Dvdy 24678

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

DEVELOPMENTAL DYNAMICS 248:10–20, 2019

DOI: 10.1002/dvdy.24678

REVIEW

Epithelial-mesenchymal Transition and Cancer


Stem Cells: At the Crossroads of Differentiation
and Dedifferentiation
Hanmin Wang and Juli J. Unternaehrer*

Division of Biochemistry, Department of Basic Sciences, Loma Linda University, Loma Linda, California

In this review, we explore the connections between epithelial-mesenchymal transition (EMT) and differentiation status. EMTs
in development have been described as differentiation events, while in most cases EMTs in cancer have been depicted as
dedifferentiation events. We will briefly summarize both embryo development and cancer progression with regard to the
involvement of EMT and cell differentiation status. We further present the studies that provide evidence that EMT results in
both differentiation and dedifferentiation. Finally, we present our resolution to this dilemma by suggesting that EMT brings
about dedifferentiation that enables subsequent differentiation. In normal development, EMT events may cause a partial rever-
sal of differentiation to overcome differentiation barriers. When EMT is aberrantly activated in cancer, cells gain attributes of
stem cells that contribute to self-renewal capabilities and are able to differentiate to all cell types represented in the tumor.
The resulting cancer stem cells attain hallmarks of cancer, including replicative immortality, resistance to cell death, and inva-
siveness. Developmental Dynamics 248:10–20, 2019. © 2018 Wiley Periodicals, Inc.
DEVELOPMENTAL DYNAMICS

Key words: development; reprogramming; miRNA regulation; epithelial-mesenchymal transition; differentiation

Submitted 29 January 2018; First Decision 29 May 2018; Accepted 27 September 2018; Published online 22 November 2018

Introduction process of differentiation (Bell and Watson, 2009; Lim and Thiery,
2012). The steps involving EMT are thus suggested to be part of
Stemness refers to a cell’s ability to self-renew and differentiate. the loss of potency (differentiation potential) associated with
It is a characteristic of all stem cell categories, including embry- development. However, some data describe cells that have under-
onic, adult, and cancer stem cells (Batlle and Clevers, 2017). Cells gone EMT as more stem cell–like than their progenitors, implying
with the greatest degree of stemness are the least differentiated. that EMT causes dedifferentiation (Thiery, 2002). These changes
Many tumors adopt an oncofetal gene expression pattern, with are caused by the action of several transcription factors, including
reversion to an embryonic phenotype, including expression of plur- Snai1 (Snail), Snai2 (Slug), Twist, ZEB1, and ZEB2, which are
ipotency genes (Al-Hajj et al., 2003; Schatton et al., 2008). Pluripo- potent repressors of epithelial gene expression (Batlle et al., 2000;
tency refers to a cell’s ability to differentiate into all embryonic Bolós et al., 2003; Cano et al., 2000; Comijn et al., 2001; Hajra
lineages and is best illustrated by embryonic stem (ESCs) and et al., 2002; Yang et al., 2004). These factors also have actions
embryonal carcinoma cells. An illustration of cell states discussed independent of the EMT, thus the process of EMT must be distin-
in this review is shown in Figure 1. Based on the degree of stem- guished from the expression of transcription factors mediating
ness (developmental potential), cell states are categorized as totipo- EMT (Vega et al., 2004; Vesuna et al., 2008; Zhang et al., 2015).
tent, pluripotent, multipotent and unipotent (Fig. 1). We compare the complete dedifferentiation that occurs in somatic
In addition to the involvement of stem cells, embryo develop- cell reprogramming (Takahashi and Yamanaka, 2006) to the par-
ment and cancer progression share other similarities. One com- tial dedifferentiation that occurs in EMT (Fig. 1). In this review,
mon feature is a process called epithelial-mesenchymal transition we focus on the hierarchical relationships between these states
(EMT) (Hay, 1995; Martin-Belmonte and Perez-Moreno, 2011; and examine the role of EMT in state transitions.
Thiery et al., 2009). Through EMT, epithelial cells lose their cell In general, EMTs in development have been described as dif-
polarity and cell-cell adhesion, and gain migratory and invasive ferentiation events, while EMT in cancer has been depicted as a
features to become mesenchymal cells. EMT is essential for dedifferentiation event. The end result of the EMT-associated
embryogenesis, which has typically been described as a gradual processes might partially cause this dichotomy. In the following
sections, we summarize the apparently contradictory literature

*Correspondence to: Juli Unternaehrer, Assistant Professor, Department of


Basic Sciences, Division of Biochemistry, Loma Linda University School of Article is online at: http://onlinelibrary.wiley.com/doi/10.1002/dvdy.
Medicine, 11085 Campus Street, Mortensen Hall 219, Loma Linda, CA 24678/abstract
92354. E-mail: junternaehrer@llu.edu © 2018 Wiley Periodicals, Inc.

10
EMT IN DIFFERENTIATION AND DEDIFFERENTIATION 11

pointing to EMT as differentiation vs. dedifferentiation events. differentiation occurs in the mesenchyme derived from the chro-
We present a synopsis of the literature pertaining to EMT and nologically earliest EMT (Pérez-Pomares and Muñoz-Chápuli,
the differentiation state in an attempt to reconcile these bodies 2002). However, this developmental potential progressively
of literature. In the end, we postulate that although the overall decreases in later EMTs. In addition, endocardial cushion mesen-
trajectory of developmental processes is toward differentiation, chyme is formed from endocardium cells through another round
cells partially and transiently dedifferentiate when they undergo of EMT (Carmona et al., 2000); cell differentiation happens during
EMT. This loss of differentiation represents an opportunity for this process (Romano and Runyan, 1999).
adjusting gene expression programs and allows new gene EMT is essential for neural crest formation. In this process, cells
expression required for formation of new tissue types in devel- undergo EMT and delaminate from the neuroepithelium, enabling
opment. Similarly, the transient loss of differentiation seen in them to migrate and form the neural crest progeny, including
cancer-associated EMT provides a window of opportunity for neurons, endocrine cells, cartilage, bone, dermis, connective tis-
acquisition of a stem cell phenotype. Thus, stem cells or differen- sue, and adipose tissues in the head and neck (Christ et al., 1983;
tiated cells in which EMT transcription factors are active may Dupin et al., 2007). Because of their remarkable differentiation
simultaneously receive signals leading to the loss of key factors potential, the neural crest has been called the “fourth germ layer”
responsible for maintaining differentiation. (Hall, 2000). In addition, neural crest delamination represents one
of the most distinct developmental mechanisms that involves
EMT. Neural crest cells originate in the neural fold or the neural
EMT in Development tube, then migrate and delaminate as mesenchymal-like cells to
EMT is essential for many developmental processes and plays a target locations, where they differentiate into various cell
role in embryonic differentiation in many contexts across the types (Gammill and Bronner-Fraser, 2003; Nikitina et al., 2008;
spectrum of metazoans (Thiery and Sleeman, 2006). EMT is Steventon et al., 2005; Weston and Thiery, 2015).
involved in the initiation of placenta formation and the implanta- Further EMTs happen throughout development, for example,
DEVELOPMENTAL DYNAMICS

tion of the embryo (Vicovac and Aplin, 1996). Subsequently, dur- in the formation of pancreas, kidney and liver; in the somites to
ing gastrulation, cells in the primitive streak (or comparable form sclerotome and myotome; and in cardiac development
structures such as the ventral furrow in Drosophila, blastoderm (Costello et al., 2011; Nakajima et al., 2000). To better under-
margin in fish) undergo EMT to cause the ingression that leads to stand EMT in development, it is also important to consider
germ layer and mesenchyme formation in metazoans (Baum mesenchymal-epithelial transition (MET), the reverse process of
et al., 2008; Leptin and Grunewald, 1990). Through invagination, EMT (Lim and Thiery, 2012). Mesenchymal cells originating from
at the primitive streak, the epiblast generates mesendoderm, which the primitive streak take part in the formation of epithelial
further separates into mesoderm and endoderm through EMT (Lim mesodermal organs like somites through MET. Waves of EMT
and Thiery, 2012). In development, Snail expression is detected as and MET occur repeatedly during organogenesis (Lim and Thi-
early as the two-cell stage in mammalian embryos (Bell and Wat- ery, 2012). During this process, cells disseminate to various parts
son, 2009). Snail is expressed in a variety of species: It is of the embryo and differentiate into many types of cells.
expressed during the formation of the ventral furrow in Drosoph- Heart formation is an illustrative example of organogenesis
ila (Leptin and Grunewald, 1990); it was also found in stroma of that involves multiple EMTs. Cardiac-specified cells first undergo
the choroid plexus of both the hindbrain and the forebrain in EMT to migrate rostrally, and a subsequent MET leads to the for-
chicken embryo development (Marin and Nieto, 2004). An ortho- mation of two cardiogenic territories, followed by the heart pri-
log of Snail is critical for primary mesenchyme cell ingression in mordium formation. Then, a second cycle of EMT/MET creates
sea urchin embryo (Wu and McClay, 2007). Snail mutant mouse the endothelial cell lining of the heart. A third cycle creates the
embryos do not survive gastrulation, confirming Snail’s impor- endocardial cushion and its derivatives. A cluster of mesothelial-
tance in early development (Carver et al., 2001). A conditional derived cells undergoes another MET to form epicardium. During
knockout of Snail in epiblast, however, affects only the mesoderm the following, fourth cycle, epicardial cells delaminate and dif-
migration, not its initial formation (Lomelí et al., 2009; Murray ferentiate into epicardial-derived cells—mesenchymal-like cells
and Gridley, 2006). Thus, multiple functions of EMT can be teased that further populate the subepicardium—and are responsible for
out, and differentiation can occur separately from migration. The the formation of endothelial cells, coronary smooth muscle, and
biological importance of Snail is illustrated by the conservation of cardiac fibroblasts (Chua et al., 2011). It takes four cycles of
its function in a wide spectrum of species. EMT for progenitor cells to form cardiac fibroblasts; after each
In the following sections, we will present EMT events involved cycle, cells differentiate into a more specialized cell type.
in embryo development that are linked to either cell differentia- EMT-related gene expression changes may also provide input
tion or cell dedifferentiation. to the differentiation state. Cells that undergo EMT change their
adhesion-molecule gene expression, down-regulating epithelial
factors, such as E-cadherin, occludin, and Traffic Jam 1 (TJ1),
Evidence that EMT Results in Differentiation and up-regulating those associated with the mesenchymal state,
Several events in natural development suggest that EMT results in such as N-cadherin (Cano et al., 2000; Hazan et al., 1997; Islam
differentiation (Fig. 2A). For instance, when cells invaginate from et al., 1996; Kim et al., 2000). Snail promotes the cadherin
the primitive streak to form the mesoderm through EMT, they dif- switch from E-cadherin to N-cadherin expression (Oda et al.,
ferentiate into lineages including blood (Eichmann et al., 2005; 1998). The expression of N-cadherin plays roles in differentia-
Fehling et al., 2003), skeletal muscle (Pu et al., 2015), and skin tion: For example, N-cadherin expression controls myogenic dif-
(Nitzan and Kalcheim, 2013). In the general process of embryo- ferentiation (Charrasse et al., 2002). Recently, Schafer
genesis, after an EMT event, differentiation of the newly produced et al. suggested that, in addition to the surface adhesion changes
mesenchyme follows. The highest diversity in cell lineage caused by the cadherin switch, Snail may serve to facilitate
12 WANG AND UNTERNAEHRER

Fig. 1. Epithelial-mesenchymal transitions in Waddington’s epigenetic landscape. Dashed arrows indicate state changes. Solid blue lines indicate
added differentiation potential of cells after EMT. EMT is proposed to be associated with partial dedifferentiation, resulting in cells with increased
stemness. In development, resulting new gene expression allows cells to differentiate into new cell types. In cancer, dedifferentiation endows cells
with self-renewal abilities and ability to regenerate all tumor cell types. Terminally differentiated cells (green to yellow) or multipotent progenitors (red
to pink) can undergo EMT and be partially reprogrammed. Classical reprogramming to pluripotency (e.g., by Yamanaka factors) is indicated by green
to blue, partial reprogramming by green to red or yellow, and direct reprogramming by green to green lines. Adapted from Waddington (Waddington,
1957) and Hochedlinger and Plath (Hochedlinger and Plath, 2009), with permission.
DEVELOPMENTAL DYNAMICS

downstream events such as Wnt signaling (Schäfer et al., 2014). et al., 2013); similarly, Twist overexpression in MSCs results in
The Wnt signaling pathway is well known to participate in cell maintenance of an immature phenotype and prevents differentia-
proliferation and cell differentiation (Reya and Clevers, 2005). tion to bone and cartilage, but allows differentiation to adipose
These gene expression changes hence indicate cell differentiation tissue (Isenmann et al., 2009). These data implicate Snail and
after EMT. Twist in maintaining the dedifferentiated state.
Evidence from pluripotent cell biology also supports the con- Several observations from the reprogramming field also provide
cept that EMT leads to differentiation. In ESCs, which display evidence that dedifferentiation results when EMT factors are
many but not all characteristics of bona fide epithelium, sponta- expressed. Somatic cells can be dedifferentiated, or reprogrammed,
neously differentiating cells have been described to undergo to pluripotency by the expression of the four “Yamanaka” factors
EMT at the periphery of colonies (Ullmann et al., 2007). This is (Park et al., 2008; Takahashi et al., 2007; Takahashi and Yamanaka,
thought to be analogous to gastrulation (Kim et al., 2014). When 2006; Yu et al., 2007). Reprogramming is a multistep process
human ESCs differentiate, they undergo EMT (Li et al., 2017). involving both EMT and MET that requires many days to accom-
Galvagni et al. (2015) used ESCs expressing tamoxifen-inducible plish (Samavarchi-Tehrani et al., 2010). Snail and other mesenchy-
Snail1 to investigate the role of Snail1 expression in early ESC mal gene expression increase early in fibroblast and keratinocyte
differentiation. Eight hours after induction, they analyzed gene reprogramming before waning as the reprogrammed cells take on
expression and found 17 self-renewal-related genes were down- an epithelial fate (Samavarchi-Tehrani et al., 2010). An early induc-
regulated, while the up-regulated genes are normally expressed tion of EMT, either by sequential application of reprogramming fac-
in later-stage, more differentiated stem cells. They conclude that tors, transient Transforming Growth Factor-beta (TGF-β) treatment,
Snail1 expression induces ESC exit from pluripotency. In this or expression of Snail or Slug, improves reprogramming efficiency
case, the EMT was associated with differentiation. (Liu et al., 2013). Inhibitors of TGFb signaling, and thus of EMT,
All the developmental events mentioned previously link EMT antagonize reprogramming when administered prior to the repro-
to differentiation. gramming factors; later, they enhance the process as expected, since
they promote MET (Maherali and Hochedlinger, 2009). Consistent
with these findings, reprogramming efficiency is decreased by
Evidence that EMT Results in Dedifferentiation inhibiting, and increased by overexpressing, Snail (Unternaehrer
Unlike the evidence presented earlier, there are studies that define et al., 2014). Thus, EMT appears to enhance early steps of
EMT as a dedifferentiation event. In embryo development, another dedifferentiation.
member of the Snail family, Slug, acts in concert with Sox9 to An important signaling pathway in pluripotency and early
induce differentiated luminal cells of the mammary gland to embryo development is the Wnt pathway, which exerts pleiotro-
acquire stem cell characteristics (Guo et al., 2012). Twist acts to pic effects dependent on context. Because of the involvement of
maintain the immature or dedifferentiated phenotype, and its loss Wnt signaling in both stemness and EMT (Fabregat et al., 2016;
allows myoblast, chondrocyte, and osteoblast differentiation Gonzalez and Medici, 2014; Micalizzi et al., 2010), studies on
(Alborzi et al., 1996; Dong et al., 2007; Hebrok et al., 1994). Over- this pathway provide insight to how the two processes relate.
expression of Snail or Twist in breast epithelial cells results in Wnt signaling maintains pluripotency and the self-renewal abil-
cells with increased differentiation potential, similar to mesenchy- ity of mouse ESCs (Anton et al., 2007; Bose et al., 2007; Essafi
mal stem cells (MSCs) (Battula et al., 2010). Snail overexpression et al., 2011). A higher percentage of ESCs formed colonies when
in MSCs blocks differentiation to bone and adipocyte fate (Batlle plated in the presence of Wnt3a protein, whereas the Wnt
EMT IN DIFFERENTIATION AND DEDIFFERENTIATION 13

signaling results in both stemness and EMT draws a further con-


nection between these phenomena.
Taken together, these findings point to a role for EMT factors
in the acquisition of pluripotency.

EMT in Cancer
EMT also occurs in cancer cells: Tumor cells recapitulate ontog-
eny, thus inappropriately activating EMT (Thiery, 2002). Transi-
tions between epithelial and mesenchymal states in cancer cells
are linked with their ability to spread (Peinado et al., 2007;
Polyak and Weinberg, 2009). A subpopulation of cancer cells in
primary tumors undergoes EMT, resulting in enhanced migratory
and invasive properties. This is an early step of establishing a
metastasis and forming secondary tumors (Thiery, 2002).
Although developmental EMT is described either as differentia-
tion or dedifferentiation, cancer studies have almost exclusively
depicted EMT as dedifferentiation (Fig. 2B).

EMTs Result in Acquisition of Stemness in Cancer Cells


Rare tumor cells possess the ability to self-renew and initiate
DEVELOPMENTAL DYNAMICS

tumors; these are known as cancer stem cells (CSCs) or tumor-


initiating cells (Sheridan et al., 2006). The source of CSCs is the
topic of much investigation and debate. Although several factors
contribute to the CSC state, much evidence connects EMT with
the acquisition of stem cell properties (Al-Hajj et al., 2003; Mani
et al., 2008; Santisteban et al., 2009; Wielenga et al., 1999).
Studies in breast cancer describe the EMT cells as having proper-
ties of mesenchymal stem cells that are able to differentiate to
multiple lineages (Battula et al., 2010). Thus, similar to some of
Fig. 2. Diagram of epithelial-mesenchymal transition effect on the EMTs that occur in development, cancer-associated EMTs
differentiation status. A: Many findings suggest that EMT results in result in more migratory cells capable of forming new tissues,
differentiation. During the process of EMT, cells lose stemness and indicating increased stemness. CSCs exhibit dynamic changes
undergo differentiation. The yellow end of the spectrum represents the
stem cell fate; the blue represents differentiated cells. B: Other findings between epithelial and mesenchymal characteristics in squamous
suggest that EMT results in dedifferentiation. Some studies have cell carcinoma and breast cancer: Epithelial cells are more prolif-
concluded the opposite, that during the process of EMT, cells erative, while mesenchymal cells are more migratory (Biddle
dedifferentiate and attain stem cell–like features. C: The data are et al., 2011; Liu et al., 2014).
consistent with a third explanation, that EMT, especially partial EMT,
results in transient dedifferentiation allowing differentiation. The action of
In support of the concept that EMT enhances stemness, cells that
EMT transcription factors may bring about a reprogramming event have undergone EMT form at least 10-fold more tumor spheres, a
resulting in dedifferentiation. The resulting stem-like cells attain self- characteristic of stem cells. Similarly, Twist inhibits CD24 expres-
renewal capabilities and display increased differentiation potential. sion, suggesting a direct link to the CSC phenotype (Vesuna et al.,
2009). (Human mammary stem cells have been characterized to
antagonist reduced, and at high concentration completely sup- express high levels of CD44 and low levels of CD24 [Al-Hajj et al.,
pressed, self-renewal (ten Berge et al., 2011). Furthermore, the 2003; Sleeman et al., 2006].) In addition to these studies related to
effect was rescued by addition of Wnt3a protein. Wnt signals are breast cancer, similar association between EMT and stemness acqui-
hence proved to be essential self-renewal factors for ESCs and sition has been observed in pancreatic (Wang et al., 2009; Wellner
are required to inhibit their differentiation into more advanced et al., 2009), prostate (Kong et al., 2010), and colorectal cancers (Fan
epiblast-derived stem cells (ten Berge et al., 2011). In addition, et al., 2012; Hwang et al., 2011; Hwang et al., 2014). Snail initiates
Wnt signaling increases the efficiency with which differentiated epithelial dedifferentiation in colon cancer cells (De Craene et al.,
cells can be reprogrammed toward pluripotency (Lluis et al., 2005). Similarly, the EMT transcription factor ZEB1 dedifferentiates
2008; Marson et al., 2008). cancer cells by repressing several epithelial fate determinants (Aigner
Wnt signaling is also crucially involved in EMT (ten Berge et al., et al., 2007). In cancers including gastric (He et al., 2012), breast
2008; Lindsley et al., 2006). The Wnt signaling pathway induces (Manning et al., 1994), liver (Tsutsumi et al., 2004), and colon (Roy
EMT in various models including colon- and breast-carcinoma cell et al., 2004), Snail expression correlates with the dedifferentiated
lines and mammary epithelial cells (Kim et al., 2002; Yook et al., phenotype. These results suggest that differentiated cancer cells that
2006). Furthermore, in breast cancer patients, markers indicating undergo EMT exhibit a more CSC-like phenotype; that is, cells
active Wnt signaling also correlate clinically with poor prognosis become less differentiated after EMT.
(Logullo et al., 2010; Prasad et al., 2009). Another study showed Not only does EMT confer stemness properties, CSCs also display
Wnt signaling engages tumor cell dedifferentiation while stabilizing more mesenchymal properties: Breast cancer cell lines with high
Snail, further inducing EMT (Yook et al., 2006). The fact that Wnt CD44+/CD24- cell numbers not only show high stem/progenitor
14 WANG AND UNTERNAEHRER

properties, but also exhibit strongly enhanced basal/mesenchymal 2015). Examples of differentiation-promoting miRNAs are the
markers (Sheridan et al., 2006). Similar results have been observed let-7 family and miR-34. The let-7 family of miRNAs is present
in nontumorigenic immortalized human mammary epithelial cells in all somatic cells (Boyerinas et al., 2010), thus presenting a
(HMLEs) as well. After EMT is induced via ectopic expression of barrier to stemness. Let-7’s pluripotency and oncogene targets
either Twist or Snail (Mani et al., 2008), most of the mesenchymal- qualify it as a tumor suppressor (Boyerinas et al., 2010). Let-7
like cells generated indeed acquire this CD44high/CD24low pattern, inhibits self-renewal genes such as Lin28, Myc, and Sall4
indicating that HMLEs also become more stem-like after undergo- (Melton et al., 2010), and its expression is decreased in many
ing EMT. The acquired stem cell properties are also observed in cancers (Dahiya et al., 2008; O’Hara et al., 2009; Takamizawa
functional assays. Tumor sphere–forming ability has long been et al., 2004). Mechanisms of let-7 loss are incompletely under-
used as a way to test cells’ stemness (Dontu et al., 2003). HMLEs in stood (Wang et al., 2012).
which Snail or Twist is overexpressed form >30-fold more tumor Let-7 is also lost during the process of somatic cell reprogram-
spheres. Also, the number of tumor-initiating cells increases at least ming, in which expression of the transcription factors Oct4,
two-fold with constitutive expression of EMT-inducing transcrip- Sox2, Klf4, and c-Myc cause differentiated cells to take on the
tion factors, Twist or Snail (Hollier et al., 2013; Mani et al., 2008; phenotype of pluripotent stem cells. Studies of reprogramming
Morel et al., 2008), indicating dedifferentiation after EMT. provide clues to mechanisms of let-7 loss. In reprogramming,
Morel et al. evaluated breast cancer stem cells by examining let-7 levels decrease to allow expression of pluripotency targets,
the expression of CD24 (Morel et al., 2008). They observed that and let-7 inhibition increases efficiency (Melton and Blelloch,
CD24-negative cells, which display mesenchymal properties as 2010). Snail binds let-7 promoters; its expression increases early
opposed to the epithelial characteristics of the CD24-positive in reprogramming in parallel with the decrease in let-7
cells, are able to grow as tumor spheres and form tumors (Unternaehrer et al., 2014). In pluripotent cells, levels of mature
in vivo. The transition to the CD24-negative fate was accom- let-7 are kept low to absent by the action of Lin28, which pre-
plished by TGF-β-induced EMT (Morel et al., 2008). Accordingly, vents its processing (Viswanathan et al., 2008). In other con-
DEVELOPMENTAL DYNAMICS

the EMT factor Snail initiates the plasticity required in tumor texts, Twist also represses let-7 transcription (Yang et al., 2012).
cells for both migration and stemness (Baulida and García de Thus, EMT factors are a potential source of transcriptional
Herreros, 2015). Results from Morel et al. and Baulida repression of let-7 and other tumor-suppressor miRNAs.
et al. indicate that cells dedifferentiate after EMT. Similar to let-7, miR-34 targets pluripotency factors, and its
In addition, Tran et al. and Ye et al. used mouse breast cancer expression is a barrier to stemness (Choi et al., 2011). As a p53 tar-
reporter models to follow the endogenous activation of Snail get, its expression is likely dysregulated in p53-mutated cancers
and demonstrated that, in primary tumors, Snail is activated fol- (He et al., 2007). Snail represses miR-34 expression (Siemens et al.,
lowed by the induction of EMT, and that cells with Snail activa- 2011), which may contribute to the stemness phenotype observed
tion have higher tumor-initiation capacity, another robust in EMT cells. Besides let-7 and miR-34, many other miRNAs could
measure of stemness (Tan et al., 2014; Ye et al., 2015). These exert similar effects in establishment of the stem cell fate in cancer
observations also contribute to our understanding of the role of (Esquela-Kerscher and Slack, 2006). In fact, the action of let-7 is
EMT in acquisition of stemness. not sufficient to overcome the action of miRNAs that activate self-
As mentioned previously, EMT causes an early step in cancer renewal (called ESC-specific cell cycle [ESCC]–regulating or ESCC
cell metastasis. Thereafter, in order to colonize a new niche and miRNAs), including the miR290 cluster (Wang et al., 2008). Thus,
form a metastatic tumor, reversion to a more epithelial state is loss of let-7 is necessary, but not sufficient, to achieve self-renewal.
required (Ocaña et al., 2012; Tsai et al., 2012). Mouse breast can- In addition, ESCC miRNAs must be expressed (Melton et al., 2010).
cer cells in vivo spontaneously undergo EMT, causing them to This illustrates the complicated path to stemness, as well as the
disseminate, after which they revert back to the epithelial state checks and balances that exist to protect the differentiated state.
to form metastases (Beerling et al., 2016). Although the first As early as 1991, the loss of E-cadherin now known to be
EMT of the metastatic process is often associated with cells gain- associated with EMT was correlated with dedifferentiation of
ing stemness, a subsequent MET might be the reason most tumors (Frixen et al., 1991). More recently, it has been shown
metastases are differentiated (Brabletz et al., 2001). that expression of individual EMT-inducing transcription factors
In summary, the literature on EMT and stemness in cancer brings about the stem cell state. ZEB1, a transcription factor that
suggests that the process of EMT may be causative for the stem induces EMT in carcinoma cells (Zhang et al., 2015), is specifi-
cell phenotype. cally up-regulated in pancreatic carcinomas with stem-like phe-
notypes and is necessary for tumor initiation in a xenograft
model (Wellner et al., 2009). Similar to the effects of Snail on
Mechanism of EMT-induced Cancer Stemness let-7 and miR-34 noted earlier, the EMT transcription factor
The mechanism by which EMT exerts these stemness effects is ZEB1 inhibits miRNAs including miR-203, miR-200, and miR-
not completely understood. In this review we focus on the direct 183, all of which down-regulate stemness factors such as Sox2,
transcriptional actions of the EMT factors. Besides their protein- KLF4, and BMI1 (Wellner et al., 2009). Hence, ZEB1 provides
coding targets, one way that EMT transcription factors act is by another link between EMT and cell dedifferentiation by inhibit-
regulating the expression of non-coding RNAs. MicroRNAs ing these miRNAs.
(miRNAs, small non-coding RNA molecules that function in In ovarian cancer development, EMT in ovarian surface epithe-
post-transcriptional regulation of gene expression) play a pivotal lial cells is reported to result in dedifferentiation (Ahmed et al.,
role in regulating differentiation (Lee et al., 2005; Takamizawa 2007; Schipper et al., 1991). EMT factors inhibit a variety of differ-
et al., 2004; Wellner et al., 2009). Factors regulating miRNA entiation pathways, including NF-kB, Tumor necrosis factor alpha,
expression and processing could thus influence the balance β-catenin, and p53 (Pan et al., 2009; Sharabi et al., 2008; Šošic
between stemness and differentiation (Takahashi and Yamanaka, et al., 2003). Another way EMT factors contribute to the
EMT IN DIFFERENTIATION AND DEDIFFERENTIATION 15

dedifferentiated phenotype is by preventing senescence. The life undergoing partial MET. In addition, compared to their progeni-
span of somatic cells, in terms of cell division, is limited: Differenti- tors, these cells displayed increased self-renewal, plasticity, and
ated cells senesce and cease dividing when they reach this limit. tumor sphere–forming ability (Grosse-Wilde et al., 2015). Partial
Both stem cells and cancer cells overcome this barrier, which allows EMT results in stemness gains both in development and in cancer
them to proliferate indefinitely. Snail and Twist contribute to this (Jolly et al., 2014; Swetha et al., 2011). Accordingly, a change in
process by directly repressing tumor suppressors such as the cell status toward either mesenchymal or epithelial phenotypes results
cycle regulator p16 (Ansieau et al., 2008; Li et al., 2018). Thus, in a loss of stem cell characteristics (Strauss et al., 2011).
several molecular mechanisms contribute to EMT-driven stemness. To illustrate our premise, cell fate decisions in both develop-
Clinically, it can also be inferred that EMT status links to stem- ment and cancer can be symbolized by Waddington’s landscape
ness. Based on gene expression profiling, Tan et al. developed a (Fig. 1). Differentiation is symbolized by downhill movement in
quantitative EMT tumor–scoring system (Tan et al., 2014). This the hills and valleys in this schema, dedifferentiation by uphill
system highlights the distinct EMT states for each cancer type. In movement. During or after the process of oncogenesis, termi-
ovarian cancer, the EMT status is closely linked to the reported nally differentiated cells (those at the bottom of the landscape)
gene expression–based molecular subtypes, where worse progno- or progenitors (partway up the hill) may be stimulated to
sis is associated with higher EMT score (Tan et al., 2013). In ovar- undergo EMT by a variety of aberrant signaling pathways. The
ian cancer patients, prognosis is often indicated by EMT process may nudge cells in an uphill direction, causing
clinicopathological parameters, such as metastasis to distant them to obtain stem cell attributes such as self-renewal as they
organs and response to chemotherapy (Gilks and Prat, 2009). dedifferentiate. Subsequent steps may involve MET, and as illus-
Metastasis and high chemoresistance also indicate a higher level trated by cardiac development previously, multiple rounds of
of pluripotency (Safa, 2016). This means that subtypes with higher EMT/MET may occur during the course of morphogenesis.
EMT scores are associated with the dedifferentiated state. Physiological developmental decisions begin at the top of the
These findings support the conclusion that in cancer cells, mountain with totipotent zygotes, which have the highest devel-
DEVELOPMENTAL DYNAMICS

potency or stemness is gained after cells have gone through EMT. opmental potential. At the bottom, cells are fully differentiated.
At each cell division in development, cells choose between two
or more potential fates. Just as a hiker chooses to go left or right
Resolution: EMT Results in Transient at a fork in the trail, cells proceed into one valley or another,
Dedifferentiation Allowing Differentiation representing varying developmental pathways. Each of these fate
decisions toward a particular differentiated cell type alters gene
Clearly, the relationship between EMT/MET and stemness is not expression patterns in ways that are necessary for the current
straightforward. EMT is not equal to dedifferentiation, and many decision but may set up barriers for subsequent decisions. Thus,
other factors and processes contribute to the complex picture of certain decisions require “back-tracking” that can be thought of
cell differentiation state. However, going through the process of as going uphill in Waddington’s landscape. EMT may provide
EMT may increase the probability of enhanced plasticity (Nieto, the mechanism for these dedifferentiation events.
2013; Wahl and Spike, 2017) (Fig. 2C). Thus, in development, when cells undergo EMT, they ulti-
Recent studies suggest that cell dedifferentiation can also be mately differentiate; but what is the status of the cells during
associated with cells undergoing a partial EMT, resulting in hybrid and immediately after EMT? EMT may result in partial dediffer-
Epithelial/Mesenchymal (E/M) phenotype (Forte et al., 2017; Jolly entiation sufficient to allow for new gene expression required in
et al., 2015; Liao and Yang, 2017; Thiery et al., 2009). Jolly the nascent tissue types (Fig. 2C). A reprogramming frame of ref-
et al. postulated that the core EMT and stemness modules, miR- erence is instructive in our model, again illustrated by Wadding-
200/ZEB and Lin28/let7, govern EMT decision making, determining ton’s epigenetic landscape. Just as expression of the Yamanaka
the position of the “stemness window” on the “EMT axis” (Bracken factors in somatic cell reprogramming to pluripotency results in
et al., 2008; Burk et al., 2008; Yang et al., 2010). They showed that expansion of developmental potential, EMT may accomplish a
under different conditions, different types of cells (epithelial, mesen- similar (though less extensive) increase in the number of genes
chymal, or epithelial-mesenchymal hybrid) could gain stemness. available for the cell’s use (Fig. 1). This could allow expression
They demonstrated that a transcription factor that is associated of genes that were turned off in a previous developmental step.
independently with both regulating stemness and EMT, OVOL, not Some possibilities of ways in which EMT might accomplish this
only stabilizes the hybrid E/M phenotype but is also predicted by include opening chromatin (McDonald et al., 2011), direct gene
mathematical models to enable hybrid E/M cells to gain stemness expression effects leading to signaling changes (Cieslik et al.,
(Jolly et al., 2015). EMT processes thus have the potential to convert 2013), and modifying post-transcriptional control of gene
stable, “terminally” differentiated states to metastable states, with expression (Shukla et al., 2015; Unternaehrer et al., 2014).
the associated increase in potential gene expression (Enver et al., In fact, a similar paradigm is emerging in the reprogramming
2009). In support of this model, it is a subset of the E/M cells that field: Sequential EMT-MET is essential for the conversion from
exhibit tumorigenesis and in vitro stem cell activity (Strauss mouse embryonic fibroblasts (MEFs) to neuronal cells (He et al.,
et al., 2011). 2017), and sequential EMT-MET occurs during human ESC (hESC)
Although further studies are needed to test this model, the differentiation into definitive endoderm in vitro, where Snail plays
idea of a moving “stemness window” alongside the epithelial- a key role (Li et al., 2017). This evidence supports our proposal
mesenchymal axis provides an extremely valuable mind set for that early EMT/MET may poise the cells in a state more suitable
future research. Recently, Grosse-Wilde et al. showed that both for further cell fate conversion. Thus, EMT is proposed to power
epithelial and mesenchymal genes can be expressed in the same the “uphill” dedifferentiation illustrated in Waddington’s land-
cell, and that these hybrid E/M cells can be generated from either scape, allowing the “downhill” differentiation that happens in
epithelial cells undergoing partial EMT or mesenchymal cells subsequent steps.
16 WANG AND UNTERNAEHRER

To illustrate one of these proposed mechanisms, as alluded to characterizing and defining the hybrid state will doubtless pro-
previously, EMT transcription factors repress key differentiation- vide useful insights regarding regulation and possible interven-
promoting miRNAs, let-7 and miR-34 (Siemens et al., 2011; tion points.
Yang et al., 2012). Inhibition of these miRNAs results in In this review, we examine the possibility that, although the
increased stemness. Our working hypothesis is that by antago- overall trajectory of developmental processes is toward differen-
nizing let-7, miR-34, and other small RNAs, EMT factors desta- tiation, cells partially and transiently dedifferentiate when they
bilize the differentiated state to allow expression of stemness undergo EMT. This loss of differentiation represents an opportu-
factors. nity for adjusting gene expression programs and allows for new
When Snail and other EMT factors are expressed in develop- gene expression required for formation of new tissue types in
ment, we propose that, in addition to allowing migration and development. Similarly, the transient loss of differentiation seen
invasion required for morphogenesis, their repression of in cancer-associated EMTs provides a window of opportunity for
differentiation-associated miRNAs allows for new gene expres- acquisition of a stem cell phenotype. Thus, tissue stem cells or
sion of the miRNA targets. A theoretical example is cardiac differentiated cells in which EMT transcription factors are active
development, which requires expression of the basic-helix-loop- might simultaneously receive signals leading to loss of key fac-
helix transcription factor Hand1, a let-7 target, as early as tors maintaining differentiation. We propose a mechanism
embryonic day 7.5–9.5 (Roche et al., 2013). By week three whereby factors intrinsic to EMT might also influence differenti-
(in the human), when cardiac development is progressing (Roche ation status.
et al., 2013), let-7 is expressed at high levels (Schulman, 2005),
and in fact let-7 plays an important role in germ layer specifica-
tion (Colas et al., 2012). Consistent with its role as a heterochro- Acknowledgements
nic gene that regulates cell fate choices, let-7 is closely regulated This work was supported by a Grant to Promote Collaboration
to control developmental decisions (Hammell et al., 2009). We and Translation from Loma Linda University (LLU), by LLU start-
DEVELOPMENTAL DYNAMICS

postulate that continued expression of let-7 in cardiac progeni- up funding, and by the Center for Health Disparities and Molec-
tors would prevent expression of Hand1. Accordingly, Snail ular Medicine at LLU. We thank Carlotta Glackin, Nozomi Hojo,
knockout embryoid bodies express Hand1 and other cardiac and Sang Nguyen for thoughtful review of the article; Mindy
genes at lower levels (Lin et al., 2014). During the developmental Lombere for generous help with illustrations; and members of
of EMT(s) leading to cardiac fate, the action of Snail to derepress the Glackin and Unternaehrer labs for insightful discussions.
Hand1 (via let-7 repression) would enable cardiac
differentiation.
Other stemness genes that are let-7 targets are re-expressed References
during development, implying the necessity for repeated cycles
Ahmed N, Thompson EW, Quinn MA. 2007. Epithelial-mesenchymal
of let-7 repression. An example is Sall4, a pluripotency factor interconversions in normal ovarian surface epithelium and ovarian
that is also important for limb, neural tube, heart, and gonad carcinomas: an exception to the norm. J Cell Physiol 213:
development (Tatetsu et al., 2016). 581–588.
Hence, we propose that the action of Snail in cancer may Aigner K, Dampier B, Descovich L, Mikula M, Sultan A, Schreiber M,
bring about a reprogramming event resulting in dedifferentia- Mikulits W, Brabletz T, Strand D, Obrist P, Sommergruber W,
Schweifer N, Wernitznig A, Beug H, Foisner R, Eger A. 2007. The
tion: Snail repression of let-7 and miR34 (among others) allows transcription factor ZEB1 (deltaEF1) promotes tumour cell dedif-
for derepression of pluripotency genes that usher in the stem cell ferentiation by repressing master regulators of epithelial polarity.
state. The resulting cancer stem cells attain self-renewal capabil- Oncogene 26:6979–6988.
ities and are able to differentiate to all cell types represented in Alborzi A, Mac K, Glackin CA, Murray SS, Zernik JH. 1996. Endo-
chondral and intramembranous fetal bone development: osteo-
the tumor. blastic cell proliferation, and expression of alkaline phosphatase,
m-twist, and histone H4. J Craniofac Genet Dev Biol 16:94–106.
Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF.
Perspectives/Conclusion 2003. Prospective identification of tumorigenic breast cancer
Although data showing roles for EMT in both differentiation and cells. Proc Natl Acad Sci USA 100:3983–3988.
Ansieau S, Bastid J, Doreau A, Morel A-P, Bouchet BP, Thomas C,
dedifferentiation appear to be contradictory, a possible explana- Fauvet F, Puisieux I, Doglioni C, Piccinin S, Maestro R, Voeltzel T,
tion that could harmonize the evidence on both sides emphasizes Selmi A, Valsesia-Wittmann S, Caron de Fromentel C, Puisieux A.
the plasticity gained by cells undergoing EMT. 2008. Induction of EMT by twist proteins as a collateral effect of
The fact that EMT occurs periodically during development as tumor-promoting inactivation of premature senescence. Cancer
Cell 14:79–89.
an essential step in formation of many tissues implies that EMT Anton R, Kestler HA, Kühl M. 2007. Beta-catenin signaling contrib-
results in differentiation. We propose that developmental EMTs, utes to stemness and regulates early differentiation in murine
rather than directly resulting in cellular differentiation, allow embryonic stem cells. FEBS Lett 581:5247–5254.
partial dedifferentiation, enabling the next round of differentia- Batlle E, Sancho E, Francí C, Domínguez D, Monfar M, Baulida J,
tion to occur (Battula et al., 2010). In a pathological analogy, García De Herreros A. 2000. The transcription factor snail is a
repressor of E-cadherin gene expression in epithelial tumour cells.
cancer-associated EMTs result in an increase in stemness, allow- Nat Cell Biol 2:84–89.
ing cancer stem cells to self-renew and differentiate to all cell Batlle E, Clevers H. 2017. Cancer stem cells revisited. Nat Med 23:
types represented in the tumor (Polyak and Weinberg, 2009). The 1124–1134.
process of EMT may be a critical step in acquisition of stemness Batlle R, Alba-Castellón L, Loubat-Casanovas J, Armenteros E,
Francí C, Stanisavljevic J, Banderas R, Martin-Caballero J,
properties seen in cancer stem cells. Bonilla F, Baulida J, Casal JI, Gridley T, García de Herreros A.
Evidence is mounting that partial EMTs resulting in hybrid 2013. Snail1 controls TGF-β responsiveness and differentiation of
cells endow cells with stem cell properties. Further studies mesenchymal stem cells. Oncogene 32:3381–3389.
EMT IN DIFFERENTIATION AND DEDIFFERENTIATION 17

Battula VL, Evans KW, Hollier BG, Shi Y, Marini FC, Ayyanan A, Cieslik M, Hoang SA, Baranova N, Chodaparambil S, Kumar M,
Wang R-Y, Brisken C, Guerra R, Andreeff M, Mani SA. 2010. Epi- Allison DF, Xu X, Wamsley JJ, Gray L, Jones DR, Mayo MW,
thelial-mesenchymal transition-derived cells exhibit multilineage Bekiranov S. 2013. Epigenetic coordination of signaling pathways
differentiation potential similar to mesenchymal stem cells. Stem during the epithelial-mesenchymal transition. Epigenetics Chro-
Cells 28:1435–1445. matin 6:28.
Baulida J, García de Herreros A. 2015. Snail1-driven plasticity of Colas AR, McKeithan WL, Cunningham TJ, Bushway PJ,
epithelial and mesenchymal cells sustains cancer malignancy. Garmire LX, Duester G, Subramaniam S, Mercola M. 2012.
Biochim Biophys Acta 1856:55–61. Whole-genome microRNA screening identifies let-7 and mir-18 as
Baum B, Settleman J, Quinlan MP. 2008. Transitions between epi- regulators of germ layer formation during early embryogenesis.
thelial and mesenchymal states in development and disease. Genes Dev 26:2567–2579.
Semin Cell Dev Biol 19:294–308. Comijn J, Berx G, Vermassen P, Verschueren K, van Grunsven L,
Beerling E, Seinstra D, de Wit E, Kester L, van der Velden D, Bruyneel E, Mareel M, Huylebroeck D, van Roy F. 2001. The two-
Maynard C, Schäfer R, van Diest P, Voest E, van Oudenaarden A, handed E box binding zinc finger protein SIP1 downregulates
Vrisekoop N, van Rheenen J. 2016. Plasticity between Epithelial E-cadherin and induces invasion. Mol Cell 7:1267–1278.
and Mesenchymal States Unlinks EMT from Metastasis- Costello I, Pimeisl I-M, Dräger S, Bikoff EK, Robertson EJ,
Enhancing Stem Cell Capacity. Cell Rep 14:2281–2288. Arnold SJ. 2011. The T-box transcription factor Eomesodermin
Bell CE, Watson AJ. 2009. SNAI1 and SNAI2 are asymmetrically acts upstream of Mesp1 to specify cardiac mesoderm during
expressed at the 2-cell stage and become segregated to the TE in mouse gastrulation. Nat Cell Biol 13:1084–1091.
the mouse blastocyst. PloS One 4:e8530. Dahiya N, Sherman-Baust CA, Wang T-L, Davidson B, Shih I-M,
Biddle A, Liang X, Gammon L, Fazil B, Harper LJ, Emich H, Zhang Y, Wood W, Becker KG, Morin PJ. 2008. MicroRNA
Costea DE, Mackenzie IC. 2011. Cancer stem cells in squamous expression and identification of putative miRNA targets in ovarian
cell carcinoma switch between two distinct phenotypes that are cancer. PloS One 3:e2436.
preferentially migratory or proliferative. Cancer Res 71:5317–5326. De Craene B, Gilbert B, Stove C, Bruyneel E, van Roy F, Berx G.
Bolós V, Peinado H, Pérez-Moreno MA, Fraga MF, Esteller M, 2005. The transcription factor snail induces tumor cell invasion
Cano A. 2003. The transcription factor Slug represses E-cadherin through modulation of the epithelial cell differentiation program.
expression and induces epithelial to mesenchymal transitions: a Cancer Res 65:6237–6244.
comparison with Snail and E47 repressors. J Cell Sci 116:499–511. Dong Y-F, Soung DY, Chang Y, Enomoto-Iwamoto M, Paris M,
DEVELOPMENTAL DYNAMICS

Bose M, Hao X, Ju J, Husain A, Park S, Lambert JD, Yang CS. O’Keefe RJ, Schwarz EM, Drissi H. 2007. Transforming growth factor-
2007. Inhibition of tumorigenesis in ApcMin/+ mice by a combina- beta and Wnt signals regulate chondrocyte differentiation through
tion of (-)-epigallocatechin-3-gallate and fish oil. J Agric Food Twist1 in a stage-specific manner. Mol Endocrinol 21:2805–2820.
Chem 55:7695–7700. Dontu G, Abdallah WM, Foley JM, Jackson KW, Clarke MF,
Boyerinas B, Park S-M, Hau A, Murmann AE, Peter ME. 2010. The Kawamura MJ, Wicha MS. 2003. In vitro propagation and tran-
role of let-7 in cell differentiation and cancer. Endocr Relat Cancer scriptional profiling of human mammary stem/progenitor cells.
17:F19-36. Genes Dev 17:1253–1270.
Brabletz T, Jung A, Reu S, Porzner M, Hlubek F, Kunz- Dupin E, Calloni G, Real C, Gonçalves-Trentin A, Le Douarin NM.
Schughart LA, Knuechel R, Kirchner T. 2001. Variable beta- 2007. Neural crest progenitors and stem cells. C R Biol 330:
catenin expression in colorectal cancers indicates tumor 521–529.
progression driven by the tumor environment. Proc Natl Acad Eichmann A, Yuan L, Moyon D, Lenoble F, Pardanaud L, Breant C.
Sci USA 98:10356–10361. 2005. Vascular development: from precursor cells to branched
Bracken CP, Gregory PA, Kolesnikoff N, Bert AG, Wang J, arterial and venous networks. Int J Dev Biol 49:259–267.
Shannon MF, Goodall GJ. 2008. A double-negative feedback loop Enver T, Pera M, Peterson C, Andrews PW. 2009. Stem cell states,
between ZEB1-SIP1 and the microRNA-200 family regulates fates, and the rules of attraction. Cell Stem Cell 4:387–397.
epithelial-mesenchymal transition. Cancer Res 68:7846–7854. Esquela-Kerscher A, Slack FJ. 2006. Oncomirs—microRNAs with a
Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, role in cancer. Nat Rev Cancer 6:259–269.
Spaderna S, Brabletz T. 2008. A reciprocal repression between Essafi A, Webb A, Berry RL, Slight J, Burn SF, Spraggon L,
ZEB1 and members of the miR-200 family promotes EMT and Velecela V, Martinez-Estrada OM, Wiltshire JH, Roberts SGE,
invasion in cancer cells. EMBO Rep 9:582–589. Brownstein D, Davies JA, Hastie ND, Hohenstein P. 2011. A
Cano A, Pérez-Moreno MA, Rodrigo I, Locascio A, Blanco MJ, del wt1-controlled chromatin switching mechanism underpins tissue-
Barrio MG, Portillo F, Nieto MA. 2000. The transcription factor specific wnt4 activation and repression. Dev Cell 21:559–574.
snail controls epithelial-mesenchymal transitions by repressing Fabregat I, Malfettone A, Soukupova J. 2016. New Insights into the
E-cadherin expression. Nat Cell Biol 2:76–83. Crossroads between EMT and Stemness in the Context of Can-
Carmona R, González-Iriarte M, Macías D, Pérez-Pomares JM, cer. J Clin Med 5.
García-Garrido L, Muñoz-Chápuli R. 2000. Immunolocalization of Fan F, Samuel S, Evans KW, Lu J, Xia L, Zhou Y, Sceusi E, Tozzi F,
the transcription factor Slug in the developing avian heart. Anat Ye X-C, Mani SA, Ellis LM. 2012. Overexpression of snail induces
Embryol (Berl) 201:103–109. epithelial-mesenchymal transition and a cancer stem cell-like phe-
Carver EA, Jiang R, Lan Y, Oram KF, Gridley T. 2001. The mouse notype in human colorectal cancer cells. Cancer Med 1:5–16.
snail gene encodes a key regulator of the epithelial-mesenchymal Fehling HJ, Lacaud G, Kubo A, Kennedy M, Robertson S, Keller G,
transition. Mol Cell Biol 21:8184–8188. Kouskoff V. 2003. Tracking mesoderm induction and its specifica-
Charrasse S, Meriane M, Comunale F, Blangy A, Gauthier- tion to the hemangioblast during embryonic stem cell differentia-
Rouvière C. 2002. N-cadherin-dependent cell-cell contact regu- tion. Dev Camb Engl 130:4217–4227.
lates Rho GTPases and beta-catenin localization in mouse C2C12 Forte E, Chimenti I, Rosa P, Angelini F, Pagano F, Calogero A,
myoblasts. J Cell Biol 158:953–965. Giacomello A, Messina E. 2017. EMT/MET at the Crossroad of
Choi YJ, Lin C-P, Ho JJ, He X, Okada N, Bu P, Zhong Y, Kim SY, Stemness, Regeneration and Oncogenesis: The Ying-Yang Equi-
Bennett MJ, Chen C, Ozturk A, Hicks GG, Hannon GJ, He L. librium Recapitulated in Cell Spheroids. Cancers (Basel) 9.
2011. miR-34 miRNAs provide a barrier for somatic cell repro- Frixen UH, Behrens J, Sachs M, Eberle G, Voss B, Warda A,
gramming. Nat Cell Biol 13:1353–1360. Löchner D, Birchmeier W. 1991. E-cadherin-mediated cell-cell
Christ B, Jacob M, Jacob HJ. 1983. On the origin and development adhesion prevents invasiveness of human carcinoma cells. J Cell
of the ventrolateral abdominal muscles in the avian embryo. An Biol 113:173–185.
experimental and ultrastructural study. Anat Embryol (Berl) 166: Galvagni F, Lentucci C, Neri F, Dettori D, De Clemente C,
87–101. Orlandini M, Anselmi F, Rapelli S, Grillo M, Borghi S, Oliviero S.
Chua K-N, Poon KL, Lim J, Sim W-J, Huang RY-J, Thiery JP. 2011. 2015. Snai1 promotes ESC exit from the pluripotency by direct
Target cell movement in tumor and cardiovascular diseases based repression of self-renewal genes. Stem Cells 33:742–750.
on the epithelial-mesenchymal transition concept. Adv Drug Deliv Gammill LS, Bronner-Fraser M. 2003. Neural crest specification:
Rev 63:558–567. migrating into genomics. Nat Rev Neurosci 4:795–805.
18 WANG AND UNTERNAEHRER

Gilks CB, Prat J. 2009. Ovarian carcinoma pathology and genetics: epithelial to mesenchymal transition and increased motility. J Cell
recent advances. Hum Pathol 40:1213–1223. Biol 151:1193–1206.
Gonzalez DM, Medici D. 2014. Signaling mechanisms of the Kim K, Lu Z, Hay ED. 2002. Direct evidence for a role of beta-cate-
epithelial-mesenchymal transition. Sci. Signal. 7:re8. nin/LEF-1 signaling pathway in induction of EMT. Cell Biol Int 26:
Grosse-Wilde A, Fouquier d’Hérouël A, McIntosh E, Ertaylan G, 463–476.
Skupin A, Kuestner RE, del Sol A, Walters K-A, Huang S. 2015. Kim Y-Y, Moon J-S, Kwon M, Shin J, Im S-K, Kim H-A, Han J-K,
Stemness of the hybrid Epithelial/Mesenchymal State in Breast Can- Kong Y-Y. 2014. Meteorin regulates mesendoderm development
cer and Its Association with Poor Survival. PloS One 10:e0126522. by enhancing nodal expression. PloS One 9:e88811.
Guo W, Keckesova Z, Donaher JL, Shibue T, Tischler V, Kong D, Banerjee S, Ahmad A, Li Y, Wang Z, Sethi S, Sarkar FH.
Reinhardt F, Itzkovitz S, Noske A, Zürrer-Härdi U, Bell G, Tam WL, 2010. Epithelial to mesenchymal transition is mechanistically
Mani SA, van Oudenaarden A, Weinberg RA. 2012. Slug and Sox9 linked with stem cell signatures in prostate cancer cells. PloS One
cooperatively determine the mammary stem cell state. Cell 148: 5:e12445.
1015–1028. Lee YS, Kim HK, Chung S, Kim K-S, Dutta A. 2005. Depletion of
Hajra KM, Chen DY-S, Fearon ER. 2002. The SLUG zinc-finger pro- human micro-RNA miR-125b reveals that it is critical for the prolif-
tein represses E-cadherin in breast cancer. Cancer Res 62: eration of differentiated cells but not for the down-regulation of
1613–1618. putative targets during differentiation. J Biol Chem 280:
Hall BK. 2000. The neural crest as a fourth germ layer and verte- 16635–16641.
brates as quadroblastic not triploblastic. Evol Dev 2:3–5. Leptin M, Grunewald B. 1990. Cell shape changes during gastrula-
Hammell CM, Karp X, Ambros V. 2009. A feedback circuit involving tion in Drosophila. Dev Camb Engl 110:73–84.
let-7-family miRNAs and DAF-12 integrates environmental signals Li Q, Hutchins AP, Chen Y, Li S, Shan Y, Liao B, Zheng D, Shi X,
and developmental timing in Caenorhabditis elegans. Proc Natl Li Y, Chan W-Y, Pan G, Wei S, Shu X, Pei D. 2017. A sequential
Acad Sci USA 106:18668–18673. EMT-MET mechanism drives the differentiation of human embry-
Hay ED. 1995. An overview of epithelio-mesenchymal transforma- onic stem cells towards hepatocytes. Nat Commun 8:15166.
tion. Acta Anat (Basel) 154:8–20. Li X, Zhang Z, Zhang Y, Cao Y, Wei H, Wu Z. 2018. Upregulation of
Hazan RB, Kang L, Whooley BP, Borgen PI. 1997. N-cadherin pro- lactate-inducible snail protein suppresses oncogene-mediated
motes adhesion between invasive breast cancer cells and the senescence through p16INK4a inactivation. J Exp Clin Cancer
stroma. Cell Adhes Commun 4:399–411. Res CR 37:39.
DEVELOPMENTAL DYNAMICS

He H, Chen W, Wang X, Wang C, Liu F, Shen Z, Xu J, Gu J, Sun Y. Liao T-T, Yang M-H. 2017. Revisiting epithelial-mesenchymal transi-
2012. Snail is an independent prognostic predictor for progression tion in cancer metastasis: the connection between epithelial plas-
and patient survival of gastric cancer. Cancer Sci 103:1296–1303. ticity and stemness. Mol Oncol 11:792–804.
He L, He X, Lim LP, de Stanchina E, Xuan Z, Liang Y, Xue W, Lim J, Thiery JP. 2012. Epithelial-mesenchymal transitions: insights
Zender L, Magnus J, Ridzon D, Jackson AL, Linsley PS, Chen C, from development. Development 139:3471–3486.
Lowe SW, Cleary MA, Hannon GJ. 2007. A microRNA component Lin Y, Li X-Y, Willis AL, Liu C, Chen G, Weiss SJ. 2014.
of the p53 tumour suppressor network. Nature 447:1130–1134. Snail1-dependent control of embryonic stem cell pluripotency and
He S, Chen J, Zhang Y, Zhang M, Yang X, Li Y, Sun H, Lin L, Fan K, lineage commitment. Nat Commun 5:3070.
Liang L, Feng C, Wang F, Zhang X, Guo Y, Pei D, Zheng H. 2017. Lindsley RC, Gill JG, Kyba M, Murphy TL, Murphy KM. 2006.
Sequential EMT-MET induces neuronal conversion through Sox2. Canonical Wnt signaling is required for development of embryonic
Cell Discov 3:17017. stem cell-derived mesoderm. Dev Camb Engl 133:3787–3796.
Hebrok M, Wertz K, Füchtbauer EM. 1994. M-twist is an inhibitor of Liu S, Cong Y, Wang D, Sun Y, Deng L, Liu Y, Martin-Trevino R,
muscle differentiation. Dev Biol 165:537–544. Shang L, McDermott SP, Landis MD, Hong S, Adams A,
Hochedlinger K, Plath K. 2009. Epigenetic reprogramming and D’Angelo R, Ginestier C, Charafe-Jauffret E, Clouthier SG,
induced pluripotency. Dev Camb Engl 136:509–523. Birnbaum D, Wong ST, Zhan M, Chang JC, Wicha MS. 2014. Breast
Hollier BG, Tinnirello AA, Werden SJ, Evans KW, Taube JH, cancer stem cells transition between epithelial and mesenchymal
Sarkar TR, Sphyris N, Shariati M, Kumar SV, Battula VL, states reflective of their normal counterparts. Stem Cell Rep 2:78–91.
Herschkowitz JI, Guerra R, Chang JT, Miura N, Rosen JM, Liu X, Sun H, Qi J, Wang L, He S, Liu J, Feng C, Chen C, Li W,
Mani SA. 2013. FOXC2 expression links epithelial-mesenchymal Guo Y, Qin D, Pan G, Chen J, Pei D, Zheng H. 2013. Sequential
transition and stem cell properties in breast cancer. Cancer Res introduction of reprogramming factors reveals a time-sensitive
73:1981–1992. requirement for individual factors and a sequential EMT-MET
Hwang W-L, Jiang J-K, Yang S-H, Huang T-S, Lan H-Y, Teng H-W, mechanism for optimal reprogramming. Nat Cell Biol 15:829–838.
Yang C-Y, Tsai Y-P, Lin C-H, Wang H-W, Yang M-H. 2014. Micro- Lluis F, Pedone E, Pepe S, Cosma MP. 2008. Periodic activation of
RNA-146a directs the symmetric division of Snail-dominant colo- Wnt/beta-catenin signaling enhances somatic cell reprogramming
rectal cancer stem cells. Nat Cell Biol 16:268–280. mediated by cell fusion. Cell Stem Cell 3:493–507.
Hwang W-L, Yang M-H, Tsai M-L, Lan H-Y, Su S-H, Chang S-C, Logullo AF, Nonogaki S, Pasini FS, Osório CABDT, Soares FA,
Teng H-W, Yang S-H, Lan Y-T, Chiou S-H, Wang H-W. 2011. Brentani MM. 2010. Concomitant expression of epithelial-
SNAIL regulates interleukin-8 expression, stem cell-like activity, mesenchymal transition biomarkers in breast ductal carcinoma:
and tumorigenicity of human colorectal carcinoma cells. Gastro- association with progression. Oncol Rep 23:313–320.
enterology 141:279–291. Lomelí H, Starling C, Gridley T. 2009. Epiblast-specific Snai1 dele-
Isenmann S, Arthur A, Zannettino ACW, Turner JL, Shi S, tion results in embryonic lethality due to multiple vascular defects.
Glackin CA, Gronthos S. 2009. TWIST family of basic helix-loop- BMC Res Notes 2:22.
helix transcription factors mediate human mesenchymal stem cell Maherali N, Hochedlinger K. 2009. Tgfbeta signal inhibition cooper-
growth and commitment. Stem Cells 27:2457–2468. ates in the induction of iPSCs and replaces Sox2 and cMyc. Curr
Islam S, Carey TE, Wolf GT, Wheelock MJ, Johnson KR. 1996. Biol 19:1718–1723.
Expression of N-cadherin by human squamous carcinoma cells Mani SA, Guo W, Liao M-J, Eaton EN, Ayyanan A, Zhou AY,
induces a scattered fibroblastic phenotype with disrupted cell-cell Brooks M, Reinhard F, Zhang CC, Shipitsin M, Campbell LL,
adhesion. J Cell Biol 135:1643–1654. Polyak K, Brisken C, Yang J, Weinberg RA. 2008. The epithelial-
Jolly MK, Huang B, Lu M, Mani SA, Levine H, Ben-Jacob E. 2014. mesenchymal transition generates cells with properties of stem
Towards elucidating the connection between epithelial- cells. Cell 133:704–715.
mesenchymal transitions and stemness. J R Soc Interface 11: Manning DL, Robertson JF, Ellis IO, Elston CW, McClelland RA,
20140962. Gee JM, Jones RJ, Green CD, Cannon P, Blamey RW. 1994. Oes-
Jolly MK, Jia D, Boareto M, Mani SA, Pienta KJ, Ben-Jacob E, trogen-regulated genes in breast cancer: association of pLIV1
Levine H. 2015. Coupling the modules of EMT and stemness: A with lymph node involvement. Eur J Cancer 30A:675–678.
tunable “stemness window” model. Oncotarget 6:25161–25174. Marin F, Nieto MA. 2004. Expression of chicken slug and snail in
Kim JB, Islam S, Kim YJ, Prudoff RS, Sass KM, Wheelock MJ, mesenchymal components of the developing central nervous sys-
Johnson KR. 2000. N-Cadherin extracellular repeat 4 mediates tem. Dev Dyn 230:144–148.
EMT IN DIFFERENTIATION AND DEDIFFERENTIATION 19

Marson A, Foreman R, Chevalier B, Bilodeau S, Kahn M, Young RA, Romano LA, Runyan RB. 1999. Slug is a mediator of epithelial-
Jaenisch R. 2008. Wnt signaling promotes reprogramming of mesenchymal cell transformation in the developing chicken heart.
somatic cells to pluripotency. Cell Stem Cell 3:132–135. Dev Biol 212:243–254.
Martin-Belmonte F, Perez-Moreno M. 2011. Epithelial cell polarity, Roy HK, Iversen P, Hart J, Liu Y, Koetsier JL, Kim Y, Kunte DP,
stem cells and cancer. Nat Rev Cancer 12:23–38. Madugula M, Backman V, Wali RK. 2004. Down-regulation of
McDonald OG, Wu H, Timp W, Doi A, Feinberg AP. 2011. Genome- SNAIL suppresses MIN mouse tumorigenesis: modulation of apo-
scale epigenetic reprogramming during epithelial-to-mesenchymal ptosis, proliferation, and fractal dimension. Mol Cancer Ther 3:
transition. Nat Struct Mol Biol 18:867–874. 1159–1165.
Melton C, Blelloch R. 2010. MicroRNA Regulation of Embryonic Safa AR. 2016. Resistance to Cell Death and Its Modulation in Can-
Stem Cell Self-Renewal and Differentiation. Adv Exp Med Biol cer Stem Cells. Crit Rev Oncog 21:203–219.
695:105–117. Samavarchi-Tehrani P, Golipour A, David L, Sung H-K, Beyer TA,
Melton C, Judson RL, Blelloch R. 2010. Opposing microRNA fami- Datti A, Woltjen K, Nagy A, Wrana JL. 2010. Functional genomics
lies regulate self-renewal in mouse embryonic stem cells. Nature reveals a BMP-driven mesenchymal-to-epithelial transition in the
463:621–626. initiation of somatic cell reprogramming. Cell Stem Cell 7:64–77.
Micalizzi DS, Farabaugh SM, Ford HL. 2010. Epithelial- Santisteban M, Reiman JM, Asiedu MK, Behrens MD, Nassar A,
mesenchymal transition in cancer: parallels between normal Kalli KR, Haluska P, Ingle JN, Hartmann LC, Manjili MH,
development and tumor progression. J. Mammary Gland Biol Radisky DC, Ferrone S, Knutson KL. 2009. Immune-induced epi-
Neoplasia 15:117–134. thelial to mesenchymal transition in vivo generates breast cancer
Morel A-P, Lièvre M, Thomas C, Hinkal G, Ansieau S, Puisieux A. stem cells. Cancer Res 69:2887–2895.
2008. Generation of breast cancer stem cells through epithelial- Schäfer G, Narasimha M, Vogelsang E, Leptin M. 2014. Cadherin
mesenchymal transition. PloS One 3:e2888. switching during the formation and differentiation of the Drosoph-
Murray SA, Gridley T. 2006. Snail family genes are required for left- ila mesoderm—implications for epithelial-to-mesenchymal transi-
right asymmetry determination, but not neural crest formation, in tions. J Cell Sci 127:1511–1522.
mice. Proc Natl Acad Sci USA 103:10300–10304. Schatton T, Murphy GF, Frank NY, Yamaura K, Waaga-Gasser AM,
Nakajima Y, Yamagishi T, Hokari S, Nakamura H. 2000. Mecha- Gasser M, Zhan Q, Jordan S, Duncan LM, Weishaupt C,
nisms involved in valvuloseptal endocardial cushion formation in Fuhlbrigge RC, Kupper TS, Sayegh MH, Frank MH. 2008. Identifi-
early cardiogenesis: roles of transforming growth factor (TGF)- cation of cells initiating human melanomas. Nature 451:345–349.
DEVELOPMENTAL DYNAMICS

beta and bone morphogenetic protein (BMP). Anat Rec 258: Schipper JH, Frixen UH, Behrens J, Unger A, Jahnke K,
119–127. Birchmeier W. 1991. E-cadherin expression in squamous cell carci-
Nieto MA. 2013. Epithelial plasticity: a common theme in embryonic nomas of head and neck: inverse correlation with tumor dedifferen-
and cancer cells. Science 342:1234850. tiation and lymph node metastasis. Cancer Res 51:6328–6337.
Nikitina N, Sauka-Spengler T, Bronner-Fraser M. 2008. Dissecting Schulman A. 2005. The search for alternative sources of human plu-
early regulatory relationships in the lamprey neural crest gene net- ripotent stem cells. Stem Cell Rev 1:291–292.
work. Proc Natl Acad Sci USA 105:20083–20088. Sharabi AB, Aldrich M, Sosic D, Olson EN, Friedman AD, Lee S-H,
Nitzan E, Kalcheim C. 2013. Neural crest and somitic mesoderm as Chen S-Y. 2008. Twist-2 controls myeloid lineage development
paradigms to investigate cell fate decisions during development. and function. PLoS Biol 6:e316.
Dev Growth Differ 55:60–78. Sheridan C, Kishimoto H, Fuchs RK, Mehrotra S, Bhat-Nakshatri P,
Ocaña OH, Córcoles R, Fabra A, Moreno-Bueno G, Acloque H, Turner CH, Goulet R, Badve S, Nakshatri H. 2006. CD44+/CD24-
Vega S, Barrallo-Gimeno A, Cano A, Nieto MA. 2012. Metastatic breast cancer cells exhibit enhanced invasive properties: an early
colonization requires the repression of the epithelial-mesenchymal step necessary for metastasis. Breast Cancer Res 8:R59.
transition inducer Prrx1. Cancer Cell 22:709–724. Shukla P, Vogl C, Wallner B, Rigler D, Müller M, Macho-Maschler S.
Oda H, Tsukita S, Takeichi M. 1998. Dynamic behavior of the 2015. High-throughput mRNA and miRNA profiling of epithelial-
cadherin-based cell-cell adhesion system during Drosophila gas- mesenchymal transition in MDCK cells. BMC Genomics 16:944.
trulation. Dev Biol 203:435–450. Siemens H, Jackstadt R, Hünten S, Kaller M, Menssen A, Götz U,
O’Hara AJ, Wang L, Dezube BJ, Harrington WJ, Damania B, Hermeking H. 2011. miR-34 and SNAIL form a double-negative
Dittmer DP. 2009. Tumor suppressor microRNAs are underrepre- feedback loop to regulate epithelial-mesenchymal transitions. Cell
sented in primary effusion lymphoma and Kaposi sarcoma. Blood Cycle Georget Tex 10:4256–4271.
113:5938–5941. Sleeman KE, Kendrick H, Ashworth A, Isacke CM, Smalley MJ.
Pan D, Fujimoto M, Lopes A, Wang Y-X. 2009. Twist-1 is a 2006. CD24 staining of mouse mammary gland cells defines lumi-
PPARdelta-inducible, negative-feedback regulator of PGC-1alpha nal epithelial, myoepithelial/basal and non-epithelial cells. Breast
in brown fat metabolism. Cell 137:73–86. Cancer Res 8:R7.
Park I-H, Zhao R, West JA, Yabuuchi A, Huo H, Ince TA, Lerou PH, Šošic D, Richardson JA, Yu K, Ornitz DM, Olson EN. 2003. Twist
Lensch MW, Daley GQ. 2008. Reprogramming of human somatic Regulates Cytokine Gene Expression through a Negative Feed-
cells to pluripotency with defined factors. Nature 451:141–146. back Loop that Represses NF-κB Activity. Cell 112:169–180.
Peinado H, Olmeda D, Cano A. 2007. Snail, Zeb and bHLH factors Steventon B, Carmona-Fontaine C, Mayor R. 2005. Genetic network
in tumour progression: an alliance against the epithelial pheno- during neural crest induction: from cell specification to cell sur-
type? Nat Rev Cancer 7:415–428. vival. Semin Cell Dev Biol 16:647–654.
Pérez-Pomares JM, Muñoz-Chápuli R. 2002. Epithelial- Strauss R, Li Z-Y, Liu Y, Beyer I, Persson J, Sova P, Möller T,
mesenchymal transitions: a mesodermal cell strategy for evolutive Pesonen S, Hemminki A, Hamerlik P, Drescher C, Urban N,
innovation in Metazoans. Anat Rec 268:343–351. Bartek J, Lieber A. 2011. Analysis of epithelial and mesenchymal
Polyak K, Weinberg RA. 2009. Transitions between epithelial and markers in ovarian cancer reveals phenotypic heterogeneity and
mesenchymal states: acquisition of malignant and stem cell traits. plasticity. PloS One 6:e16186.
Nat Rev Cancer 9:265–273. Swetha G, Chandra V, Phadnis S, Bhonde R. 2011. Glomerular pari-
Prasad CP, Rath G, Mathur S, Bhatnagar D, Parshad R, Ralhan R. etal epithelial cells of adult murine kidney undergo EMT to gener-
2009. Expression analysis of E-cadherin, Slug and GSK3beta in ate cells with traits of renal progenitors. J Cell Mol Med 15:
invasive ductal carcinoma of breast. BMC Cancer 9:325. 396–413.
Pu Q, Patel K, Huang R. 2015. The lateral plate mesoderm: a novel Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K,
source of skeletal muscle. Results Probl Cell Differ 56:143–163. Yamanaka S. 2007. Induction of pluripotent stem cells from adult
Reya T, Clevers H. 2005. Wnt signalling in stem cells and cancer. human fibroblasts by defined factors. Cell 131:861–872.
Nature 434:843–850. Takahashi K, Yamanaka S. 2006. Induction of pluripotent stem cells
Roche J, Nasarre P, Gemmill R, Baldys A, Pontis J, Korch C, from mouse embryonic and adult fibroblast cultures by defined
Guilhot J, Ait-Si-Ali S, Drabkin H. 2013. Global Decrease of His- factors. Cell 126:663–676.
tone H3K27 Acetylation in ZEB1-Induced Epithelial to Mesenchy- Takahashi K, Yamanaka S. 2015. A developmental framework for
mal Transition in Lung Cancer Cells. Cancers 5:334–356. induced pluripotency. Dev Camb Engl 142:3274–3285.
20 WANG AND UNTERNAEHRER

Takamizawa J, Konishi H, Yanagisawa K, Tomida S, Osada H, Waddington CH. 1957. The strategy of the genes: a discussion of
Endoh H, Harano T, Yatabe Y, Nagino M, Nimura Y, Mitsudomi T, some aspects of theoretical biology. London: Allen & Unwin.
Takahashi T. 2004. Reduced expression of the let-7 microRNAs in 262 p.
human lung cancers in association with shortened postoperative Wahl GM, Spike BT. 2017. Cell state plasticity, stem cells, EMT, and
survival. Cancer Res 64:3753–3756. the generation of intra-tumoral heterogeneity. NPJ Breast Cancer
Tan TZ, Miow QH, Huang RY-J, Wong MK, Ye J, Lau JA, Wu MC, 3:14.
Bin Abdul Hadi LH, Soong R, Choolani M, Davidson B, Wang Y, Baskerville S, Shenoy A, Babiarz JE, Baehner L, Blelloch R.
Nesland JM, Wang L-Z, Matsumura N, Mandai M, Konishi I, 2008. Embryonic stem cell-specific microRNAs regulate the G1-S
Goh B-C, Chang JT, Thiery JP, Mori S. 2013. Functional geno- transition and promote rapid proliferation. Nat Genet 40:
mics identifies five distinct molecular subtypes with clinical rele- 1478–1483.
vance and pathways for growth control in epithelial ovarian Wang Y, Hu X, Greshock J, Shen L, Yang X, Shao Z, Liang S,
cancer. EMBO Mol Med 5:1051–1066. Tanyi JL, Sood AK, Zhang L. 2012. Genomic DNA copy-number
Tan TZ, Miow QH, Miki Y, Noda T, Mori S, Huang RY-J, Thiery JP. alterations of the let-7 family in human cancers. PloS One 7:
2014. Epithelial-mesenchymal transition spectrum quantification e44399.
and its efficacy in deciphering survival and drug responses of can- Wang Z, Li Y, Kong D, Banerjee S, Ahmad A, Azmi AS, Ali S,
cer patients. EMBO Mol Med 6:1279–1293. Abbruzzese JL, Gallick GE, Sarkar FH. 2009. Acquisition of
Tatetsu H, Kong NR, Chong G, Amabile G, Tenen DG, Chai L. 2016. epithelial-mesenchymal transition phenotype of gemcitabine-
SALL4, the missing link between stem cells, development and resistant pancreatic cancer cells is linked with activation of the
cancer. Gene 584:111–119. notch signaling pathway. Cancer Res 69:2400–2407.
ten Berge D, Koole W, Fuerer C, Fish M, Eroglu E, Nusse R. 2008. Wellner U, Schubert J, Burk UC, Schmalhofer O, Zhu F, Sonntag A,
Wnt signaling mediates self-organization and axis formation in Waldvogel B, Vannier C, Darling D, zur Hausen A, Brunton VG,
embryoid bodies. Cell Stem Cell 3:508–518. Morton J, Sansom O, Schüler J, Stemmler MP, Herzberger C,
ten Berge D, Kurek D, Blauwkamp T, Koole W, Maas A, Eroglu E, Hopt U, Keck T, Brabletz S, Brabletz T. 2009. The EMT-activator
Siu RK, Nusse R. 2011. Embryonic stem cells require Wnt pro- ZEB1 promotes tumorigenicity by repressing stemness-inhibiting
teins to prevent differentiation to epiblast stem cells. Nat Cell Biol microRNAs. Nat Cell Biol 11:1487–1495.
13:1070–1075. Weston JA, Thiery JP. 2015. Pentimento: Neural Crest and the origin
Thiery JP. 2002. Epithelial-mesenchymal transitions in tumour pro- of mesectoderm. Dev Biol 401:37–61.
DEVELOPMENTAL DYNAMICS

gression. Nat Rev Cancer 2:442–454. Wielenga VJ, Smits R, Korinek V, Smit L, Kielman M, Fodde R,
Thiery JP, Acloque H, Huang RYJ, Nieto MA. 2009. Epithelial- Clevers H, Pals ST. 1999. Expression of CD44 in Apc and Tcf
mesenchymal transitions in development and disease. Cell 139: mutant mice implies regulation by the WNT pathway. Am J Pathol
871–890. 154:515–523.
Thiery JP, Sleeman JP. 2006. Complex networks orchestrate Wu S-Y, McClay DR. 2007. The Snail repressor is required for PMC
epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol 7: ingression in the sea urchin embryo. Dev Camb Engl 134:
131–142. 1061–1070.
Tsai JH, Donaher JL, Murphy DA, Chau S, Yang J. 2012. Spatiotem- Yang J, Mani SA, Donaher JL, Ramaswamy S, Itzykson RA,
poral regulation of epithelial-mesenchymal transition is essential Come C, Savagner P, Gitelman I, Richardson A, Weinberg RA.
for squamous cell carcinoma metastasis. Cancer Cell 22:725–736. 2004. Twist, a master regulator of morphogenesis, plays an
Tsutsumi S, Yanagawa T, Shimura T, Kuwano H, Raz A. 2004. Auto- essential role in tumor metastasis. Cell 117:927–939.
crine motility factor signaling enhances pancreatic cancer metas- Yang W-H, Lan H-Y, Huang C-H, Tai S-K, Tzeng C-H, Kao S-Y,
tasis. Clin Cancer Res 10:7775–7784. Wu K-J, Hung M-C, Yang M-H. 2012. RAC1 activation mediates
Ullmann U, In’t Veld P, Gilles C, Sermon K, De Rycke M, Van de Twist1-induced cancer cell migration. Nat Cell Biol 14:366–374.
Velde H, Van Steirteghem A, Liebaers I. 2007. Epithelial- Yang X, Lin X, Zhong X, Kaur S, Li N, Liang S, Lassus H, Wang L,
mesenchymal transition process in human embryonic stem cells Katsaros D, Montone K, Zhao X, Zhang Y, Bützow R, Coukos G,
cultured in feeder-free conditions. Mol Hum Reprod 13:21–32. Zhang L. 2010. Double-negative feedback loop between repro-
Unternaehrer JJ, Zhao R, Kim K, Cesana M, Powers JT, gramming factor LIN28 and microRNA let-7 regulates aldehyde
Ratanasirintrawoot S, Onder T, Shibue T, Weinberg RA, Daley GQ. dehydrogenase 1-positive cancer stem cells. Cancer Res 70:
2014. The epithelial-mesenchymal transition factor SNAIL paradoxi- 9463–9472.
cally enhances reprogramming. Stem Cell Rep 3:691–698. Ye X, Tam WL, Shibue T, Kaygusuz Y, Reinhardt F, Ng Eaton E,
Vega S, Morales AV, Ocaña OH, Valdés F, Fabregat I, Nieto MA. Weinberg RA. 2015. Distinct EMT programs control normal mam-
2004. Snail blocks the cell cycle and confers resistance to cell mary stem cells and tumour-initiating cells. Nature 525:256–260.
death. Genes Dev 18:1131–1143. Yook JI, Li X-Y, Ota I, Hu C, Kim HS, Kim NH, Cha SY, Ryu JK,
Vesuna F, van Diest P, Chen JH, Raman V. 2008. Twist is a tran- Choi YJ, Kim J, Fearon ER, Weiss SJ. 2006. A Wnt-
scriptional repressor of E-cadherin gene expression in breast can- Axin2-GSK3beta cascade regulates Snail1 activity in breast can-
cer. Biochem Biophys Res Commun 367:235–241. cer cells. Nat Cell Biol 8:1398–1406.
Vesuna F, Lisok A, Kimble B, Raman V. 2009. Twist modulates Yu J, Vodyanik MA, Smuga-Otto K, Antosiewicz-Bourget J,
breast cancer stem cells by transcriptional regulation of CD24 Frane JL, Tian S, Nie J, Jonsdottir GA, Ruotti V, Stewart R,
expression. Neoplasia 11:1318–1328. Slukvin II, Thomson JA. 2007. Induced pluripotent stem cell lines
ovac L, Aplin JD. 1996. Epithelial-mesenchymal transition during
Vic derived from human somatic cells. Science 318:1917–1920.
trophoblast differentiation. Acta Anat (Basel) 156:202–216. Zhang P, Sun Y, Ma L. 2015. ZEB1: at the crossroads of epithelial-
Viswanathan SR, Daley GQ, Gregory RI. 2008. Selective blockade of mesenchymal transition, metastasis and therapy resistance. Cell
microRNA processing by Lin28. Science 320:97–100. Cycle Georget Tex 14:481–487.

You might also like