-200 signaling network regulates establishment and maintenance of epithelial-mesenchymal transition
.
Mol Biol Cell
2011
;
22
:
1686
98
.
35.
De Craene
B
,
Berx
G
. 
Regulatory networks defining EMT during cancer initiation and progression
.
Nat Rev Cancer
2013
;
13
:
97
110
.