Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2018 Dec 1.
Published in final edited form as: J Cell Physiol. 2017 Apr 10;232(12):3261–3272. doi: 10.1002/jcp.25797

Epithelial-mesenchymal transition (EMT): A biological process in the development, stem cell differentiation and tumorigenesis

Tong Chen 1,4,*, Yanan You 2,4, Hua Jiang 2, Zack Z Wang 3,*
PMCID: PMC5507753  NIHMSID: NIHMS843471  PMID: 28079253

Abstract

The lineage transition between epithelium and mesenchyme is a process known as epithelial-mesenchymal transition (EMT), by which polarized epithelial cells lose their adhesion property and obtain mesenchymal cell phenotypes. EMT is a biological process that is often involved in embryogenesis and diseases, such as cancer invasion and metastasis. The EMT and the reverse process, mesenchymal-epithelial transition (MET), also play important roles in stem cell differentiation and de-differentiation (or reprogramming). In this review, we will discuss current research progress of EMT in embryonic development, cellular differentiation and reprogramming, and cancer progression, all of which are representative models for researches of stem cell biology in normal and in diseases. Understanding of EMT and MET may help to identify specific markers to distinguish normal stem cells from cancer stem cells in future.

Keywords: EMT, MET, stem cells, reprogramming, cancer stem cells

1. Introduction

Pluripotent stem cells (PSCs) possess a pluripotent potential to give rise to cell types in three germ lines. PSC differentiation or reprogramming to generate induced pluripotent stem cells (iPSCs) is related to the changes of cellular morphology and surface markers, orchestrated by tremendous molecular regulating processes accompanying with the cellular mobility changes. Transition between epithelial features and mesenchymal counterparts has been documented existing in either pluripotent stem cells or differentiated cells. The epithelium is characterized by apical-basal polarity and tight cell-cell junctions between neighboring epithelial cells, which is associated with cell integrity and stability (Pasti and Labouesse, 2014). On the contrary, mesenchymal cells are identified as fibroblast-like, elongated, and spindle-shaped in appearance by loose cell-cell interaction without tight intracellular adhesion, resulting in more mobility to increase cell migration and anchorage distantly (Pittenger and Martin, 2004). By morphological transition, cells obtain motility to migrate into their organogenetic destinations while de-mobility enables the cells to colonize in site, which is a common mission in stem cell fate (Kavanagh et al., 2014).

The phenomenon that epithelial cells acquire mesenchymal traits, termed as epithelial-mesenchymal transition (EMT), has been observed in physiological and pathological processes, including embryogenesis (Gros and Tabin, 2014; Yoshino et al., 2014), inflammation (Correa-Costa et al., 2014), fibrosis (Grande et al., 2015; Lovisa et al., 2015; Schneider et al., 2012), wound healing (Banerjee et al., 2015), and cancer progression (Ruscetti et al., 2015; Wei et al., 2015). This morphology-related plasticity is also a common theme in carcinogenesis where cancer-initiating cells generate and invade. There are three types of EMT process, named as EMT-Type 1, 2, 3, being proposed to relate to embryo formation, tissue regeneration and cancer progression (Kalluri and Weinberg, 2009), whereas the EMT standing at the stage of embryonic development is sequentially divided as primary, secondary and tertiary segments, representing early gastrulation, mesodermal development, and endocardium morphogenesis, respectively (Thiery et al., 2009).

Loss of E-cadherin expression is considered a key event in an EMT where the cell-cell contacting modulators, cell polarity-related cytoskeleton and extracellular matrix are involved (Puisieux et al., 2014). Since first described in 1982 (Greenburg and Hay, 1982), the EMT has been documented to be regulated by various transcription factors (TFs) (Galvan et al., 2015; Wei et al., 2015; Yu et al., 2015), small non-coding RNA (Dang et al., 2015; Zhou et al., 2015), epigenetic modulators (Choi et al., 2015a; Cicchini et al., 2015), and exogenous inducers (An et al., 2015; Buczek et al., 2015; Choi et al., 2015b; Hwang-Verslues et al., 2013; Park et al., 2015; Zavadil et al., 2001). Families of the zinc-finger proteins Snails (Snai1, Snai2/Slug, Snai3/Smuc), the E-box-binding proteins Zebs (Zeb1/Tcf8, Zeb2/Sip1), the basic helix-loop-helix protein Twists (Twist1, Twist2), and the forkhead box proteins FOXCs (FOXC1, FOXC2) are major TFs that repress the expression of E-cadherin directly (Puisieux et al., 2014). These molecular regulators form an intricate network to mediate the cellular conversion in processes of development and pathologies, among which stem cells lead an important role (Taube et al., 2010).

However, in the studies of relationship between epithelial/mesenchymal lineage and stem cells, whether the EMT or its reversal process, mesenchymal-epithelial transition (MET), enables cells to obtain the pluripotency still remains controversial. Clarification of the relationship between cellular transformation and pluripotent characters will shed light on harnessing cell fate and provide a clue to determine disease status. This review mostly addresses the events in embryo development that is involve in epithelial and mesenchymal lineage transition, and discuss its relevance in stem cell differentiation and reprogramming in normal and diseases.

2. The EMT in embryo development

Fertilized oocytes are a type of homozygous stem cells that have potential to generate all tissue types in development (Lin et al., 2003). Accompanied with its implantation into uterus and differentiation process, the monoplast generates triploblastic body, which is composed of differentiated cell clusters and experiences cellular changes to satisfy the environment stress and the request for survival. Mapping the journey of morphological variety from totipotency to lineage specification will help to explore the relevance in cellular homostasis and tissue morphogenesis.

2.1. The mesenchymal transition in embryo implantation and pre-implantation

Primary EMT takes place in the cells that never undergo the cellular aspect changes, and is confined in the early embryo developmental segments, including embryo implantation and early gastrulation (Figure 1) (Acloque et al., 2009; Thiery et al., 2009). The mammalian embryogenesis is a self-organizing process that is determined not only by maternal distributed factors but also by the cellular interaction within the embryo. A zygote undergoes cellular cleavage to establish the apolar inner cell mass (ICM) and the polar trophectoderm (TE), the first two lineages that appears in the blastocyst stage localizing at inside and outside of the embryo, respectively. Differentiation of the ICM and the TE is initiated from late 8-cell stage when the cells are compacted to assemble conjunctional complex in a distinguishable apical-basal polarized layer. The ICM will give rise to the embryo and extraembryo tissue, while TE contributes to the placenta to provide the interacting signaling between fetus and maternal body (Saiz and Plusa, 2013).

Figure 1. Schematic relationship between embryonic differentiation and E-M transition.

Figure 1

Embryonic differentiation is triggered after the oocyte is fertilized. Primary epithelial (blue)-mesenchymal (red) transition takes place in the cells that never undergo the cellular aspect changes, which is confined in the early embryo developmental segments, including embryo implantation and early gastrulation. During gastrulation, the cells undergoing EMT contribute to ingressive mesoderm formation. Differentiation of ESC/iPSCs, an ex vivo embryonic cellular model, generates mesodermal characterized cells inside of embryonic body (EBs), whereas reprogramming of fibroblast to iPSCs undergoes the counterpart of EMT, the MET. The pluripotency and cellular epithelial/mesenchymal characterization is showed along with in vivo and ex vivo embryonic differentiation flow.

The embryo implantation process involves blastocyst migration, apposition, attachment, adhesion, and invasion into the epithelial lining of the endometrium, where it is classified as an immediate reception in rodents/primates and a pre-receptive phase in other domestic animals (Bowen and Burghardt, 2000). Differentiation of TE lineage is symbolized by Cdx2 expression in outer cells, a specific gene for trophectoderm formation, which co-expresses with a pluripotent POU-family transcription factor Oct3/4 in a reciprocal repressive model (Toyooka et al., 2016). Upregulation of Cdx2 requiring to switch off Oct3/4 indicates that establishment of TE is the first differentiation event in mammalian embryogenesis (Niwa et al., 2005; Strumpf et al., 2005). The sequential superficial/central implantation in ruminant species having a prolonged pre-attachment period provides a window to look into the molecular and cellular changes during peri-attachment periods. The TE in pre-implanted bovine conceptuses was found to express epithelial cytokeratin as well as mesenchymal vimentin and N-cadherin. The EMT-related transcripts, SNALs, ZEBs and TWISTs, were upregulated in pre-implanted conceptuses of day 22, compared to those in day 17 and day 20 conceptuses (Yamakoshi et al., 2012). Loss of E-cadherin, an epithelial adhesion molecule, is associated with invasive phenotype of extravillous trophoblasts, while a reduction in N-cadherin, the mesenchymal adhesion molecule, decreased the invasive capacity of human trophoblast cells (Duzyj et al., 2015; Ng et al., 2012). Interestingly, SNAL1 and SNAL2 are expressed not in inner cells but in outer cells at 2-cell to 8-cell of blastocyst, indicating that the implantation process for noninvasive early-stage trophoblasts requires asymmetrically partial EMT to have special extracellular matrix expression as well (Bell and Watson, 2009).

The significance of the epiblast as epithelial integrity is associated with the selective counteracting mechanical stress and is unique to the early development of amniotes (Sheng, 2015). The polarity-dependent and position-dependent models are both associated with the cell fate segregation in mammal embryos (Saiz and Plusa, 2013; Sasaki, 2010). Cellular localization in murine embryos are related to the expression of transcription factors that are critical for cell differentiation (Toyooka et al., 2016). E-cadherin was showed to be important to ICM compaction and inner-outer lineage segregation. Lacking E-cadherin in embryo resulted in impaired cell adhesion, delayed compaction and disorganized cell allocation, indicating that it is rather epithelial cell-cell interaction than mesenchymal phenotype acting to anchor intracellular signaling in the embryo preimplantation stage (Bessonnard et al., 2015). Even though the development prior to the appearance of pre-gastrulation epiblast is variable in different species, a fully-epithelialized, unilaminar epiblast is a conserved model of start point in embryogenesis of all amniotes.

2.2. Primary EMT in early gastrulation

Gastrulation is a process of epithelial rearrangement resulted from cell division-mediated intercalations, which is necessary for the cellular spatial-patterning movements (Firmino et al., 2016). It is a morphogenetic process to form a three-layer organism consisting of the endoderm layer inside, the ectoderm outside, and the mesoderm in the middle, represented by internalization of the mesendoderm, convergence to the midline, and extension along the anteroposterior axis, all of which is conserved throughout evolution in various species (Thiery et al., 2009). These dramatic shape-changes require locally produced and anisotropically applied forces. Depletion of myosin regulatory light chain in the embryo was able to block force generation at gastrulation by destabilizing the myosin II (MII) hexameric complex and inhibiting MII contractility (Pfister et al., 2016). Interestingly, most subapical clusters in early mesoderm move apically and enhance in density and intensity. This phenomenon depended on MII and was correlated with the pulse actomyosin accumulation before the cells gained morphology change, indicating that contractile myosin-driven cell movement is prior to transcript-driven EMT during early gastrulation (Weng and Wieschaus, 2016).

The establishment of the embryonic apical-basal polarity is contributed to well-defined intercellular adhesive structures. This complex process is coordinated by disruption of epithelial cell-cell junction, breakdown of cell-basement membrane interaction, and changes in cytoskeletal architecture. Decomposition of basement membrane is the first recognized step in EMT process during gastrulation, which is mediated by certain molecular families. Inhibition of Rho pathway caused disruption of cell-basement interaction and microtubule instability (Nakaya et al., 2008; Stankova et al., 2015). Epithelialization and differentiation of the apical membrane during blastoderm stages is regulated by some transmembrane signals, one of which is Crumbs homolog 2 (Crb2). Crb2-mutant murine embryo showed a disturbed epiblast polarity and impaired EMT process at the primitive streak, resulting in impaired blastoderm formation and embryonic lethality (Xiao et al., 2011).

Gastrulation lasts a short duration and thus requires rapid changes in gene expression and function. Downregulation of E-cadherin, for example, is controlled both at the transcriptional level by Snai1, Zeb, Twist, and by P38 interacting protein (IP)-p38-MAP kinase complex (Lamouille et al., 2014). Reciprocal transcriptional repression of Snail2 and Sox3 factors mediated the mesendoderm internalization in chick embryos. Snail-expressing cells ingressed into the primitive streak, whereas Sox3-positive cells formed ectodermal derivatives outside, indicating that the relationship between Snail and Sox determines the embryonic cellular localization(Acloque et al., 2011). In addition, FGFR was known to be required for cell migration during the gastrulation by regulating several pathways, including RhoA, the non-canonical Wnt pathway, PDGF signaling, and the cell adhesion protein N-cadherin. In FGFR1-knockout mice, epiblast cells in the primitive streak were less mobile on account of sustained E-cadherin expression (Hardy et al., 2011). O-fucosyltransferase-2 (POFUT2) specifically adds O-fucose to Thrombospondin type 1 repeat (TSR) superfamily regulating diverse biological activities ranging from cell motility to inhibition of angiogenesis. Pofut2-mutant murine embryos underwent extensive mesoderm differentiation when the embryo established anterior/posterior polarity, demonstrating that O-fucosylation of TSRs is essential for restricting pattern of EMT during gastrulation (Du et al., 2010).

2.3. EMT from primary to tertiary in embryo development

During gastrulation, the majority cells undergoing EMT contribute to ingressive mesoderm which is crucial to be separated from adjacent non-ingressing neuroectoderm cells to generate anterior/posterior regionalization. During cellular transition between epithelial and mesenchymal phenotype, a hybrid epithelial-mesenchymal state might exist to enable the cells to move collectively, to identify an external signaling, and to acquire maximum cellular transition (Jolly et al., 2016). A fully-transformed mesenchymal fate is more associated with mesoderm development, while in diploblastic and some triploblastic animals the EMT process may be only partially involved in endoderm formation (Nakaya and Sheng, 2008). The neural crest delamination in ectoderm is also a consequence of EMT which is coordinated by Snails, FoxD3 and SoxE factors expression, even though the precise combination varies temporally and spatially (Theveneau and Mayor, 2012).

Snail expression in presomitic mesoderm is asymmetric at left-right pattern and induces desynchronic somitogenesis. The endogenous retinoic acid-induced asymmetrically regression of Snail prevents the cellular invasiveness and ensures bilateral synchronic mesoderm segmentation (Morales et al., 2007). The migratory cells during early-gastrulation thus acquire a condense population mediated by a transient mesenchymal-epithelial transition (MET) process to obtain an epithelial homogeneity, which is initiated and stabilized by integrating signals to control the reorganization of the ECM, cytoskeleton, and adhesion junctions (Rowton et al., 2013). These secondary epithelia undergo a secondary EMT with more restricted differentiated potential to generate different types of cells, including the notochord, somites, heart, gut, kidney, body wall and lining of the coelom (Ohta et al., 2010; Thiery et al., 2009).

The somite epithelialization shares a common regulation way with intersomitic border formation which progresses in a rostral to caudal direction along the axis. The stimulators initiating secondary EMT remain largely unknown, one of which is postulated to be the signals emanated from the neural tube and notochord. Another proposed mechanism is that in a passive way, ventral somitic EMT is merely due to lack of epithelial-maintaining signals when the somatic cells escape from the pro-epithelializing activity (Kalcheim, 2015). Interestingly, caudal FGF signaling was demonstrated to control the trunk neural crest cell specification and emigration, whereas rostrally generated retinoic acid (RA) activity gradient is only required to control early neural crest cell specification. This opposing gradient of FGF and RA pathway orchestrates progressive neural and limb development (Martinez-Morales et al., 2011). BMP and Wnt signaling pathways were reported to be required for proper pattern of the neural tube and somites, providing evidence that both genes are crucial for regulating mammalian anterior-posterior (A–P) axial elongation during the EMT process (Andre et al., 2015; Martinez-Morales et al., 2011). The Wnt-regulated basic helix-loop-helix (bHLH) transcription factor mesogenin 1 (Msgn1) is a major regulator controlling paraxial pre-somitic mesoderm (PSM) differentiation, by directly activating the transcriptional programs that define PSM identity, the EMT, cellular motility, and segmentation (Chalamalasetty et al., 2014). BMP family was also reported to be close related to Snails expression in the primitive streak along its A–P axis. SMADs, the downstream factors of BMP signaling pathway, also expresses on the pre-migratory cells adjacent to the primitive streak and lateral neural crest, demonstrating that BMP-SMAD signaling induces EMT via MMP-mediated basal membrane disruption in early tail bud stage, whereas Noggin inhibits BMP signaling and results in the suppression of EMT and basement membrane formation in late gastrulation (Ohta et al., 2010).

Cellular migration and differentiation of epicardium-derived progenitor cells (EPDC) is one of the major cardio-vasculogenesis events. Epicardium is a single cell layer of mesothelium lining the outer heart. In tamoxifen-induced Wt1-knockout immortalized epicardial cells (Cre+ CoMEECs), loss of Wt1 led to a robust increase in E-cadherin expression in a dose-dependent manner, indicating that Wt1 is important in regulating epicardial EMT-related development. Treatment of the Cre+ CoMEECs with tamoxifen led not only to the changes in the EMT-expressing pattern, but also to reduced cell migration. The mesodermal lineage deficiency was rescued by increased expression of Snai1, underscoring the importance of the EMT in generating differentiated cells (Martinez-Estrada et al., 2010). The key EMT-stimulator transforming growth factor (TGF)-β was reported to lead nuclear accumulation of myocardin-related transcription factors (MRTFs) in the epicardium. Mice lacking MRTFs showed a series of cardio-vasculogenic disorganization, including coronary plexus malformation, endothelial cell dysfunction and sub-epicardial hemorrhage (Trembley et al., 2015).

The EMT process was also investigated during other embryonic mesodermal lineage development. Stroma components have been demonstrated to provide important inter-epithelial signals for the formation of overlying coelomic epithelial cell sheet during nephric duct development. The mesonephros is a transient structure in which a single epithelial cell layer nephric coelomic epithelium (Neph-CE) covers multiple types of cells. Ablation of inter-epithelial crosstalk caused increased sensitivity to the EMT-induced external stress (Yoshino et al., 2014). Moreover, Wnt signaling is also related to the EMT and is critical for proper formation of the female reproductive tract. Less cellular proliferation and delayed uterine gland formation were seen in embryos with stabilization of β-catenin, accompanied with the induction of an EMT event from pre-birth to 5 days of post-birth (Stewart et al., 2013).

An in vitro analysis showed that embryonic hematopoiesis regionally distributed at different embryo stages. The hematopoietic bud was dispersive in the entire epiblast during early- and mid-gastrulation but was restricted in the posterior at the headfold stage, whereas the EMT-inducers Activin A and BMP-4 enhanced hematogenic potency of anterior part (Kanatsu and Nishikawa, 1996). One of the EMT transcripts Zeb2/Sip1 was demonstrated to be essential for murine embryonic hematopoietic stem and progenitor cell differentiation and mobilization (Goossens et al., 2011). The population fitting the description of cells in the EMT process was observed in the early embryonic hematopoietic liver microenvironment but not in nonhematopoietic liver at the late gestation and the post-natal stage. Furthermore, the hematopoietic supportive capacity of the EMT cells disappeared after hepatocytic maturation, indicating that the EMT process does contribute to hematopoiesis in embryonic stage (Chagraoui et al., 2003).

3. Epithelial or mesenchymal specificity of pluripotent stem cells

Stem cells are a class of undifferentiated or poorly differentiated cells, which are able to self-renew and to produce well-differentiated daughter cells. A variety of markers, such as Oct4, Nanog, alkaline phosphatase (ALP), stage specific embryonic antigen (SSEA), CD133, and aldehyde dehydrogenase (ALDH), have been indicated to relate to cellular pluripotency (Hess et al., 2006; Li et al., 2001; Thomson et al., 1998; Tondreau et al., 2005). Stem cells can be further classified as totipotent, pluripotent, and multi/uni-potent by their differentiation potential. Among them, embryonic stem cells (ESCs), which are generated from ICM in embryo blastocyst, are a totipotent in vitro model able to mimic embryo development (Henderson et al., 2002; Thomson et al., 1998). Reprogramming of somatic cells to induced pluripotent stem cells (iPSCs) provides an unprecedented opportunity to generate a new patient-specific cell source (Guha et al., 2013; Inoue et al., 2014; Maekawa et al., 2011; Takahashi et al., 2007; Takahashi and Yamanaka, 2006). ESCs and iPSCs, joint named pluripotent stem cells (PSCs), share similar gene expression profiles and development potential and thus provide a novel cellular tool for regenerative medicine, disease modeling, and preclinical drug screening (Halevy and Urbach, 2014; Liang and Zhang, 2013). As for the widely recognized differentiating potential of stem cells, determination of the epithelial and mesenchymal specificity during stem cell differentiation and reprogramming will provide more evidences to the relationship between E-M characteristics and pluripotency.

3.1. EMT and PSC differentiation

ESCs are derived from inner cell mass (ICM) in the blastocyst stage, providing the possibility that undifferentiated ESC cells dictate the feature of ICMs to form an accessible and tractable differentiating model to investigate the lineage selection at gastrulation (Slawny and O’Shea, 2013). An in vitro EMT process has been investigated during ESCs and iPSCs differentiation into three germ layers, which are regulated by special transcripts and modulators.

In murine ESCs, E-cadherin positively express on the intercellular membrane accompanied by pluripotent markers (Nanog, Oct4, Sox2 and alkaline phosphatase). Depletion of Snail did not impact the expression of pluripotent markers, Phospho-histone H3 and TUNEL experiment, indicating that Snail is dispensable for pluripotency maintenance, cell cycle and cell survival regulation. Undifferentiated ESCs retained the epithelial phenotype while the differentiation procedure was showed as an endogenous burst of Wnt signaling-mediated Snail expression, which regulates neuroectodermal and the consequent lineage fate defining mesoderm commitment (Lin et al., 2014). Another EMT-transcript, Zeb2, was indicated to modulate the cell-fate decision between neuroectoderm and mesoderm in the duration from undifferentiated ESCs to primary germ layer differentiation (Chng et al., 2010). Moreover, miR-200a was demonstrated to exert an opposite function to Snail to stall ESC differentiation. In consideration of repressive actions of miR-200 family on the EMT by targeting the transcription factors Zeb1 and Zeb2, this sequential orchestration of EMT-related transcripts and miRNAs plays an important role in regulating ESC-derived early lineage selection (Gill et al., 2011).

3.2. MET in somatic reprogramming

Reprogramming refers to re-establishment of epigenetic marks and induction of pluripotent stem cells by transfer of nuclear contents into oocytes (Noggle et al., 2011; Yamada et al., 2014), by fusion with ES cells (Ambrosi et al., 2007; Lluis et al., 2008), and by transfection of defined factors into adult mature cells (Takahashi et al., 2007; Takahashi and Yamanaka, 2006). Among them, somatic reprogrammed iPSCs, which share similar plasticity to ESCs (Bilic and Izpisua Belmonte, 2012) and thus undergo the same trend of EMT during differentiation (Li and Niyibizi, 2012), have captured more and more attention because of their potential to differentiate into patient-specific cells. Whether somatic reprogramming fits the opposite process of EMT is an attractive question, and the clarification of this question will shed light on controlling the efficiency of iPSC reprogramming.

The reprogramming of somatic cells into iPSCs requires defined factors, such as Sox2, Oct4, c-myc and Klf4 (called as Yamanaka factors). It was demonstrated that Sox2, Oct4, and c-myc suppress TGF-β signaling while Klf4 activates multiple epithelial genes. During reprogramming, the cells aggregated into a distinguished epithelial-like colony with well-defined intercellular junctions. A pan-cytokeratin containing E-cadherin was upregulated after introducing Yamanaka factors. Expression of mesenchymal marker Vimentin was repressed concomitantly to the emergence of iPSC colony (Hoffding and Hyttel, 2015). Meanwhile, a reduction of the mesenchymal-like marker fibronectin (Fn) and Snail was observed, demonstrating that reprograming to iPSCs experiences the reverse process of EMT, the MET (Li et al., 2010). The morphology change of fibroblasts to iPSCs resembled an artificial rewinding of mesenchyme to epithelial-like epiblast. A more detailed lineage dissection found that a short period of TGF-β treatment enhanced reprogramming process, whereas a 12-day of full-term TGF-β treatment blocked reprogramming, indicating that optimal de-differentiation requires a time-sensitive trend that the efficiency of reprogramming is increased by a sequential early-EMT and delayed-MET in order (Liu et al., 2013).

4. Distinct story of relationship between EMT/MET and pluripotency in cancers

Compared to PSCs, adult stem cells exist in post-natal tissues or organs that have restricted differentiation potential. These adult stem cells are capable of self-renewal and producing more mature functional offspring of limited number of cell types (Visvader and Clevers, 2016; Wang et al., 2016). The cells residing in tumors having limited potential of differentiation and self-renewal are termed as tumor-initiating cells (TICs) or cancer stem cells (CSCs) (Ponti et al., 2005; Singh et al., 2004; Wang et al., 2006). This small population is documented to be selected by cellular surface antigens (CD133, CD34, CD44, ABCB5) (Jin et al., 2006; Ricci-Vitiani et al., 2007; Schatton et al., 2008; Trempus et al., 2007), pluripotent markers (SSEA-1, Oct-4) (Levings et al., 2009; Son et al., 2009), Hoechst 33342 dye-efflux capability (Harris et al., 2008; Ho et al., 2007) and Aldehyde Dehydrogenase (ALDH) (Carpentino et al., 2009; Visus et al., 2011), and was indicated to be responsible for tumorigenesis (O’Brien et al., 2007). The transition between epithelial and mesenchymal counterparts exists in various tumors and contributes to tumor metastasis and chemoresistance (Fischer et al., 2015; Zheng et al., 2015). However, whether the direction of EMT or MET relates to the emergence of CSCs remains controversial in different CSC-purifying methods and in different tissues (Brabletz, 2012).

4.1. The EMT is related to mesenchymal-like CSC initiation

In 2008, Mani et al. reported the association between the EMT and the gain of epithelial stem cell property (Mani et al., 2008). In their study, the neoplastic mammary stem cells were determined by CD44high/CD24low phenotype and the capability to form a mammosphere. Ectopic expression of Snail or Twist in human mammary epithelial cells (HMLEs) or exposing HMLEs to TGF-β stimulation resulted in the acquisition of CD44high/CD24low population in the appearance of mesenchymal growth pattern, with the ability to form mammosphere and to generate the CD44low/CD24high non-mammosphere-forming cells in vitro, indicating that the cells that undergo an EMT are associated with CSC acquisition (Mani et al., 2008). A distinct localization of normal gland-reconstituting mammary stem cells and breast CSCs were investigated by the same group afterwards, and found that the former is in basal layer and the latter is originated in the luminal layer, where Snail, but not Slug serves as the key regulator in CSC-initiation (Ye et al., 2015). The tumorigenicity can be also determined by an in vitro sphere-forming assay. The feedback loop of TF-Zeb with suppression of stemness-inhibitor miRNAs was investigated to be linked with increased sphere-forming capability in pancreatic CD44+/CD24+ cancer stem cells (Wellner et al., 2009). The downstream effector of tumor suppressor and the cyclin-dependent kinase inhibitor p21CIP1/WAF1 was showed to be associated with the mammary neoplastic EMT. Expression of Sox2, Klf4, and Oct-4 was increased in the Ras-p21CIP1−/− and c-Myc-p21CIP1−/− tumors, elucidating that loss of p21CIP1/WAF1 can induce a Ha-Ras- and c-Myc-dependent EMT and an enrichment of CD44high/CD24low phenotype with an embryonic stem-like gene expression pattern in immortal human mammary MCF10A-c-Myc cells (Liu et al., 2009).

The CSC markers are sub-assorted to the cellular function (Hoechst 33342 dye-effluxing assay), nuclear transcription factor (Oct-4), and cell surface antigens (CD44, CD133, CD34, SSEA, ABCB5, ALDH). Since there is no unified CSC-specific marker in tissues and organs, the CSCs purified by different methods might exhibit distinct characters. Among them, CD44 is also used as a mesenchymal marker to specify mesenchymal stroma cells. The cells derived from the EMT process behave similarly to mesenchymal stem cells (MSCs) with the potential to differentiate into multiple lineages, share many properties that are typical for MSCs, including CD44(+), CD24(−), CD45(−), EMT-TFs, mesenchyme forkhead 1 (FOXC2), and CD140b (Battula et al., 2010). This CD44+/CD24− mesenchymal phenotype with more adherent protein expression and more mobile ability may enhance the cells to form aggregates in vitro, one of which meets the requirement of CSC’s features in culture, and enable the cells to exhibit more metastatic capability of migration, invasion and chemoresistance (Kawamura et al., 1991). Knockdown of CD44 induced MET and subsequently resulted in impaired migration and invasion of cancer cells both in vitro and in vivo (Gao et al., 2015).

4.2. The EMT is related to epithelial CSC differentiation

Other studies of the relationship between the lineage transition and the tumor initiating capability indicated that induction of EMT in breast cancer cells is not associated with enhancing tumor-initiating capacity, but instead, conferred a CD44+/CD24− phenotype and the malignant properties, including cell proliferation, migration, and resistance to chemotherapy and radiation. On the other hand, MET does not lead to inhibition or loss of the tumor-initiating capacity, but markedly attenuated other malignant properties (Xie et al., 2014). Repression of the EMT inducer Prrx1 reverted the cells to the epithelial phenotype concomitant with the acquisition of the stem cell properties (Ocana et al., 2012).

Hoechst 33342 is a vital dye used the first time in 1996 to stain a small population in bone marrow which expresses multipotential hematopoietic stem cells and ATP-binding cassette transporter G (ABCG) family able to efflux Hoechst 33342 fluorescence dye (Goodell et al., 1996). This population was named as side population (SP) and had been applied as one of the representative methods to purify various tissue pluripotent cells and in cancers (Boesch et al., 2014; Goodell et al., 1997; Hirschmann-Jax et al., 2004; Jackson et al., 1999; Jonker et al., 2005). Unlike mesenchymal specificity of CD44, Hoechst 33342-effluxing SP cells may exhibit the characters associating with their lineage origins. We found that SP cells in ovarian cancers exhibited an epithelial phenotype and showed a distinct pluripotent gene profile with reduced expression of cell adhesion molecules. Cultivation of SP cells into non-SP cells resulted in decreased expression of Oct-4 and Nanog with more mesenchymal phenotype and increased invasive potential (Jiang et al., 2012). SP cells were able to be depleted by treatment of TGF-β-directed EMT which was associated with down-regulation of ABCG2 expression, while removal of TGF-β restored SP abundance with an epithelial phenotype (Yin et al., 2008). However, heterogeneity still exists in side population, and their relevance to EMT need to be further investigated. A stem-purifying method that is not associated with epithelial or mesenchymal in nature may facilitate to distinguish this difference.

5. Summary

The transition between epithelial and mesenchymal lineages is a common process in embryonic development and cancer progression, both of which encounter the developmental processes including cellular invasion into the basal membrane, migration to the distant site, seeding in the proper circumstance and growing to form aggregates, tissues, organs and metastatic sites.

Embryogenesis is a complicated process whose details are far from clarification. The embryogenic EMT enables cells to move to the accurately positioned environment for further development. The sequential transformations involved in embryogenesis are served as development-oriented events which are able to generate further differentiated cells. The ex vivo cellular model which mimics pluripotent embryo in nature, ESCs or iPSCs, are demonstrated to mainly experience EMT in their differentiation and MET in their de-differentiation. Even though a short period of EMT was seen in early reprogramming of iPSCs, it is much like the short period of MET in embryogenesis to enable the cells to acquire a unified status for next step differentiation, to generate tight junction between the reprogramming cells, and surrounding feeders to facilitate the de-differentiation (Figure 1).

The mature body is grown from a fertilized egg, the primary homozygous stem cell, in a vital life, while the cancers are recently documented to be initiated from a small population of cells having stem cell-like properties. The normal stem cells and CSCs share some properties but, in some ways, behave differently. The accurate description of EMT and MET in CSCs is remained controversial, and much is caused by the heterogeneity in cancers and the inconsistency of CSC feature in different tissues (Table 1). The CSCs isolated by epithelial or mesenchymal markers will be bound to show their corresponding lineage characteristics, resulting in the uncertainty in the studies of relationship between EMT/MET and differentiation/de-differentiation. Finding a unified CSC-isolating method independent of lineage markers will help to accurately clarify this question in the future.

Table 1.

Distinct relationship between E/M lineage and CSC features

Lineage relationship Origin Models Inducer Stem cell features Biological function of E-M transition Reference
CSC marker Other feature
Mesenchymal lineage relatedCSC initiation Breast HMLEs TGF-β stimulationEctopic expression of Snail or Twist CD44+/CD24− Mammosphere-forming cells CSC initiation (Mani et al., 2008)
Pancreas pancreatic cancer cells Zeb1-Mir200 loop CD44+/CD24+ Sphere-forming capacity Self-renewal, drug resistance (Wellner et al., 2009)
Breast MCF10A loss of p21CIP1/WAF1 CD44+/CD24− Expression of Sox2, Klf4 and Oct-4 Enrichment of CD44+/CD24− phenotype (Liu et al., 2009)
Breast HMECs Ectopic expressing Twist, Snail, or TGF-β1 stimulation CD44+/CD24− CD45(−), EMT-TFs, FOXC2(+), and CD140b(+) Derive cells similar to MSCs (Battula et al., 2010)
Breast Slug+ or Snail+ knock-in mice Slug+ or Snail+ knock-in CD44+/CD24− Epcam(−) A distinct localization of normal mammary stem cells and breast CSCs (Ye et al., 2015)

Epithelial lineage related CSC initiation Breast and other Chicken embryo, zebrafish, human cancer cells, MDCK cells Prrx1 knock-in or knock out CD44+/CD24− MET concomitant with the acquisition of the stem cell properties (Ocana et al., 2012)
Ovarian Ovarian cancer cells TGF-β stimulation SP cells Expression of Oct-4, Nanog, sphere-forming capacity Enhanced differentiation of SP cells into non-SP cells (Jiang et al., 2012)
Breast MCF7 cells TGF-β stimulation SP cells In vivo tumorigenicity Depletion of SP cells by transcriptional repression of the ABCG2 gene (Yin et al., 2008)

CSCs: cancer stem cells, HMLEs or HMECs: human mammary epithelial cells, SP: side population, MSCs: mesenchymal stem cells, MDCK cells: Madin-Darby Canine Kidney

Acknowledgments

This study was partially supported by National Natural Science Foundation of China (81561138002, 31371480, 91542109) and Program of Shanghai Subject Chief Scientist (16XD1400600) to T.C, and NIH/NIDDK (R01 DK106109) to Z.Z.W.

Footnotes

Disclosures

The authors indicate no potential conflict of interest.

References

  1. Acloque H, Adams MS, Fishwick K, Bronner-Fraser M, Nieto MA. Epithelial-mesenchymal transitions: the importance of changing cell state in development and disease. The Journal of clinical investigation. 2009;119(6):1438–1449. doi: 10.1172/JCI38019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Acloque H, Ocana OH, Matheu A, Rizzoti K, Wise C, Lovell-Badge R, Nieto MA. Reciprocal repression between Sox3 and snail transcription factors defines embryonic territories at gastrulation. Developmental cell. 2011;21(3):546–558. doi: 10.1016/j.devcel.2011.07.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Ambrosi DJ, Tanasijevic B, Kaur A, Obergfell C, O’Neill RJ, Krueger W, Rasmussen TP. Genome-wide reprogramming in hybrids of somatic cells and embryonic stem cells. Stem cells. 2007;25(5):1104–1113. doi: 10.1634/stemcells.2006-0532. [DOI] [PubMed] [Google Scholar]
  4. An H, Stoops SL, Deane NG, Zhu J, Zi J, Weaver C, Waterson AG, Zijlstra A, Lindsley CW, Beauchamp RD. Small molecule/ML327 mediated transcriptional de-repression of E-cadherin and inhibition of epithelial-to-mesenchymal transition. Oncotarget. 2015;6(26):22934–22948. doi: 10.18632/oncotarget.4473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Andre P, Song H, Kim W, Kispert A, Yang Y. Wnt5a and Wnt11 regulate mammalian anterior-posterior axis elongation. Development. 2015;142(8):1516–1527. doi: 10.1242/dev.119065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Banerjee P, Venkatachalam S, Mamidi MK, Bhonde R, Shankar K, Pal R. Vitiligo patient-derived keratinocytes exhibit characteristics of normal wound healing via epithelial to mesenchymal transition. Experimental dermatology. 2015;24(5):391–393. doi: 10.1111/exd.12671. [DOI] [PubMed] [Google Scholar]
  7. Battula VL, Evans KW, Hollier BG, Shi Y, Marini FC, Ayyanan A, Wang RY, Brisken C, Guerra R, Andreeff M, Mani SA. Epithelial-mesenchymal transition-derived cells exhibit multilineage differentiation potential similar to mesenchymal stem cells. Stem cells. 2010;28(8):1435–1445. doi: 10.1002/stem.467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Bell CE, Watson AJ. SNAI1 and SNAI2 are asymmetrically expressed at the 2-cell stage and become segregated to the TE in the mouse blastocyst. PloS one. 2009;4(12):e8530. doi: 10.1371/journal.pone.0008530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Bessonnard S, Mesnard D, Constam DB. PC7 and the related proteases Furin and Pace4 regulate E-cadherin function during blastocyst formation. The Journal of cell biology. 2015;210(7):1185–1197. doi: 10.1083/jcb.201503042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Bilic J, Izpisua Belmonte JC. Concise review: Induced pluripotent stem cells versus embryonic stem cells: close enough or yet too far apart? Stem cells. 2012;30(1):33–41. doi: 10.1002/stem.700. [DOI] [PubMed] [Google Scholar]
  11. Boesch M, Zeimet AG, Reimer D, Schmidt S, Gastl G, Parson W, Spoeck F, Hatina J, Wolf D, Sopper S. The side population of ovarian cancer cells defines a heterogeneous compartment exhibiting stem cell characteristics. Oncotarget. 2014;5(16):7027–7039. doi: 10.18632/oncotarget.2053. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Bowen JA, Burghardt RC. Cellular mechanisms of implantation in domestic farm animals. Seminars in cell & developmental biology. 2000;11(2):93–104. doi: 10.1006/scdb.2000.0155. [DOI] [PubMed] [Google Scholar]
  13. Brabletz T. EMT and MET in metastasis: where are the cancer stem cells? Cancer cell. 2012;22(6):699–701. doi: 10.1016/j.ccr.2012.11.009. [DOI] [PubMed] [Google Scholar]
  14. Buczek ME, Miles AK, Green W, Johnson C, Boocock DJ, Pockley AG, Rees RC, Hulman G, van Schalkwyk G, Parkinson R, Hulman J, Powe DG, Regad T. Cytoplasmic PML promotes TGF-beta-associated epithelial-mesenchymal transition and invasion in prostate cancer. Oncogene. 2015 doi: 10.1038/onc.2015.409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Carpentino JE, Hynes MJ, Appelman HD, Zheng T, Steindler DA, Scott EW, Huang EH. Aldehyde dehydrogenase-expressing colon stem cells contribute to tumorigenesis in the transition from colitis to cancer. Cancer research. 2009;69(20):8208–8215. doi: 10.1158/0008-5472.CAN-09-1132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Chagraoui J, Lepage-Noll A, Anjo A, Uzan G, Charbord P. Fetal liver stroma consists of cells in epithelial-to-mesenchymal transition. Blood. 2003;101(8):2973–2982. doi: 10.1182/blood-2002-05-1341. [DOI] [PubMed] [Google Scholar]
  17. Chalamalasetty RB, Garriock RJ, Dunty WC, Jr, Kennedy MW, Jailwala P, Si H, Yamaguchi TP. Mesogenin 1 is a master regulator of paraxial presomitic mesoderm differentiation. Development. 2014;141(22):4285–4297. doi: 10.1242/dev.110908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Chng Z, Teo A, Pedersen RA, Vallier L. SIP1 mediates cell-fate decisions between neuroectoderm and mesendoderm in human pluripotent stem cells. Cell stem cell. 2010;6(1):59–70. doi: 10.1016/j.stem.2009.11.015. [DOI] [PubMed] [Google Scholar]
  19. Choi HJ, Park JH, Park M, Won HY, Joo HS, Lee CH, Lee JY, Kong G. UTX inhibits EMT-induced breast CSC properties by epigenetic repression of EMT genes in cooperation with LSD1 and HDAC1. EMBO reports. 2015a;16(10):1288–1298. doi: 10.15252/embr.201540244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Choi SH, Hong ZY, Nam JK, Lee HJ, Jang J, Yoo RJ, Lee YJ, Lee CY, Kim KH, Park S, Ji YH, Lee YS, Cho J, Lee YJ. A Hypoxia-Induced Vascular Endothelial-to-Mesenchymal Transition in Development of Radiation-Induced Pulmonary Fibrosis. Clinical cancer research : an official journal of the American Association for Cancer Research. 2015b;21(16):3716–3726. doi: 10.1158/1078-0432.CCR-14-3193. [DOI] [PubMed] [Google Scholar]
  21. Cicchini C, de Nonno V, Battistelli C, Cozzolino AM, De Santis Puzzonia M, Ciafre SA, Brocker C, Gonzalez FJ, Amicone L, Tripodi M. Epigenetic control of EMT/MET dynamics: HNF4alpha impacts DNMT3s through miRs-29. Biochimica et biophysica acta. 2015;1849(8):919–929. doi: 10.1016/j.bbagrm.2015.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Correa-Costa M, Andrade-Oliveira V, Braga TT, Castoldi A, Aguiar CF, Origassa CS, Rodas AC, Hiyane MI, Malheiros DM, Rios FJ, Jancar S, Camara NO. Activation of platelet-activating factor receptor exacerbates renal inflammation and promotes fibrosis. Laboratory investigation; a journal of technical methods and pathology. 2014;94(4):455–466. doi: 10.1038/labinvest.2013.155. [DOI] [PubMed] [Google Scholar]
  23. Dang TT, Esparza MA, Maine EA, Westcott JM, Pearson GW. DeltaNp63alpha Promotes Breast Cancer Cell Motility through the Selective Activation of Components of the Epithelial-to-Mesenchymal Transition Program. Cancer research. 2015;75(18):3925–3935. doi: 10.1158/0008-5472.CAN-14-3363. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Du J, Takeuchi H, Leonhard-Melief C, Shroyer KR, Dlugosz M, Haltiwanger RS, Holdener BC. O-fucosylation of thrombospondin type 1 repeats restricts epithelial to mesenchymal transition (EMT) and maintains epiblast pluripotency during mouse gastrulation. Developmental biology. 2010;346(1):25–38. doi: 10.1016/j.ydbio.2010.07.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Duzyj CM, Buhimschi IA, Motawea H, Laky CA, Cozzini G, Zhao G, Funai EF, Buhimschi CS. The invasive phenotype of placenta accreta extravillous trophoblasts associates with loss of E-cadherin. Placenta. 2015;36(6):645–651. doi: 10.1016/j.placenta.2015.04.001. [DOI] [PubMed] [Google Scholar]
  26. Firmino J, Rocancourt D, Saadaoui M, Moreau C, Gros J. Cell Division Drives Epithelial Cell Rearrangements during Gastrulation in Chick. Developmental cell. 2016;36(3):249–261. doi: 10.1016/j.devcel.2016.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Fischer KR, Durrans A, Lee S, Sheng J, Li F, Wong ST, Choi H, El Rayes T, Ryu S, Troeger J, Schwabe RF, Vahdat LT, Altorki NK, Mittal V, Gao D. Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance. Nature. 2015;527(7579):472–476. doi: 10.1038/nature15748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Galvan JA, Zlobec I, Wartenberg M, Lugli A, Gloor B, Perren A, Karamitopoulou E. Expression of E-cadherin repressors SNAIL, ZEB1 and ZEB2 by tumour and stromal cells influences tumour-budding phenotype and suggests heterogeneity of stromal cells in pancreatic cancer. British journal of cancer. 2015;112(12):1944–1950. doi: 10.1038/bjc.2015.177. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  29. Gao Y, Ruan B, Liu W, Wang J, Yang X, Zhang Z, Li X, Duan J, Zhang F, Ding R, Tao K, Dou K. Knockdown of CD44 inhibits the invasion and metastasis of hepatocellular carcinoma both in vitro and in vivo by reversing epithelial-mesenchymal transition. Oncotarget. 2015;6(10):7828–7837. doi: 10.18632/oncotarget.3488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Gill JG, Langer EM, Lindsley RC, Cai M, Murphy TL, Kyba M, Murphy KM. Snail and the microRNA-200 family act in opposition to regulate epithelial-to-mesenchymal transition and germ layer fate restriction in differentiating ESCs. Stem cells. 2011;29(5):764–776. doi: 10.1002/stem.628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Goodell MA, Brose K, Paradis G, Conner AS, Mulligan RC. Isolation and functional properties of murine hematopoietic stem cells that are replicating in vivo. The Journal of experimental medicine. 1996;183(4):1797–1806. doi: 10.1084/jem.183.4.1797. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Goodell MA, Rosenzweig M, Kim H, Marks DF, DeMaria M, Paradis G, Grupp SA, Sieff CA, Mulligan RC, Johnson RP. Dye efflux studies suggest that hematopoietic stem cells expressing low or undetectable levels of CD34 antigen exist in multiple species. Nature medicine. 1997;3(12):1337–1345. doi: 10.1038/nm1297-1337. [DOI] [PubMed] [Google Scholar]
  33. Goossens S, Janzen V, Bartunkova S, Yokomizo T, Drogat B, Crisan M, Haigh K, Seuntjens E, Umans L, Riedt T, Bogaert P, Haenebalcke L, Berx G, Dzierzak E, Huylebroeck D, Haigh JJ. The EMT regulator Zeb2/Sip1 is essential for murine embryonic hematopoietic stem/progenitor cell differentiation and mobilization. Blood. 2011;117(21):5620–5630. doi: 10.1182/blood-2010-08-300236. [DOI] [PubMed] [Google Scholar]
  34. Grande MT, Sanchez-Laorden B, Lopez-Blau C, De Frutos CA, Boutet A, Arevalo M, Rowe RG, Weiss SJ, Lopez-Novoa JM, Nieto MA. Snail1-induced partial epithelial-to-mesenchymal transition drives renal fibrosis in mice and can be targeted to reverse established disease. Nature medicine. 2015;21(9):989–997. doi: 10.1038/nm.3901. [DOI] [PubMed] [Google Scholar]
  35. Greenburg G, Hay ED. Epithelia suspended in collagen gels can lose polarity and express characteristics of migrating mesenchymal cells. The Journal of cell biology. 1982;95(1):333–339. doi: 10.1083/jcb.95.1.333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Gros J, Tabin CJ. Vertebrate limb bud formation is initiated by localized epithelial-to-mesenchymal transition. Science. 2014;343(6176):1253–1256. doi: 10.1126/science.1248228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Guha P, Morgan JW, Mostoslavsky G, Rodrigues NP, Boyd AS. Lack of immune response to differentiated cells derived from syngeneic induced pluripotent stem cells. Cell stem cell. 2013;12(4):407–412. doi: 10.1016/j.stem.2013.01.006. [DOI] [PubMed] [Google Scholar]
  38. Halevy T, Urbach A. Comparing ESC and iPSC-Based Models for Human Genetic Disorders. Journal of clinical medicine. 2014;3(4):1146–1162. doi: 10.3390/jcm3041146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Hardy KM, Yatskievych TA, Konieczka J, Bobbs AS, Antin PB. FGF signalling through RAS/MAPK and PI3K pathways regulates cell movement and gene expression in the chicken primitive streak without affecting E-cadherin expression. BMC developmental biology. 2011;11:20. doi: 10.1186/1471-213X-11-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Harris MA, Yang H, Low BE, Mukherjee J, Guha A, Bronson RT, Shultz LD, Israel MA, Yun K. Cancer stem cells are enriched in the side population cells in a mouse model of glioma. Cancer research. 2008;68(24):10051–10059. doi: 10.1158/0008-5472.CAN-08-0786. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Henderson JK, Draper JS, Baillie HS, Fishel S, Thomson JA, Moore H, Andrews PW. Preimplantation human embryos and embryonic stem cells show comparable expression of stage-specific embryonic antigens. Stem cells. 2002;20(4):329–337. doi: 10.1634/stemcells.20-4-329. [DOI] [PubMed] [Google Scholar]
  42. Hess DA, Wirthlin L, Craft TP, Herrbrich PE, Hohm SA, Lahey R, Eades WC, Creer MH, Nolta JA. Selection based on CD133 and high aldehyde dehydrogenase activity isolates long-term reconstituting human hematopoietic stem cells. Blood. 2006;107(5):2162–2169. doi: 10.1182/blood-2005-06-2284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Hirschmann-Jax C, Foster AE, Wulf GG, Nuchtern JG, Jax TW, Gobel U, Goodell MA, Brenner MK. A distinct “side population” of cells with high drug efflux capacity in human tumor cells. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(39):14228–14233. doi: 10.1073/pnas.0400067101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Ho MM, Ng AV, Lam S, Hung JY. Side population in human lung cancer cell lines and tumors is enriched with stem-like cancer cells. Cancer research. 2007;67(10):4827–4833. doi: 10.1158/0008-5472.CAN-06-3557. [DOI] [PubMed] [Google Scholar]
  45. Hoffding MK, Hyttel P. Ultrastructural visualization of the Mesenchymal-to-Epithelial Transition during reprogramming of human fibroblasts to induced pluripotent stem cells. Stem cell research. 2015;14(1):39–53. doi: 10.1016/j.scr.2014.11.003. [DOI] [PubMed] [Google Scholar]
  46. Hwang-Verslues WW, Chang PH, Jeng YM, Kuo WH, Chiang PH, Chang YC, Hsieh TH, Su FY, Lin LC, Abbondante S, Yang CY, Hsu HM, Yu JC, Chang KJ, Shew JY, Lee EY, Lee WH. Loss of corepressor PER2 under hypoxia up-regulates OCT1-mediated EMT gene expression and enhances tumor malignancy. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(30):12331–12336. doi: 10.1073/pnas.1222684110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  47. Inoue H, Nagata N, Kurokawa H, Yamanaka S. iPS cells: a game changer for future medicine. The EMBO journal. 2014;33(5):409–417. doi: 10.1002/embj.201387098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Jackson KA, Mi T, Goodell MA. Hematopoietic potential of stem cells isolated from murine skeletal muscle. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(25):14482–14486. doi: 10.1073/pnas.96.25.14482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Jiang H, Lin X, Liu Y, Gong W, Ma X, Yu Y, Xie Y, Sun X, Feng Y, Janzen V, Chen T. Transformation of epithelial ovarian cancer stemlike cells into mesenchymal lineage via EMT results in cellular heterogeneity and supports tumor engraftment. Molecular medicine. 2012;18:1197–1208. doi: 10.2119/molmed.2012.00075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Jin L, Hope KJ, Zhai Q, Smadja-Joffe F, Dick JE. Targeting of CD44 eradicates human acute myeloid leukemic stem cells. Nature medicine. 2006;12(10):1167–1174. doi: 10.1038/nm1483. [DOI] [PubMed] [Google Scholar]
  51. Jolly MK, Tripathi SC, Jia D, Mooney SM, Celiktas M, Hanash SM, Mani SA, Pienta KJ, Ben-Jacob E, Levine H. Stability of the hybrid epithelial/mesenchymal phenotype. Oncotarget. 2016 doi: 10.18632/oncotarget.8166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Jonker JW, Freeman J, Bolscher E, Musters S, Alvi AJ, Titley I, Schinkel AH, Dale TC. Contribution of the ABC transporters Bcrp1 and Mdr1a/1b to the side population phenotype in mammary gland and bone marrow of mice. Stem cells. 2005;23(8):1059–1065. doi: 10.1634/stemcells.2005-0150. [DOI] [PubMed] [Google Scholar]
  53. Kalcheim C. Epithelial-Mesenchymal Transitions during Neural Crest and Somite Development. Journal of clinical medicine. 2015;5(1) doi: 10.3390/jcm5010001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Kalluri R, Weinberg RA. The basics of epithelial-mesenchymal transition. The Journal of clinical investigation. 2009;119(6):1420–1428. doi: 10.1172/JCI39104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  55. Kanatsu M, Nishikawa SI. In vitro analysis of epiblast tissue potency for hematopoietic cell differentiation. Development. 1996;122(3):823–830. doi: 10.1242/dev.122.3.823. [DOI] [PubMed] [Google Scholar]
  56. Kavanagh DP, Robinson J, Kalia N. Mesenchymal stem cell priming: fine-tuning adhesion and function. Stem cell reviews. 2014;10(4):587–599. doi: 10.1007/s12015-014-9510-7. [DOI] [PubMed] [Google Scholar]
  57. Kawamura K, Fujiwara S, Sugino YM. Budding-specific lectin induced in epithelial cells is an extracellular matrix component for stem cell aggregation in tunicates. Development. 1991;113(3):995–1005. doi: 10.1242/dev.113.3.995. [DOI] [PubMed] [Google Scholar]
  58. Lamouille S, Xu J, Derynck R. Molecular mechanisms of epithelial-mesenchymal transition. Nature reviews Molecular cell biology. 2014;15(3):178–196. doi: 10.1038/nrm3758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Levings PP, McGarry SV, Currie TP, Nickerson DM, McClellan S, Ghivizzani SC, Steindler DA, Gibbs CP. Expression of an exogenous human Oct-4 promoter identifies tumor-initiating cells in osteosarcoma. Cancer research. 2009;69(14):5648–5655. doi: 10.1158/0008-5472.CAN-08-3580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Li F, Lu S, Vida L, Thomson JA, Honig GR. Bone morphogenetic protein 4 induces efficient hematopoietic differentiation of rhesus monkey embryonic stem cells in vitro. Blood. 2001;98(2):335–342. doi: 10.1182/blood.v98.2.335. [DOI] [PubMed] [Google Scholar]
  61. Li F, Niyibizi C. Cells derived from murine induced pluripotent stem cells (iPSC) by treatment with members of TGF-beta family give rise to osteoblasts differentiation and form bone in vivo. BMC cell biology. 2012;13:35. doi: 10.1186/1471-2121-13-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Li R, Liang J, Ni S, Zhou T, Qing X, Li H, He W, Chen J, Li F, Zhuang Q, Qin B, Xu J, Li W, Yang J, Gan Y, Qin D, Feng S, Song H, Yang D, Zhang B, Zeng L, Lai L, Esteban MA, Pei D. A mesenchymal-to-epithelial transition initiates and is required for the nuclear reprogramming of mouse fibroblasts. Cell stem cell. 2010;7(1):51–63. doi: 10.1016/j.stem.2010.04.014. [DOI] [PubMed] [Google Scholar]
  63. Liang G, Zhang Y. Embryonic stem cell and induced pluripotent stem cell: an epigenetic perspective. Cell research. 2013;23(1):49–69. doi: 10.1038/cr.2012.175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Lin H, Lei J, Wininger D, Nguyen MT, Khanna R, Hartmann C, Yan WL, Huang SC. Multilineage potential of homozygous stem cells derived from metaphase II oocytes. Stem cells. 2003;21(2):152–161. doi: 10.1634/stemcells.21-2-152. [DOI] [PubMed] [Google Scholar]
  65. Lin Y, Li XY, Willis AL, Liu C, Chen G, Weiss SJ. Snail1-dependent control of embryonic stem cell pluripotency and lineage commitment. Nature communications. 2014;5:3070. doi: 10.1038/ncomms4070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Liu M, Casimiro MC, Wang C, Shirley LA, Jiao X, Katiyar S, Ju X, Li Z, Yu Z, Zhou J, Johnson M, Fortina P, Hyslop T, Windle JJ, Pestell RG. p21CIP1 attenuates Ras- and c-Myc-dependent breast tumor epithelial mesenchymal transition and cancer stem cell-like gene expression in vivo. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(45):19035–19039. doi: 10.1073/pnas.0910009106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Liu X, Sun H, Qi J, Wang L, He S, Liu J, Feng C, Chen C, Li W, Guo Y, Qin D, Pan G, Chen J, Pei D, Zheng H. Sequential introduction of reprogramming factors reveals a time-sensitive requirement for individual factors and a sequential EMT-MET mechanism for optimal reprogramming. Nature cell biology. 2013;15(7):829–838. doi: 10.1038/ncb2765. [DOI] [PubMed] [Google Scholar]
  68. Lluis F, Pedone E, Pepe S, Cosma MP. Periodic activation of Wnt/beta-catenin signaling enhances somatic cell reprogramming mediated by cell fusion. Cell stem cell. 2008;3(5):493–507. doi: 10.1016/j.stem.2008.08.017. [DOI] [PubMed] [Google Scholar]
  69. Lovisa S, LeBleu VS, Tampe B, Sugimoto H, Vadnagara K, Carstens JL, Wu CC, Hagos Y, Burckhardt BC, Pentcheva-Hoang T, Nischal H, Allison JP, Zeisberg M, Kalluri R. Epithelial-to-mesenchymal transition induces cell cycle arrest and parenchymal damage in renal fibrosis. Nature medicine. 2015;21(9):998–1009. doi: 10.1038/nm.3902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  70. Maekawa M, Yamaguchi K, Nakamura T, Shibukawa R, Kodanaka I, Ichisaka T, Kawamura Y, Mochizuki H, Goshima N, Yamanaka S. Direct reprogramming of somatic cells is promoted by maternal transcription factor Glis1. Nature. 2011;474(7350):225–229. doi: 10.1038/nature10106. [DOI] [PubMed] [Google Scholar]
  71. Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, Campbell LL, Polyak K, Brisken C, Yang J, Weinberg RA. The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 2008;133(4):704–715. doi: 10.1016/j.cell.2008.03.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Martinez-Estrada OM, Lettice LA, Essafi A, Guadix JA, Slight J, Velecela V, Hall E, Reichmann J, Devenney PS, Hohenstein P, Hosen N, Hill RE, Munoz-Chapuli R, Hastie ND. Wt1 is required for cardiovascular progenitor cell formation through transcriptional control of Snail and E-cadherin. Nature genetics. 2010;42(1):89–93. doi: 10.1038/ng.494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Martinez-Morales PL, Diez del Corral R, Olivera-Martinez I, Quiroga AC, Das RM, Barbas JA, Storey KG, Morales AV. FGF and retinoic acid activity gradients control the timing of neural crest cell emigration in the trunk. The Journal of cell biology. 2011;194(3):489–503. doi: 10.1083/jcb.201011077. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Morales AV, Acloque H, Ocana OH, de Frutos CA, Gold V, Nieto MA. Snail genes at the crossroads of symmetric and asymmetric processes in the developing mesoderm. EMBO reports. 2007;8(1):104–109. doi: 10.1038/sj.embor.7400867. [DOI] [PMC free article] [PubMed] [Google Scholar]
  75. Nakaya Y, Sheng G. Epithelial to mesenchymal transition during gastrulation: an embryological view. Development, growth & differentiation. 2008;50(9):755–766. doi: 10.1111/j.1440-169X.2008.01070.x. [DOI] [PubMed] [Google Scholar]
  76. Nakaya Y, Sukowati EW, Wu Y, Sheng G. RhoA and microtubule dynamics control cell-basement membrane interaction in EMT during gastrulation. Nature cell biology. 2008;10(7):765–775. doi: 10.1038/ncb1739. [DOI] [PubMed] [Google Scholar]
  77. Ng YH, Zhu H, Leung PC. Twist modulates human trophoblastic cell invasion via regulation of N-cadherin. Endocrinology. 2012;153(2):925–936. doi: 10.1210/en.2011-1488. [DOI] [PubMed] [Google Scholar]
  78. Niwa H, Toyooka Y, Shimosato D, Strumpf D, Takahashi K, Yagi R, Rossant J. Interaction between Oct3/4 and Cdx2 determines trophectoderm differentiation. Cell. 2005;123(5):917–929. doi: 10.1016/j.cell.2005.08.040. [DOI] [PubMed] [Google Scholar]
  79. Noggle S, Fung HL, Gore A, Martinez H, Satriani KC, Prosser R, Oum K, Paull D, Druckenmiller S, Freeby M, Greenberg E, Zhang K, Goland R, Sauer MV, Leibel RL, Egli D. Human oocytes reprogram somatic cells to a pluripotent state. Nature. 2011;478(7367):70–75. doi: 10.1038/nature10397. [DOI] [PubMed] [Google Scholar]
  80. O’Brien CA, Pollett A, Gallinger S, Dick JE. A human colon cancer cell capable of initiating tumour growth in immunodeficient mice. Nature. 2007;445(7123):106–110. doi: 10.1038/nature05372. [DOI] [PubMed] [Google Scholar]
  81. Ocana OH, Corcoles R, Fabra A, Moreno-Bueno G, Acloque H, Vega S, Barrallo-Gimeno A, Cano A, Nieto MA. Metastatic colonization requires the repression of the epithelial-mesenchymal transition inducer Prrx1. Cancer cell. 2012;22(6):709–724. doi: 10.1016/j.ccr.2012.10.012. [DOI] [PubMed] [Google Scholar]
  82. Ohta S, Schoenwolf GC, Yamada G. The cessation of gastrulation: BMP signaling and EMT during and at the end of gastrulation. Cell adhesion & migration. 2010;4(3):440–446. doi: 10.4161/cam.4.3.12000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Park SY, Kim MJ, Park SA, Kim JS, Min KN, Kim DK, Lim W, Nam JS, Sheen YY. Combinatorial TGF-beta attenuation with paclitaxel inhibits the epithelial-to-mesenchymal transition and breast cancer stem-like cells. Oncotarget. 2015;6(35):37526–37543. doi: 10.18632/oncotarget.6063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Pasti G, Labouesse M. Epithelial junctions, cytoskeleton, and polarity. WormBook : the online review of C elegans biology. 2014:1–35. doi: 10.1895/wormbook.1.56.2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Pfister K, Shook DR, Chang C, Keller R, Skoglund P. Molecular model for force production and transmission during vertebrate gastrulation. Development. 2016;143(4):715–727. doi: 10.1242/dev.128090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  86. Pittenger MF, Martin BJ. Mesenchymal stem cells and their potential as cardiac therapeutics. Circulation research. 2004;95(1):9–20. doi: 10.1161/01.RES.0000135902.99383.6f. [DOI] [PubMed] [Google Scholar]
  87. Ponti D, Costa A, Zaffaroni N, Pratesi G, Petrangolini G, Coradini D, Pilotti S, Pierotti MA, Daidone MG. Isolation and in vitro propagation of tumorigenic breast cancer cells with stem/progenitor cell properties. Cancer research. 2005;65(13):5506–5511. doi: 10.1158/0008-5472.CAN-05-0626. [DOI] [PubMed] [Google Scholar]
  88. Puisieux A, Brabletz T, Caramel J. Oncogenic roles of EMT-inducing transcription factors. Nature cell biology. 2014;16(6):488–494. doi: 10.1038/ncb2976. [DOI] [PubMed] [Google Scholar]
  89. Ricci-Vitiani L, Lombardi DG, Pilozzi E, Biffoni M, Todaro M, Peschle C, De Maria R. Identification and expansion of human colon-cancer-initiating cells. Nature. 2007;445(7123):111–115. doi: 10.1038/nature05384. [DOI] [PubMed] [Google Scholar]
  90. Rowton M, Ramos P, Anderson DM, Rhee JM, Cunliffe HE, Rawls A. Regulation of mesenchymal-to-epithelial transition by PARAXIS during somitogenesis. Developmental dynamics : an official publication of the American Association of Anatomists. 2013;242(11):1332–1344. doi: 10.1002/dvdy.24033. [DOI] [PubMed] [Google Scholar]
  91. Ruscetti M, Quach B, Dadashian EL, Mulholland DJ, Wu H. Tracking and Functional Characterization of Epithelial-Mesenchymal Transition and Mesenchymal Tumor Cells during Prostate Cancer Metastasis. Cancer research. 2015;75(13):2749–2759. doi: 10.1158/0008-5472.CAN-14-3476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Saiz N, Plusa B. Early cell fate decisions in the mouse embryo. Reproduction. 2013;145(3):R65–80. doi: 10.1530/REP-12-0381. [DOI] [PubMed] [Google Scholar]
  93. Sasaki H. Mechanisms of trophectoderm fate specification in preimplantation mouse development. Development, growth & differentiation. 2010;52(3):263–273. doi: 10.1111/j.1440-169X.2009.01158.x. [DOI] [PubMed] [Google Scholar]
  94. Schatton T, Murphy GF, Frank NY, Yamaura K, Waaga-Gasser AM, Gasser M, Zhan Q, Jordan S, Duncan LM, Weishaupt C, Fuhlbrigge RC, Kupper TS, Sayegh MH, Frank MH. Identification of cells initiating human melanomas. Nature. 2008;451(7176):345–349. doi: 10.1038/nature06489. [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Schneider DJ, Wu M, Le TT, Cho SH, Brenner MB, Blackburn MR, Agarwal SK. Cadherin-11 contributes to pulmonary fibrosis: potential role in TGF-beta production and epithelial to mesenchymal transition. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2012;26(2):503–512. doi: 10.1096/fj.11-186098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Sheng G. Epiblast morphogenesis before gastrulation. Developmental biology. 2015;401(1):17–24. doi: 10.1016/j.ydbio.2014.10.003. [DOI] [PubMed] [Google Scholar]
  97. Singh SK, Hawkins C, Clarke ID, Squire JA, Bayani J, Hide T, Henkelman RM, Cusimano MD, Dirks PB. Identification of human brain tumour initiating cells. Nature. 2004;432(7015):396–401. doi: 10.1038/nature03128. [DOI] [PubMed] [Google Scholar]
  98. Slawny N, O’Shea KS. Geminin promotes an epithelial-to-mesenchymal transition in an embryonic stem cell model of gastrulation. Stem cells and development. 2013;22(8):1177–1189. doi: 10.1089/scd.2012.0050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Son MJ, Woolard K, Nam DH, Lee J, Fine HA. SSEA-1 is an enrichment marker for tumor-initiating cells in human glioblastoma. Cell stem cell. 2009;4(5):440–452. doi: 10.1016/j.stem.2009.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Stankova V, Tsikolia N, Viebahn C. Rho kinase activity controls directional cell movements during primitive streak formation in the rabbit embryo. Development. 2015;142(1):92–98. doi: 10.1242/dev.111583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  101. Stewart CA, Wang Y, Bonilla-Claudio M, Martin JF, Gonzalez G, Taketo MM, Behringer RR. CTNNB1 in mesenchyme regulates epithelial cell differentiation during Mullerian duct and postnatal uterine development. Molecular endocrinology. 2013;27(9):1442–1454. doi: 10.1210/me.2012-1126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  102. Strumpf D, Mao CA, Yamanaka Y, Ralston A, Chawengsaksophak K, Beck F, Rossant J. Cdx2 is required for correct cell fate specification and differentiation of trophectoderm in the mouse blastocyst. Development. 2005;132(9):2093–2102. doi: 10.1242/dev.01801. [DOI] [PubMed] [Google Scholar]
  103. Takahashi K, Tanabe K, Ohnuki M, Narita M, Ichisaka T, Tomoda K, Yamanaka S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell. 2007;131(5):861–872. doi: 10.1016/j.cell.2007.11.019. [DOI] [PubMed] [Google Scholar]
  104. Takahashi K, Yamanaka S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell. 2006;126(4):663–676. doi: 10.1016/j.cell.2006.07.024. [DOI] [PubMed] [Google Scholar]
  105. Taube JH, Herschkowitz JI, Komurov K, Zhou AY, Gupta S, Yang J, Hartwell K, Onder TT, Gupta PB, Evans KW, Hollier BG, Ram PT, Lander ES, Rosen JM, Weinberg RA, Mani SA. Core epithelial-to-mesenchymal transition interactome gene-expression signature is associated with claudin-low and metaplastic breast cancer subtypes. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(35):15449–15454. doi: 10.1073/pnas.1004900107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Theveneau E, Mayor R. Neural crest delamination and migration: from epithelium-to-mesenchyme transition to collective cell migration. Developmental biology. 2012;366(1):34–54. doi: 10.1016/j.ydbio.2011.12.041. [DOI] [PubMed] [Google Scholar]
  107. Thiery JP, Acloque H, Huang RY, Nieto MA. Epithelial-mesenchymal transitions in development and disease. Cell. 2009;139(5):871–890. doi: 10.1016/j.cell.2009.11.007. [DOI] [PubMed] [Google Scholar]
  108. Thomson JA, Itskovitz-Eldor J, Shapiro SS, Waknitz MA, Swiergiel JJ, Marshall VS, Jones JM. Embryonic stem cell lines derived from human blastocysts. Science. 1998;282(5391):1145–1147. doi: 10.1126/science.282.5391.1145. [DOI] [PubMed] [Google Scholar]
  109. Tondreau T, Meuleman N, Delforge A, Dejeneffe M, Leroy R, Massy M, Mortier C, Bron D, Lagneaux L. Mesenchymal stem cells derived from CD133-positive cells in mobilized peripheral blood and cord blood: proliferation, Oct4 expression, and plasticity. Stem cells. 2005;23(8):1105–1112. doi: 10.1634/stemcells.2004-0330. [DOI] [PubMed] [Google Scholar]
  110. Toyooka Y, Oka S, Fujimori T. Early preimplantation cells expressing Cdx2 exhibit plasticity of specification to TE and ICM lineages through positional changes. Developmental biology. 2016;411(1):50–60. doi: 10.1016/j.ydbio.2016.01.011. [DOI] [PubMed] [Google Scholar]
  111. Trembley MA, Velasquez LS, de Mesy Bentley KL, Small EM. Myocardin-related transcription factors control the motility of epicardium-derived cells and the maturation of coronary vessels. Development. 2015;142(1):21–30. doi: 10.1242/dev.116418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Trempus CS, Morris RJ, Ehinger M, Elmore A, Bortner CD, Ito M, Cotsarelis G, Nijhof JG, Peckham J, Flagler N, Kissling G, Humble MM, King LC, Adams LD, Desai D, Amin S, Tennant RW. CD34 expression by hair follicle stem cells is required for skin tumor development in mice. Cancer research. 2007;67(9):4173–4181. doi: 10.1158/0008-5472.CAN-06-3128. [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Visus C, Wang Y, Lozano-Leon A, Ferris RL, Silver S, Szczepanski MJ, Brand RE, Ferrone CR, Whiteside TL, Ferrone S, DeLeo AB, Wang X. Targeting ALDH(bright) human carcinoma-initiating cells with ALDH1A1-specific CD8(+) T cells. Clinical cancer research : an official journal of the American Association for Cancer Research. 2011;17(19):6174–6184. doi: 10.1158/1078-0432.CCR-11-1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Visvader JE, Clevers H. Tissue-specific designs of stem cell hierarchies. Nature cell biology. 2016;18(4):349–355. doi: 10.1038/ncb3332. [DOI] [PubMed] [Google Scholar]
  115. Wang H, Diao D, Shi Z, Zhu X, Gao Y, Gao S, Liu X, Wu Y, Rudolph KL, Liu G, Li T, Ju Z. SIRT6 Controls Hematopoietic Stem Cell Homeostasis through Epigenetic Regulation of Wnt Signaling. Cell stem cell. 2016;18(4):495–507. doi: 10.1016/j.stem.2016.03.005. [DOI] [PubMed] [Google Scholar]
  116. Wang S, Garcia AJ, Wu M, Lawson DA, Witte ON, Wu H. Pten deletion leads to the expansion of a prostatic stem/progenitor cell subpopulation and tumor initiation. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(5):1480–1485. doi: 10.1073/pnas.0510652103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Wei SC, Fattet L, Tsai JH, Guo Y, Pai VH, Majeski HE, Chen AC, Sah RL, Taylor SS, Engler AJ, Yang J. Matrix stiffness drives epithelial-mesenchymal transition and tumour metastasis through a TWIST1-G3BP2 mechanotransduction pathway. Nature cell biology. 2015;17(5):678–688. doi: 10.1038/ncb3157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Wellner U, Schubert J, Burk UC, Schmalhofer O, Zhu F, Sonntag A, Waldvogel B, Vannier C, Darling D, zur Hausen A, Brunton VG, Morton J, Sansom O, Schuler J, Stemmler MP, Herzberger C, Hopt U, Keck T, Brabletz S, Brabletz T. The EMT-activator ZEB1 promotes tumorigenicity by repressing stemness-inhibiting microRNAs. Nature cell biology. 2009;11(12):1487–1495. doi: 10.1038/ncb1998. [DOI] [PubMed] [Google Scholar]
  119. Weng M, Wieschaus E. Myosin-dependent remodeling of adherens junctions protects junctions from Snail-dependent disassembly. The Journal of cell biology. 2016;212(2):219–229. doi: 10.1083/jcb.201508056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Xiao Z, Patrakka J, Nukui M, Chi L, Niu D, Betsholtz C, Pikkarainen T, Vainio S, Tryggvason K. Deficiency in Crumbs homolog 2 (Crb2) affects gastrulation and results in embryonic lethality in mice. Developmental dynamics : an official publication of the American Association of Anatomists. 2011;240(12):2646–2656. doi: 10.1002/dvdy.22778. [DOI] [PubMed] [Google Scholar]
  121. Xie G, Ji A, Yuan Q, Jin Z, Yuan Y, Ren C, Guo Z, Yao Q, Yang K, Lin X, Chen L. Tumour-initiating capacity is independent of epithelial-mesenchymal transition status in breast cancer cell lines. British journal of cancer. 2014;110(10):2514–2523. doi: 10.1038/bjc.2014.153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  122. Yamada M, Johannesson B, Sagi I, Burnett LC, Kort DH, Prosser RW, Paull D, Nestor MW, Freeby M, Greenberg E, Goland RS, Leibel RL, Solomon SL, Benvenisty N, Sauer MV, Egli D. Human oocytes reprogram adult somatic nuclei of a type 1 diabetic to diploid pluripotent stem cells. Nature. 2014;510(7506):533–536. doi: 10.1038/nature13287. [DOI] [PubMed] [Google Scholar]
  123. Yamakoshi S, Bai R, Chaen T, Ideta A, Aoyagi Y, Sakurai T, Konno T, Imakawa K. Expression of mesenchymal-related genes by the bovine trophectoderm following conceptus attachment to the endometrial epithelium. Reproduction. 2012;143(3):377–387. doi: 10.1530/REP-11-0364. [DOI] [PubMed] [Google Scholar]
  124. Ye X, Tam WL, Shibue T, Kaygusuz Y, Reinhardt F, Ng Eaton E, Weinberg RA. Distinct EMT programs control normal mammary stem cells and tumour-initiating cells. Nature. 2015;525(7568):256–260. doi: 10.1038/nature14897. [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Yin L, Castagnino P, Assoian RK. ABCG2 expression and side population abundance regulated by a transforming growth factor beta-directed epithelial-mesenchymal transition. Cancer research. 2008;68(3):800–807. doi: 10.1158/0008-5472.CAN-07-2545. [DOI] [PubMed] [Google Scholar]
  126. Yoshino T, Saito D, Atsuta Y, Uchiyama C, Ueda S, Sekiguchi K, Takahashi Y. Interepithelial signaling with nephric duct is required for the formation of overlying coelomic epithelial cell sheet. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(18):6660–6665. doi: 10.1073/pnas.1316728111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Yu JM, Sun W, Hua F, Xie J, Lin H, Zhou DD, Hu ZW. BCL6 induces EMT by promoting the ZEB1-mediated transcription repression of E-cadherin in breast cancer cells. Cancer letters. 2015;365(2):190–200. doi: 10.1016/j.canlet.2015.05.029. [DOI] [PubMed] [Google Scholar]
  128. Zavadil J, Bitzer M, Liang D, Yang YC, Massimi A, Kneitz S, Piek E, Bottinger EP. Genetic programs of epithelial cell plasticity directed by transforming growth factor-beta. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(12):6686–6691. doi: 10.1073/pnas.111614398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Zheng X, Carstens JL, Kim J, Scheible M, Kaye J, Sugimoto H, Wu CC, LeBleu VS, Kalluri R. Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic cancer. Nature. 2015;527(7579):525–530. doi: 10.1038/nature16064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  130. Zhou JN, Zeng Q, Wang HY, Zhang B, Li ST, Nan X, Cao N, Fu CJ, Yan XL, Jia YL, Wang JX, Zhao AH, Li ZW, Li YH, Xie XY, Zhang XM, Dong Y, Xu YC, He LJ, Yue W, Pei XT. MicroRNA-125b attenuates epithelial-mesenchymal transitions and targets stem-like liver cancer cells through small mothers against decapentaplegic 2 and 4. Hepatology. 2015;62(3):801–815. doi: 10.1002/hep.27887. [DOI] [PubMed] [Google Scholar]

RESOURCES