Skip to main content
Erschienen in: Molecular Cancer 1/2013

Open Access 01.12.2013 | Review

Epithelial-mesenchymal transition: focus on metastatic cascade, alternative splicing, non-coding RNAs and modulating compounds

verfasst von: Timur R Samatov, Alexander G Tonevitsky, Udo Schumacher

Erschienen in: Molecular Cancer | Ausgabe 1/2013

Abstract

Epithelial-mesenchymal transition (EMT) is a key process in embryonic development and metastases formation during malignant progression. This review focuses on transcriptional regulation, non-coding RNAs, alternative splicing events and cell adhesion molecules regulation during EMT. Additionally, we summarize the knowledge with regard to the small potentially druggable molecules capable of modulating EMT for cancer therapy.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1476-4598-12-107) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors’ contribution

TRS wrote the first draft of the article, AGT and US finalized the manuscript. All authors read and approved the final manuscript.
Abkürzungen
EMT
Epithelial-mesenchymal transition
MET
Mesenchymal-epithelial transition
TGFβ
Transforming growth factor β
CAM
Cell adhesion molecule
CTC
Circulating tumor cell
miRNA
MicroRNA
lncRNA
Long non-coding RNA.

Introduction

Epithelial-mesenchymal transition (EMT) is a multi-step morphogenetic process during which epithelial cells downregulate their epithelial properties and upregulate mesenchymal characteristics (Figure 1). Namely, static epithelial cells lose cell to cell junctions and as a consequence they lose apico-basal polarity to become migratory mesenchymal-like cells. This process of down-regulation of the epithelial phenotype mimics the normal developmental process of gastrulation, in which cells from the epithelial sheet of the ectoderm start to form the third germinal layer, the mesoderm, whose migratory cells are called mesenchymal cells. This process is therefore aptly called the epithelial-mesenchymal transition, which is currently classified into three subtypes[1].
Type 1 EMT is associated with the original embryonic development and also occurs during postnatal growth. The steps of this EMT type are specific and well-defined. Epithelial cells are cuboidal to cylindrical in shape and are in contact with each other via adherent and tight junctions. Primary migratory mesenchymal cells generated this way may potentially go through a reverse step to become epithelia again. This step is called the mesenchymal-epithelial transition (MET) and generates secondary epithelia in the developing embryo[2]. Differentiated cells in almost all organs in adults developed as a result of EMT-MET.
Type 2 EMT is initiated by injury and results in generation of fibroblasts to rebuild wounded tissues[3]. During inflammation fibroblasts and immune cells release cytokines and other pro-inflammatory factors as well as extracellular matrix proteins which results in stimulation of cells to undergo EMT. If inflammation pathologically persists, continuous EMT of normal epithelial cells can result in fibrosis and organ damage[4].
Oncogenic type 3 EMT enables epithelial cells to acquire invasive mesenchymal phenotype characteristics which are essential in metastatic spread[5]. Typical developmental EMT features are recapitulated in oncogenic EMT[6], however, they are less ordered and coordinated. As a result of this disordered EMT, hybrid phenotypes can often arise having the properties of both epithelial and mesenchymal cell types[7].

Transcription factors regulating EMT

There are a number of transcription factors known to be involved in the regulation of EMT. The most characterized are ZEB 1 and ZEB 2, snail, slug and twist (Figure 2).
ZEB 1 and ZEB 2 are highly conserved zinc finger proteins which can directly bind to the promoter regions of target genes and thus repress the expression of E-cadherin and some other epithelial markers[8] and induce the expression of vimentin and a number of other mesenchymal markers[9]. ZEB 1 and 2 are induced by TGFβ, hypoxic conditions and inflammatory cytokines, factors which all initiate EMT. ZEBs play an important role in normal embryonic development and they are reported to be upregulated in many tumors[10].
Snail and slug belong to the snail family of transcription factors, with C-terminal zinc finger binding to E-boxes of the regulatory regions of target genes[11]. Snail factors repress E-cadherin expression by direct binding to its promoter and can also repress other epithelial proteins including desmoplakin and claudins. At the same time snail proteins activate expression of pro-invasive genes (vimentin, fibronectin, MMPs) promoting cell migration[12]. Like the two ZEB transcription factors, snail and slug can be induced by TGFβ, hypoxic conditions and other EMT-related signaling pathways[13]. Snail transcription factors are not present in normal epithelial cells, however they are found in the invasive front of tumors and considered to be prognostic factors for poor survival in a number of carcinomas[11].
The twist protein contains a basic/helix-loop-helix domain which provides for binding to DNA and dimerization. Its C-terminal end contains a “twist box” responsible for both transcriptional activation (e. g. for N-cadherin) and repression (E-cadherin)[14]. Regulation of genes by twist depends on its binding to other transcriptional factors, post-translational modifications, and choice of partner for dimerization. Twist is upregulated in human cancers and its abundancy increases during tumor progression. Its expression also correlates with higher tumor grade, invasiveness, and metastasis, cellular processes being considered as prognostic factors for enhanced tumor aggressiveness, tumor recurrence, and poorer survival[11].
Remarkably, there is a significant overlap in the regulatory signals of these transcription factors. Namely, expression of ZEB factors is regulated by snail[15, 16]. Snail in turn also increases the stability of twist which then activates the transcription of slug[15, 17]. This interaction network may play a role in spatial and temporal regulation of EMT.

EMT and metastatic cascade

One of the classical models for cancer metastasis is Stephen Paget’s seed and soil hypothesis in which the tumor cell is the seed and the organ in which the metastasis grows is the soil[18]. This model implies that certain tumor cells have an affinity to the particular organ which provides a growth advantage to them. Thus the site of metastasis is dependent on the affinity of the tumor for the given microenvironment, which elegantly explains why some organs (lung, liver, bone marrow) are particularly prone to harbour metastases while others are not (intestine, skeletal muscle, skin). After passing the endothelial barrier, additional factors such as local growth factor production play a role in stimulating the growth of these evasive tumor cells[19]. According to this later expanded model, metastasis formation starts when the primary malignant cell divides and once the cell mass has reached the size of a few dozens cells, it sends out angiogenic signals, thus leading to the ingrowth of blood vessels into the newly formed tumor. As a next step, future metastatic cells have to free themselves from the primary tumor mass, have to degrade the surrounding extracellular matrix including the basement membrane, must enter the blood vessels and survive within the circulation (= the seed). Once they have reached the target organ of the future metastasis (= the soil), the tumor cell has to attach to the endothelium in this organ and has to migrate through it. When this process is accomplished, the metastatic cancer cell has - probably under the influence of local growth factors – to start to divide again in order to form a clinically detectable metastasis. Once proliferation has started, this metatsatic cycle resumes in order to spawn further metastases originating from a metastasis.
Different cell adhesion molecules (CAMs) play vital and opposing roles during this process. Due to their very epithelial nature cancer cells form more or less tight homologous epithelial cell to epithelial cell contacts at the site of the primary tumors. Molecularly this encompasses often homologous CAMs which are part of desmosomes, tight junctions and gap junctions (see Table 1). In addition, cell to basal lamina contacts (focal adhesions, hemidesmosomes) are formed from those cells directly adjacent to a basal lamina. In order to escape from the primary tumor, the proteins forming these junctions have to be down-regulated in order to allow cell migration. The contact of epithelial cells to the basal lamina not only hinders migration but also prevents cell death. If a normal epithelial cell looses the contact to the basal lamina, a special form of detachment-induced apoptosis, termed anoikis by Steven Frisch[20], is triggered as the cell’s integrins are detached from their ligands in the basal lamina. As mesenchymal cells do not necessarily have a direct contact to the basal lamina, they are not subject to anoikis and the EMT would therefore aid survival of the loosened cancer cells.
Table 1
Homologous CAMs
Type of junction
Type of protein
Protein
Gene name
Function
Desmosome
Cadherin (calcium-dependent)
Desmoglein 1, Desmoglein 2, Desmoglein 3, Desmoglein 4
DSG1 DSG2, DSG3, DSG4
Play important roles in cell adhesion, ensuring that cells within tissues are bound together. Cadherins behave as both receptors and ligands.
  
Desmocollin 1, Desmocollin 2, Desmocollin 3, Desmocollin 4
DSC1 DSC2 DSC3
 
 
Catenin
Junction plakoglobin ( JUP)
JUP
JUP can bind to the desmoglein I.
Tight junctions
Claudins
Claudin 1
CLDN1
The main component of the tight junctions
Occludins
Occludin
OCLN
The main component of the tight junctions
Cadherin
E-cadherin
CDH1
Loss of E-cadherin function or expression has been implicated in cancer progression and metastasis. E-cadherin downregulation decreases the strength of cellular adhesion within a tissue, resulting in an increase of cellular motility. This in turn may allow cancer cells to cross the basement membrane and invade surrounding tissues[21].
  
F11 receptor (JCAM)
JAM-1
The ligand for the integrin LFA1, a platelet receptor
 
Catenins
α-(E, N,T), β-, δ-catenins, γ-catenin (or Junction plakoglobin, JUP)
CTNNA1 (CAP102), CTNNA2 (CAPR), CTNNA3 (VR22), CTNNB1, CTNND1, CTNND2, JUP
Catenins belong to a family of proteins found in complexes with cadherin cell adhesion molecules. The primary mechanical role of catenins is connecting cadherins to actin filaments, specifically in these adhesion junctions of epithelial cells[22]. β-catenin may play a role in telling the cell to stop proliferating, as there is no room for more cells in the area.
The role of catenin in EMT has also received a lot of recent attention for its contributions to cancer development. It has been shown that HIF-1α can induce the EMT pathway, as well as the Wnt/β-catenin signaling pathway, thus enhancing the invasive potential of LNCaP cells (human prostate cancer cells)[23]. As a result, it is possible that the EMT associated with upregulated HIF-1α is controlled by signals from this Wnt/β-catenin pathway[23]. Catenin and EMT interactions may also play a role in hepatocellular carcinoma. VEGF-B treatment of hepatoma carcinoma cells can cause α-catenin to move from its normal location on the membrane into the nucleus and E-cadherin expression to decrease, thus promoting EMT and tumor invasiveness[24].
JUP protein is the only known constituent common to submembranous plaques of both desmosomes and intermediate junctions. JUP also associates with classical cadherins such as E-cadherin; in that context. Plakoglobin is O-glycosylated.
Cingulin
Cingulin
CGN
Cingulin is specifically localized at tight junctions in epithelial cells, unlike ZO-1, which is also detected at adherens-type junctions in non-epithelial cells. Cingulin interacts with ZO-1 and several other tight junction proteins, in addition to interacting with actin and myosin[25, 26].
Actin
α-, β-, γ-actins
ACTA1, ACTA2, ACTB, ACTG1, ACTG2
Participates in many important cellular processes, including cell motility, cell division and cytokinesis, vesicle and organelle movement, cell signalling, and the establishment and maintenance of cell junctions and cell shape.
Gap junctions
Connexin (or hemichannel)
Connexins
GJA1, GJC1, GJB4 etc.
Connexins are assembled in groups of six to form hemichannels, or connexons, and two hemichannels then combine to form a gap junction. The connexin gene family is diverse, with 21 identified members in the sequenced human genome.
The molecules forming homologous epithelial cell to epithelial cell tight contacts.
After EMT has enabled the tumor cells to migrate out of the primary tumor, they have to enter circulation and survive within it (Figure 3). Later, they must adhere to the microvascular endothelial cells at the site of the target organ and by this adhesion they have to communicate to the endothelial cells to open their cell junctions. This allows the passage of the cancer cell through the endothelium to the connective tissue space of the host organ. Again, CAMs mediate this process, however, these CAMs are different from those forming the intra-epithelial cell adhesion. Here, heterologous CAMs mediating cell adhesion between different cell types – tumor cells and endothelial cells - are important. Similarly to the mimicry of the EMT, cancer cells evading circulation mimic the leukocyte adhesion cascade (see Table 2). The CAMs and their ligands used in this adhesion are selectin glycoconjugate ligands, integrins and their extracellular matrix ligands, ALCAM and ICAMs. In contrast to the epithelial CAMs, which were down-regulated during EMT, these CAMs were up-regulated as part of the mesenchymal phenotype during EMT. These down- and up-regulations of cell adhesion molecule expression are governed by transcription factors which are important during gastrulation including twist, snail, slug, brachyury and ZEB 1 and ZEB 2.
Table 2
Heterologous CAMs
Adhesion molecule (receptor)
Gene name
Localization and other information
Ligand
Gene name of the ligand
Localization of the ligand and other information
Integrins
Integrin alpha (CD11a)
ITGAL (CD11A, p180)
Integrin alpha combines with the beta 2 chain (ITGB2) to form the integrin lymphocyte function-associated antigen-1 (LFA-1). LFA-1 plays a central role in leukocyte intercellular adhesion through interactions with its ligands, ICAMs 1–3 (intercellular adhesion molecules 1 through 3), as a rolling and signaling molecule[27], and also functions in lymphocyte costimulatory signaling.
ICAM1 (CD54)
ICAM1
A member of the immunoglobulin superfamily. A glycoprotein which is typically expressed on endothelial cells and cells of the immune system.
Integrin beta-2 (CD18)
ITGB2
ICAM-1 can be induced by (IL-1) and (TNFα) and is expressed by the vascular endothelium, macrophages, and lymphocytes. ICAM-1 is a ligand for LFA-1 (integrin), a receptor found on leukocytes.
Integrin alpha M (ITGAM)
ITGAM (CD11B, CR3A)
Integrin alpha M is one protein subunit that forms the heterodimeric integrin alpha-M beta-2 (αMβ2) molecule, also known as macrophage-1 antigen (Mac-1) or complement receptor 3 (CR3). αMβ2 is expressed on the surface of many leukocytes involved in the innate immune system. It mediates leukocyte adhesion and migration.
Integrin alpha 4 (CD49d)
ITGA4
VLA4 (α4β1-integrin) is found on leukocytes and endothelial cells.
VCAM1[28]
VCAM1 (CD106)
VLA4-interections support lymphocyte rolling in venules of the central nervous system in conjunction with P-selectin or can directly mediate rapid adhesion independent of P-selectin engagement[27].
Integrin beta-1 (CD29)
ITGB1
Fibronectin
FN1
Fibronectin is a high-molecular weight glycoprotein of the extracellular matrix[29]. Insoluble cellular fibronectin is a major component of the extracellular matrix. It is secreted by various cells. Fibronectin plays a major role in cell adhesion, growth, migration, and differentiation.
     
Altered fibronectin expression, degradation, and organization are associated with a number of pathologies, including cancer and fibrosis[30].
Integrin
 
α4β7-integrin
MADCAM-1
MADCAM1
MADCAM-1 is a cell adhesion leukocyte receptor expressed by mucosal venules. It helps to direct lymphocyte traffic into mucosal tissues. It can bind both integrin alpha-4/beta-7 and L-selectin regulating both the passage and retention of leukocytes. Isoform 2 lacking the mucin-like domain may be specialized in supporting integrin alpha-4/beta-7-dependent adhesion strengthening, independent of L-selectin binding.
Selectins
P-selectin
SELP
P-selectin is expressed on activated endothelial cells and platelets. Synthesis of P-selectin can be induced by thrombin, leukotriene B4, complement fragment C5a, histamine, TNFα or LPS.
PSGL-1 (P-selectin glycoprotein ligand-1)
SELPLG (CD16)
PSGL-1 is found on white blood cells and endothelial cells. PSGL-1 can bind to all three members of the selectin family however it binds to P-selectin with the highest affinity.
  
P-selectin plays an active role in the rolling of leukocytes[27].
  
see above and: PSGL-1 was shown contribute to E-selectin-mediated initial leukocyte capture and rolling in vivo[31].
E-selectin (CD62E, ELAM-1)
SELE (CD62E, ELAM-1)
E-selectin is expressed on activated endothelial cells. E-selectin is not stored within the cell and has to be
PSGL-1
SELPLG (CD16)
transported to the cell surface. Synthesis of E-selectin follows shortly after P-selectin synthesis, induced by cytokines such as IL-1, TNFα and lipopolysaccharide (LPS). Shear forces can also affect E-selectin expression. E-selectin may interact indiscriminately with many glycoproteins and glycolipids[31].
ESL-1 (golgi glycoprotein 1)
GLG1
ESL-1 is a glycoprotein and a variant of a receptor for fibroblast growth factor.
ESL-1 is a major E-selectin ligand on leukocytes[31].
CD44
CD44
CD44 is expressed in a large number of mammalian cell types. This protein participates in a variety of cellular functions including lymphocyte activation, recirculation and homing, hematopoiesis, and tumor metastasis.
E-selectin was shown to play a pivotal role in mediating cell–cell interactions between breast cancer cells and endothelial monolayers during metastasis[32].
E-selectin plays an active role in the rolling of leukocytes[27].
The contribution of CD44 is significant only at the later stages of the leukocyte recruitment cascade[31].
GlyCAM-1
GLYCAM1
In breast cancer the splice variant 4 of CD44 was shown as a major E-selectin ligand in facilitating tumor cell migration across endothelial monolayers[32].
L-selectin (CD62L)
SELL (CD62L, LAM1)
L-selectin found on lymphocytes and preimplantation embryo. It plays important roles in lymphocyte-endothelial cell interactions.
GlyCAM-1 is a proteoglycan ligand expressed on cells of the high endothelial venules in lymph nodes.
CD34
CD34
A cell surface glycoprotein which functions as a cell-cell adhesion factor. It may also mediate the attachment of stem cells to bone marrow extracellular matrix or directly to stromal cells.
Cells expressing CD34 are normally found in the umbilical cord and bone marrow as hematopoietic cells, a subset of mesenchymal stem cells, endothelial progenitor cells, endothelial cells of blood vessels but not lymphatics (except pleural lymphatics). CD34 is also an important adhesion molecule and is required for T cells to enter lymph nodes. It is expressed on lymph node endothelia whereas the L-selectin to which it binds is on the T cell.
MADCAM-1
MADCAM1
MADCAM-1 is a cell adhesion leukocyte receptor expressed by mucosal venules. It helps to direct lymphocyte traffic into mucosal tissues. It can bind both integrin alpha-4/beta-7 and L-selectin, regulating both the passage and retention of leukocytes.
   
PSGL-1
SELPLG (CD16)
See above
The molecules which are responsible for the leukocyte adhesion cascade involved in the inflammatory response.
Circulating tumor cells (CTCs) are cells which have already separated from the tumor and entered the bloodstream. It has been demonstrated that the number of CTCs in blood is an important prognostic marker for breast[33], prostate[34], lung[35], bladder[36] and colon[37] cancer patients. CTCs are a heterogeneous population of tumor cells, some of them presumably underwent EMT and hence possess mesenchymal features, while others have not and still represent with a more epithelial phenotype. It has been demonstrated on groups of patients with distinct breast cancer stages that CTCs with mesenchymal markers are more typical for the late metastatic stage[38] and provide for the reliable prognosis of recurrence[39]. Another recently reported observation is that mesenchymal CTCs in patients with advanced cancer comprise multicellular clusters rather than single cells, in contrast to epithelial ones[40]. The authors explained this observation with the proliferation of the mesenchymal cell that has undergone EMT and after proliferation differentiated back into a more epithelially differentiated cell cluster which, however, seems contradictory to the typical individual mesenchymal phenotype. Alternatively, the authours hypothesized simultaneous EMT of a pre-existing cluster of CTCs in the bloodstream mediated by TGF-β released from platelets.
Despite the many efforts, the detection of CTCs still suffers from technical complexities and non-reliability of their isolation. These problems are due to the low abundance and heterogeneity of CTCs. The CellSearch and AdnaTest systems approved by the FDA in USA and by EU authorities, respectively, are based on the detection of epithelial markers. However, if the cells in the bloodstream are more of the mesenchymal phenotype, some important cell population might be missed by using these isolation techniques. Currently there is no reliable method and no defined list of markers for the detection of dedifferentiated EMT-derived CTCs[41].

EMT and alternative splicing

More than 88% of human pre-mRNAs are alternatively spliced, thus generating protein diversity in an organism[42]. Alternative splicing events are regulated in a cell- and tissue type-specific manner, at different developmental stages or in response to extra-cellular stimuli and activation of specific signalling pathways[43, 44]. As many of these processes occur during EMT, alternative splicing is of importance in EMT as well (Figure 2). Examples of the best characterized EMT-dependent alternatively spliced genes are FGFR2, CD44, p120-catenin and Mena.
The fibroblast growth factor receptor 2 (FGFR2) encodes for a fibroblast growth factor-activated transmembrane receptor tyrosine kinase and is the first discovered example of EMT-related alternative splicing[45]. The second half of the third extra-cellular immunoglobulin-like domain of the FGFR2 is encoded by one of two mutually exclusive exons IIIb (expressed in epithelial cells) or IIIc (characteristic for mesenchymal cells). The functional model suggests that epithelial cells expressing the FGFR2-IIIb form specifically interact with fibroblast growth factors produced by mesenchymal cells. Accordingly, the factors expressed by epithelial cells interact with FGFR2-IIIc[46]. These interactions have been demonstrated to be important during embryonic development and limb outgrowth and lung-branching morphogenesis. Remarkably, targeted down-regulation of mesenchymal-specific FGFR2-IIIc isoform was shown to decrease metastatic ability of TSU-PrI bladder cancer cells and to increase survival following in vivo inoculation in mice[47]. Interestingly, alternative splicing of a similar protein, namely FGFR3, is regulated by snoRNA HBII-180C[48]. This finding implies that non-coding RNAs regulate EMT through modulation of alternative splicing.
The CD44 gene encodes for a transmembrane protein which maintains tissue structure by mediating cell-cell adhesion[49]. The N-terminal domain of CD44 is extracellular and interacts with the extracellular matrix glycosaminoglycan hyaluronic acid (HA) facilitating the binding of a number of extracellular ligands. The formed complex initiates a downstream signaling cascade via the interaction of the intracellular domain with binding partners. The CD44 pre-mRNA comprises exons 1–5 at the 5’ end and exons 16–20 at the 3’ end that are spliced together into the standard isoform CD44s. This isoform is the smallest and is present on the membrane of most vertebrate cells. Between exons 5 and 16 are ten alternatively spliced variable exons (v1–v10). These alternatively spliced variants are longer than the standard isoform of CD44 and the proteins encoded by these variants show extended extracellular membrane-proximal regions which form a glycosylated stalk-like structure providing interaction sites for additional molecules[50]. The CD44E isoform containing exons v8–10 is predominantly expressed in epithelial cells correlating with the expression of E-cadherin[51]. Remarkably, induction of EMT in cultured cells resulted in a switch from CD44E to standard isoform, and expression of the latter was upregulated in human breast cancers and was correlated with the mesenchymal marker N-cadherin in these tumors[52].
Splicing is not a particular feature of CD44, indeed, CAMs in general are alternatively spliced. The most remarkable example is DSCAM (Down Syndrome Cell Adhesion Molecule) which has up to 18,000 splice isoforms[53]. This Ig-like receptor is involved in innate immunity and neural wiring and its gene is located on 21 chromosome.
p120-Catenin regulates cadherin stability and modulates Rho GTPase activity[54, 55]. The isoforms containing exons 2 and 3 are expressed in mesenchymal cells. Epithelial cells skip these exons producing a shorter protein isoform. Consistently, EMT induces the expression of mesenchymal p120-catenin isoform[56]. Rho GTPases are known to regulate actin cytoskeleton and cell motility[57]. The full-length mesenchymal isoform of p120-catenin can bind RhoA GTPase, reducing its activity, and promote migration and invasiveness of the cells[58].
Mena (also known as Enah, mammalian enabled homolog of Drosophila protein Ena) is expressed in various cell types and regulates the branching actin filaments[59]. The isoform which contains the exon 11a is characteristic for epithelial cells and is not found in mesenchymal cells. Remarkably, it has been also found to be expressed in primary tumor cells but not in invasive tumor cells[60]. So far it is not clear what kind of functional implications this protein has on EMT.
Recently genome-wide approaches were used to determine EMT-related alternative splicing signatures[61]. It was shown that EMT-related extensive changes in alternative splicing are regulated by epithelial splicing regulatory proteins 1 and 2 (ESRP1 and ESRP2)[62]. These proteins are present in epithelial cells. Their siRNA-mediated knock down resulted in a splicing switch of FGFR2, CD44, p120 and Mena genes to mesenchymal phenotype. The reverse effect was observed when the ectopic expression of ESRP1 and ESRP2 was performed in mesenchymal cells.
Thus, there are clearly distinct profiles of alternative splicing which allow discrimination between epithelial and mesenchymal cell types.

EMT and non-coding RNAs

MiRNAs are one family of small (20–22 nucleotides) non-coding RNAs. Their function is to regulate gene expression post-transcriptionally through binding to the sites which are perfectly complementary, or which may contain mismatches (“non-canonical sites”). These sites are located in 3’ UTRs, however recent reports demonstrate that miRNAs can also function through binding to other regions of target mRNAs[63, 64]. By binding to target mRNAs, miRNAs play important roles in regulating diverse biological processes[65]. These processes include regulation of the EMT, in which various miRNAs are involved[66]. Remarkably, the regulatory miRNA-mRNA networks can be rapidly regulated[67]. It should also be mentioned that another recently reported interesting function of miRNAs, which may play a certain role in EMT regulation, is paracrine-mode intercellular signaling[68].
The miR200 and the miR205 families were shown to be highly associated with EMT and a strong correlation between the expression of the miR200 family and E-cadherin expression in different cell lines and epithelial tissues has been demonstrated[69, 70]. During EMT, expression of miR-200 family is repressed by ZEB transcription factors. These factors in turn are the targets for miR-200 family thus comprising a double negative feedback loop[71]. It was shown recently that miR200c also regulates EMT through targeting fibronectin, moesin and other proteins that normally suppress cell migration and resistance to anoikis[72]. Moreover, the same lab found that miR200c targets a NF-κB-dependent neurotrophic tyrosine receptor kinase, which also suppresses resistance to anoikis, and this miRNA is down-regulated in highly aggressive triple negative breast cancers[73]. Other EMT-related downstream targets of the miR200 family are miR141 inhibiting TGFβ2[74] and miR200a suppressing β-catenin (CTNNB1)[75].
The EMT-related transcription factors have been described as transcriptional regulators of miRNAs as well. For example, miR21 is abundant in various tumors and known to induce metastasis through EMT. The promoter regions of miR21 contain consensus E-box sequences comprising binding sites for ZEB1[76]. Binding of ZEB1 induces transcription of miR21[77]. MiR10b is also known to be associated with cell migration, invasion, and metastasis of breast cancer cells. It was shown that the transcription factor twist can bind to the E-box element close to the predicted promoter of miR10b and activate its transcription, thus promoting twist-mediated EMT[77]. Overall regulation of miR10b is complex and context dependent: ZEB1 increases the expression of miR10b in colorectal cancer cells but decreases expression in breast cancer cells[74]. Similarly, snail reduces the expression of miR10b in human breast cancer cells[77]. These data suggest that miRNAs can be considered as markers for EMT through the activity of EMT-related transcription factors.
MiRNAs were shown to be associated with the TGFβ signaling pathway. The TGFβ-mediated induction of EMT in mammary epithelial cells results in loss of tight junctions and cell polarity and up-regulates the expression of miR155[78]. The target of miR155 is RhoA which is important for the control of actin cytoskeleton and cell invasion. RhoA contains three conserved regions which are potential binding sites for miR155[78]. Down-regulation of RhoA leads to actin cytoskeleton rearrangements and increased cell motility[79].
The TGFβ-induced EMT in mammary epithelial cells also leads to the higher expression levels of miR29a and miR21[78, 80]. Ectopic expression of miR29a suppresses the expression of tristetraprolin and promotes to EMT in cooperation with the Ras pathway[80].
It has been demonstrated that miR9 regulates the mRNA encoding for E-cadherin[81]. Hence the increased expression of miR9 induced EMT in human mammary epithelial cells[81].
Remarkably, it has been demonstrated recently that circulating miRNAs in plasma of metastatic breast cancer patients can indicate their CTC status[82]. Circulating miRNAs are easier to isolate and handle than CTCs, which will probably make them prognostic markers of choice in future.
Long non-coding RNAs (lncRNAs) are an emerging class of RNAs longer than 200 nt. Our current understanding of their functional role is limited, however there are reports describing their involvement in the regulation of gene expression, chromatin remodeling, transcription, post-transcriptional RNA processing and cancer progression[83]. Metastasis-associated lncRNAs MALAT1 (8000 nt), HOTAIR (2200 nt) and ANRIL (3800 nt) are up-regulated in some tumors and can be potentially considered as EMT-related as they regulate EMT transcription[84]. More specifically, siRNA-mediated MALAT1 silencing resulted in down-regulation of the EMT-associated transcription factors ZEB1, ZEB2 and slug, and up-regulation of E-cadherin[85]. Moreover, MALAT-1 promoted EMT by activating the Wnt signaling pathway. It also has been demonstrated that MALAT-1 levels were significantly increased in primary tumors that subsequently metastasized comparing to those tumors that did not metastasize.

Small molecule compounds modulating EMT

There are numerous kinases involved in TGF-β, Wnt, hedgehog and other signalling pathways regulating EMT and thus malignant progression. The basis of modern molecular targeted cancer therapeutics is the development of small molecule inhibitors capable of binding to the ATP-binding site of the dysregulated kinases. Thus the majority of the compounds affect EMT target kinases. For example, gefitinib and erlotinib, which are competitive inhibitors of EGFR, currently used for the treatment of advanced carcinomas, also demonstrate a protective effect against pulmonary fibrosis and hepatic fibrosis/cirrhosis, which supports their EMT-inhibiting activity[86, 87]. Other well-known compounds are antiangiogenic drugs sorafenib and sunitinib that inhibit VEGFR and PDGFR, exhibit antifibrotic effects in the liver and have been demonstrated to inhibit EMT in in vitro cell culture models[8890]. Compounds EW-7195 and EW-7203 target TGF-β type I receptor kinase/activin receptor like kinase-5 (ALK5) in a similar way, inhibiting TGF-β-induced EMT of mammary epithelial cells and preventing breast cancer metastasis to lung[91, 92].
The drug BI 5700 directly inhibits kinase IKK2, a member of NF-κB signaling pathway whose activation causes EMT, and which has been demonstrated to revert EMT in metastasizing mouse colon carcinoma[93]. Another compound SL0101 targets ribosomal protein S6 kinase (RSK)-2 which is an important component of RON and TGF-β signaling pathways[94]. Both pathways regulate EMT, and inhibition of RSK-2 results in suppression of EMT-associated cell migration in an in vitro experimental system[94].
Interestingly, the complete reversal of EMT in vitro was achieved when a combination of the inhibitors of kinases TβRI (inhibitor SB431542) and ROCK (inhibitor Y27632) was used[28]. SB431542 down-regulate ZEB1 and ZEB2 levels, thus blocking mesenchymal gene expression of TGF-β-induced mesenchymal renal tubular epithelial cells. The Rho pathway inhibiting Y27632 was necessary to fully eliminate mesenchymal actin stress fibers.
Another type of small molecule modulator 4Ei-1 is a non-toxic nucleotide analogue which prevents the association of eIF4E and the mRNA cap. It inhibited cap-dependent translation in a dose-dependent manner in zebrafish embryos without causing developmental abnormalities and prevented eIF4E from triggering EMT in zebrafish explant model[95]. This compound can be considered as a potential anti-cancer drug and investigation of its effect on the tumors would be of a great interest.
Recently a high-throughput assay was developed to screen for small molecules interfering with EMT initiated by growth factor signalling using a model carcinoma reporter cell line NBT-II[96]. In this assay both cell growth and cell migration can be analysed simultaneously via time-course imaging in multi-well plates. The authors have validated several compounds targeting ALK5, MEK, and SRC kinases as efficient EMT inhibitors. This work highlights the growing interest in the small molecule compounds able to modulate EMT. Metastases are responsible for >90% of the cancer associated deaths. Therefore new strategies to prevent EMT which leads to metastases formation might be a promising novel approach in oncology.

Conclusions

Epithelial-mesenchymal transition remains in the focus of a large number of researchers today due to its fundamental nature and important clinical implications. Non-coding RNAs and alternative splicing switches discussed in this review play important roles in EMT and cancer progression and can serve as markers for distinct epithelial or mesenchymal states of cells. Also, there are a growing number of discovered small molecules, belonging mostly to kinase inhibitors, which modulate EMT and have anti-cancer effect.

Acknowledgements

We would like to thank Dr D. Maltseva for the help with preparation of the tables and Ms E. Grundy for the carefully correcting our manuscript. This work was supported by funds from the Bundesministerium für Bildung und Forschung (BMBF No. RUS 10/022) and Russian Ministry of Science Contracts (No. 14.512.11.0024 and 14.512.11.0083).
This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors’ contribution

TRS wrote the first draft of the article, AGT and US finalized the manuscript. All authors read and approved the final manuscript.
Anhänge

Authors’ original submitted files for images

Literatur
2.
Zurück zum Zitat Chaffer CL, Thompson EW, Williams ED: Mesenchymal to epithelial transition in development and disease. Cells Tissues Organs. 2007, 185: 7-19.CrossRefPubMed Chaffer CL, Thompson EW, Williams ED: Mesenchymal to epithelial transition in development and disease. Cells Tissues Organs. 2007, 185: 7-19.CrossRefPubMed
3.
Zurück zum Zitat Zeisberg EM, Tarnavski O, Zeisberg M, Dorfman AL, McMullen JR, Gustafsson E, Chandraker A, Yuan X, Pu WT, Roberts AB, Neilson EG, Sayegh MH, Izumo S, Kalluri R: Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med. 2007, 13: 952-961.CrossRefPubMed Zeisberg EM, Tarnavski O, Zeisberg M, Dorfman AL, McMullen JR, Gustafsson E, Chandraker A, Yuan X, Pu WT, Roberts AB, Neilson EG, Sayegh MH, Izumo S, Kalluri R: Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med. 2007, 13: 952-961.CrossRefPubMed
4.
Zurück zum Zitat Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, Brumwell AN, Sheppard D, Chapman HA: Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrix. Proc Natl Acad Sci U S A. 2006, 103: 13180-13185.PubMedCentralCrossRefPubMed Kim KK, Kugler MC, Wolters PJ, Robillard L, Galvez MG, Brumwell AN, Sheppard D, Chapman HA: Alveolar epithelial cell mesenchymal transition develops in vivo during pulmonary fibrosis and is regulated by the extracellular matrix. Proc Natl Acad Sci U S A. 2006, 103: 13180-13185.PubMedCentralCrossRefPubMed
5.
Zurück zum Zitat Thiery JP: Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002, 2: 442-454.CrossRefPubMed Thiery JP: Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002, 2: 442-454.CrossRefPubMed
6.
Zurück zum Zitat Kopantzev EP, Monastyrskaya GS, Vinogradova TV, Zinovyeva MV, Kostina MB, Filyukova OB, Tonevitsky AG, Sukhikh GT, Sverdlov ED: Differences in gene expression levels between early and later stages of human lung development are opposite to those between normal lung tissue and non-small lung cell carcinoma. Lung Cancer. 2008, 62: 23-34.CrossRefPubMed Kopantzev EP, Monastyrskaya GS, Vinogradova TV, Zinovyeva MV, Kostina MB, Filyukova OB, Tonevitsky AG, Sukhikh GT, Sverdlov ED: Differences in gene expression levels between early and later stages of human lung development are opposite to those between normal lung tissue and non-small lung cell carcinoma. Lung Cancer. 2008, 62: 23-34.CrossRefPubMed
7.
Zurück zum Zitat Lee JM, Dedhar S, Kalluri R, Thompson EW: The epithelial-mesenchymal transition: new insights in signaling, development, and disease. J Cell Biol. 2006, 172: 973-981.PubMedCentralCrossRefPubMed Lee JM, Dedhar S, Kalluri R, Thompson EW: The epithelial-mesenchymal transition: new insights in signaling, development, and disease. J Cell Biol. 2006, 172: 973-981.PubMedCentralCrossRefPubMed
8.
Zurück zum Zitat Vandewalle C, Van Roy F, Berx G: The role of the ZEB family of transcription factors in development and disease. Cell Mol Life Sci. 2009, 66: 773-787.CrossRefPubMed Vandewalle C, Van Roy F, Berx G: The role of the ZEB family of transcription factors in development and disease. Cell Mol Life Sci. 2009, 66: 773-787.CrossRefPubMed
9.
Zurück zum Zitat Bindels S, Mestdagt M, Vandewalle C, Jacobs N, Volders L, Noe¨l A, Van Roy F, Berx G, Foidart JM, Gilles C: Regulation of vimentin by SIP1 in human epithelial breast tumor cells. Oncogene. 2006, 25: 4975-4985.CrossRefPubMed Bindels S, Mestdagt M, Vandewalle C, Jacobs N, Volders L, Noe¨l A, Van Roy F, Berx G, Foidart JM, Gilles C: Regulation of vimentin by SIP1 in human epithelial breast tumor cells. Oncogene. 2006, 25: 4975-4985.CrossRefPubMed
10.
Zurück zum Zitat De Craene B, Berx G: Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer. 2013, 13: 97-110.CrossRefPubMed De Craene B, Berx G: Regulatory networks defining EMT during cancer initiation and progression. Nat Rev Cancer. 2013, 13: 97-110.CrossRefPubMed
11.
Zurück zum Zitat Sánchez-Tilló E, Liu Y, de Barrios O, Siles L, Fanlo L, Cuatrecasas M, Darling DS, Dean DC, Castells A, Postigo A: EMT-activating transcription factors in cancer: beyond EMT and tumor invasiveness. Cell Mol Life Sci. 2012, 69: 3429-3456.CrossRefPubMed Sánchez-Tilló E, Liu Y, de Barrios O, Siles L, Fanlo L, Cuatrecasas M, Darling DS, Dean DC, Castells A, Postigo A: EMT-activating transcription factors in cancer: beyond EMT and tumor invasiveness. Cell Mol Life Sci. 2012, 69: 3429-3456.CrossRefPubMed
12.
Zurück zum Zitat Peinado H, Marin F, Cubillo E, Stark HJ, Fusenig N, Nieto MA, Cano A: Snail and E47 repressors of E-cadherin induce distinct invasive and angiogenic properties in vivo. J Cell Sci. 2004, 117: 2827-2839.CrossRefPubMed Peinado H, Marin F, Cubillo E, Stark HJ, Fusenig N, Nieto MA, Cano A: Snail and E47 repressors of E-cadherin induce distinct invasive and angiogenic properties in vivo. J Cell Sci. 2004, 117: 2827-2839.CrossRefPubMed
13.
Zurück zum Zitat Thiery JP, Acloque H, Huang RY, Nieto MA: Epithelial mesenchymal transitions in development and disease. Cell. 2009, 139: 871-890.CrossRefPubMed Thiery JP, Acloque H, Huang RY, Nieto MA: Epithelial mesenchymal transitions in development and disease. Cell. 2009, 139: 871-890.CrossRefPubMed
14.
Zurück zum Zitat Qin Q, Xu Y, He T, Qin C, Xu J: Normal and disease related biological functions of Twist1 and underlying molecular mechanisms. Cell Res. 2012, 22: 90-106.PubMedCentralCrossRefPubMed Qin Q, Xu Y, He T, Qin C, Xu J: Normal and disease related biological functions of Twist1 and underlying molecular mechanisms. Cell Res. 2012, 22: 90-106.PubMedCentralCrossRefPubMed
15.
Zurück zum Zitat Dave N, Guaita-Esteruelas S, Gutarra S, Frias A, Beltran M, Peiro S, de Herreros AG: Functional cooperation between Snail1 and twist in the regulation of ZEB1 expression during epithelial to mesenchymal transition. J Biol Chem. 2011, 286: 12024-12032.PubMedCentralCrossRefPubMed Dave N, Guaita-Esteruelas S, Gutarra S, Frias A, Beltran M, Peiro S, de Herreros AG: Functional cooperation between Snail1 and twist in the regulation of ZEB1 expression during epithelial to mesenchymal transition. J Biol Chem. 2011, 286: 12024-12032.PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Beltran M, Puig I, Peña C, Garcia JM, Alvarez AB, Peña R, Bonilla F, de Herreros AG: A natural antisense transcript regulates Zeb2/Sip1 gene expression during Snail1-induced epithelial-mesenchymal transition. Genes Dev. 2008, 22: 756-769.PubMedCentralCrossRefPubMed Beltran M, Puig I, Peña C, Garcia JM, Alvarez AB, Peña R, Bonilla F, de Herreros AG: A natural antisense transcript regulates Zeb2/Sip1 gene expression during Snail1-induced epithelial-mesenchymal transition. Genes Dev. 2008, 22: 756-769.PubMedCentralCrossRefPubMed
17.
Zurück zum Zitat Casas E, Kim J, Bendesky A, Ohno-Machado L, Wolfe CJ, Yang J: Snail2 is an essential mediator of Twist1-induced epithelial mesenchymal transition and metastasis. Cancer Res. 2011, 71: 245-254.PubMedCentralCrossRefPubMed Casas E, Kim J, Bendesky A, Ohno-Machado L, Wolfe CJ, Yang J: Snail2 is an essential mediator of Twist1-induced epithelial mesenchymal transition and metastasis. Cancer Res. 2011, 71: 245-254.PubMedCentralCrossRefPubMed
18.
Zurück zum Zitat Ribatti D, Mangialardi G, Vacca A: Stephen Paget and the ‘seed and soil’ theory of metastatic dissemination. Clin Exp Med. 2006, 6: 145-149.CrossRefPubMed Ribatti D, Mangialardi G, Vacca A: Stephen Paget and the ‘seed and soil’ theory of metastatic dissemination. Clin Exp Med. 2006, 6: 145-149.CrossRefPubMed
19.
20.
Zurück zum Zitat Frisch SM, Francis H: Disruption of epithelial cell-matrix interactions induces apoptosis. J Cell Biol. 1994, 124: 619-626.CrossRefPubMed Frisch SM, Francis H: Disruption of epithelial cell-matrix interactions induces apoptosis. J Cell Biol. 1994, 124: 619-626.CrossRefPubMed
21.
Zurück zum Zitat Weinberg R: The Biology of Cancer. 2006, New York: Garland Science Weinberg R: The Biology of Cancer. 2006, New York: Garland Science
22.
Zurück zum Zitat Hirohashi S, Kanai Y: Cell adhesion system and human cancer morphogenesis. Cancer Sci. 2003, 94: 575-581.CrossRefPubMed Hirohashi S, Kanai Y: Cell adhesion system and human cancer morphogenesis. Cancer Sci. 2003, 94: 575-581.CrossRefPubMed
23.
Zurück zum Zitat Zhao JH, Luo Y, Jiang YG, He DL, Wu CT: Knockdown of β-Catenin through shRNA cause a reversal of EMT and metastatic phenotypes induced by HIF-1α. Cancer Invest. 2011, 29: 377-382.CrossRefPubMed Zhao JH, Luo Y, Jiang YG, He DL, Wu CT: Knockdown of β-Catenin through shRNA cause a reversal of EMT and metastatic phenotypes induced by HIF-1α. Cancer Invest. 2011, 29: 377-382.CrossRefPubMed
24.
Zurück zum Zitat Yi ZY, Feng LJ, Xiang Z, Yao H: Vascular endothelial growth factor receptor-1 activation mediates epithelial to mesenchymal transition in hepatocellular carcinoma cells. J Invest Surg. 2011, 24: 67-76.CrossRefPubMed Yi ZY, Feng LJ, Xiang Z, Yao H: Vascular endothelial growth factor receptor-1 activation mediates epithelial to mesenchymal transition in hepatocellular carcinoma cells. J Invest Surg. 2011, 24: 67-76.CrossRefPubMed
25.
Zurück zum Zitat Cordenonsi M, D’Atri F, Hammar E, Parry DA, Kendrick-Jones J, Shore D, Citi S: Cingulin contains globular and coiled-coil domains and interacts with ZO-1, ZO-2, ZO-3, and myosin. J Cell Biol. 1999, 147: 1569-1582.PubMedCentralCrossRefPubMed Cordenonsi M, D’Atri F, Hammar E, Parry DA, Kendrick-Jones J, Shore D, Citi S: Cingulin contains globular and coiled-coil domains and interacts with ZO-1, ZO-2, ZO-3, and myosin. J Cell Biol. 1999, 147: 1569-1582.PubMedCentralCrossRefPubMed
26.
27.
Zurück zum Zitat Ley K, Laudanna C, Cybulsky MI, Nourshargh S: Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol. 2007, 7: 678-689.CrossRefPubMed Ley K, Laudanna C, Cybulsky MI, Nourshargh S: Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol. 2007, 7: 678-689.CrossRefPubMed
28.
Zurück zum Zitat Das S, Becker BN, Hoffmann FM, Mertz JE: Complete reversal of epithelial to mesenchymal transition requires inhibition of both ZEB expression and the Rho pathway. BMC Cell Biol. 2009, 10: 94-PubMedCentralCrossRefPubMed Das S, Becker BN, Hoffmann FM, Mertz JE: Complete reversal of epithelial to mesenchymal transition requires inhibition of both ZEB expression and the Rho pathway. BMC Cell Biol. 2009, 10: 94-PubMedCentralCrossRefPubMed
30.
Zurück zum Zitat Williams CM, Engler AJ, Slone RD, Galante LL, Schwarzbauer JE: Fibronectin expression modulates mammary epithelial cell proliferation during acinar differentiation. Cancer Res. 2008, 68: 3185-3192.PubMedCentralCrossRefPubMed Williams CM, Engler AJ, Slone RD, Galante LL, Schwarzbauer JE: Fibronectin expression modulates mammary epithelial cell proliferation during acinar differentiation. Cancer Res. 2008, 68: 3185-3192.PubMedCentralCrossRefPubMed
31.
Zurück zum Zitat Hidalgo A, Peired AJ, Wild MK, Vestweber D, Frenette PS: Complete identification of E-selectin ligands on neutrophils reveals distinct functions of PSGL-1, ESL-1, and CD44. Immunity. 2007, 26: 477-89.PubMedCentralCrossRefPubMed Hidalgo A, Peired AJ, Wild MK, Vestweber D, Frenette PS: Complete identification of E-selectin ligands on neutrophils reveals distinct functions of PSGL-1, ESL-1, and CD44. Immunity. 2007, 26: 477-89.PubMedCentralCrossRefPubMed
32.
Zurück zum Zitat Zen K, Liu DQ, Guo YL, Wang C, Shan J, Fang M, Zhang CY, Liu Y: CD44v4 is a major E-selectin ligand that mediates breast cancer cell transendothelial migration. PLoS One. 2008, 3: e1826-PubMedCentralCrossRefPubMed Zen K, Liu DQ, Guo YL, Wang C, Shan J, Fang M, Zhang CY, Liu Y: CD44v4 is a major E-selectin ligand that mediates breast cancer cell transendothelial migration. PLoS One. 2008, 3: e1826-PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Zhang L, Riethdorf S, Wu G, Wang T, Yang K, Peng G, Liu J, Pantel K: Meta-analysis of the prognostic value of circulating tumor cells in breast cancer. Clin Cancer Res. 2012, 18: 5701-5710.CrossRefPubMed Zhang L, Riethdorf S, Wu G, Wang T, Yang K, Peng G, Liu J, Pantel K: Meta-analysis of the prognostic value of circulating tumor cells in breast cancer. Clin Cancer Res. 2012, 18: 5701-5710.CrossRefPubMed
34.
Zurück zum Zitat Delacruz A: Using circulating tumor cells as a prognostic indicator in metastatic castration-resistant prostate cancer. Clin J Oncol Nurs. 2012, 16: E44-47.CrossRefPubMed Delacruz A: Using circulating tumor cells as a prognostic indicator in metastatic castration-resistant prostate cancer. Clin J Oncol Nurs. 2012, 16: E44-47.CrossRefPubMed
35.
Zurück zum Zitat O’Flaherty JD, Gray S, Richard D, Fennell D, O’Leary JJ, Blackhall FH, O’Byrne KJ: Circulating tumour cells, their role in metastasis and their clinical utility in lung cancer. Lung Cancer. 2012, 76: 19-25.CrossRefPubMed O’Flaherty JD, Gray S, Richard D, Fennell D, O’Leary JJ, Blackhall FH, O’Byrne KJ: Circulating tumour cells, their role in metastasis and their clinical utility in lung cancer. Lung Cancer. 2012, 76: 19-25.CrossRefPubMed
36.
Zurück zum Zitat Msaouel P, Koutsilieris M: Diagnostic value of circulating tumor cell detection in bladder and urothelial cancer: systematicreview and meta-analysis. BMC Cancer. 2011, 11: 336-PubMedCentralCrossRefPubMed Msaouel P, Koutsilieris M: Diagnostic value of circulating tumor cell detection in bladder and urothelial cancer: systematicreview and meta-analysis. BMC Cancer. 2011, 11: 336-PubMedCentralCrossRefPubMed
37.
Zurück zum Zitat Hashimoto M, Tanaka F, Yoneda K, Kondo N, Takuwa T, Matsumoto S, Kuroda A, Noda M, Tomita N, Hasegawa S: Circulating tumor cells as a potential biomarker in selecting patients for pulmonary metastasectomy from colorectal cancer: report of a case. Case Rep Oncol. 2012, 5: 542-545.PubMedCentralCrossRefPubMed Hashimoto M, Tanaka F, Yoneda K, Kondo N, Takuwa T, Matsumoto S, Kuroda A, Noda M, Tomita N, Hasegawa S: Circulating tumor cells as a potential biomarker in selecting patients for pulmonary metastasectomy from colorectal cancer: report of a case. Case Rep Oncol. 2012, 5: 542-545.PubMedCentralCrossRefPubMed
38.
Zurück zum Zitat Kallergi G, Papadaki MA, Politaki E, Mavroudis D, Georgoulias V, Agelaki S: Epithelial to mesenchymal transition markers expressed in circulating tumour cells of early and metastatic breast cancer patients. Breast Cancer Res. 2011, 13: R59-PubMedCentralCrossRefPubMed Kallergi G, Papadaki MA, Politaki E, Mavroudis D, Georgoulias V, Agelaki S: Epithelial to mesenchymal transition markers expressed in circulating tumour cells of early and metastatic breast cancer patients. Breast Cancer Res. 2011, 13: R59-PubMedCentralCrossRefPubMed
39.
Zurück zum Zitat Gradilone A, Raimondi C, Nicolazzo C, Petracca A, Gandini O, Vincenzi B, Naso G, Aglianò AM, Cortesi E, Gazzaniga P: Circulating tumour cells lacking cytokeratin in breast cancer: the importance of being mesenchymal. J Cell Mol Med. 2011, 15: 1066-1070.PubMedCentralCrossRefPubMed Gradilone A, Raimondi C, Nicolazzo C, Petracca A, Gandini O, Vincenzi B, Naso G, Aglianò AM, Cortesi E, Gazzaniga P: Circulating tumour cells lacking cytokeratin in breast cancer: the importance of being mesenchymal. J Cell Mol Med. 2011, 15: 1066-1070.PubMedCentralCrossRefPubMed
40.
Zurück zum Zitat Yu M, Bardia A, Wittner BS, Stott SL, Smas ME, Ting DT, Isakoff SJ, Ciciliano JC, Wells MN, Shah AM, Concannon KF, Donaldson MC, Sequist LV, Brachtel E, Sgroi D, Baselga J, Ramaswamy S, Toner M, Haber DA, Maheswaran S: Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science. 2013, 339: 580-584.PubMedCentralCrossRefPubMed Yu M, Bardia A, Wittner BS, Stott SL, Smas ME, Ting DT, Isakoff SJ, Ciciliano JC, Wells MN, Shah AM, Concannon KF, Donaldson MC, Sequist LV, Brachtel E, Sgroi D, Baselga J, Ramaswamy S, Toner M, Haber DA, Maheswaran S: Circulating breast tumor cells exhibit dynamic changes in epithelial and mesenchymal composition. Science. 2013, 339: 580-584.PubMedCentralCrossRefPubMed
41.
Zurück zum Zitat Kasimir-Bauer S, Hoffmann O, Wallwiener D, Kimmig R, Fehm T: Expression of stem cell and epithelial-mesenchymal transition markers in primary breast cancer patients with circulating tumor cells. Breast Cancer Res. 2012, 14: R15-PubMedCentralCrossRefPubMed Kasimir-Bauer S, Hoffmann O, Wallwiener D, Kimmig R, Fehm T: Expression of stem cell and epithelial-mesenchymal transition markers in primary breast cancer patients with circulating tumor cells. Breast Cancer Res. 2012, 14: R15-PubMedCentralCrossRefPubMed
42.
Zurück zum Zitat Kampa D, Cheng J, Kapranov P, Yamanaka M, Brubaker S, Cawley S, Drenkow J, Piccolboni A, Bekiranov S, Helt G, Tammana H, Gingeras TR: Novel RNAs identified from an in-depth analysis of the transcriptome of human chromosomes 21 and 22. Genome Res. 2004, 14: 331-342.PubMedCentralCrossRefPubMed Kampa D, Cheng J, Kapranov P, Yamanaka M, Brubaker S, Cawley S, Drenkow J, Piccolboni A, Bekiranov S, Helt G, Tammana H, Gingeras TR: Novel RNAs identified from an in-depth analysis of the transcriptome of human chromosomes 21 and 22. Genome Res. 2004, 14: 331-342.PubMedCentralCrossRefPubMed
44.
Zurück zum Zitat Chen M, Manley JL: Mechanisms of alternative splicing regulation: insights from molecular and genomics approaches. Nat Rev Mol Cell Biol. 2009, 10: 741-754.PubMedCentralPubMed Chen M, Manley JL: Mechanisms of alternative splicing regulation: insights from molecular and genomics approaches. Nat Rev Mol Cell Biol. 2009, 10: 741-754.PubMedCentralPubMed
45.
Zurück zum Zitat Savagner P, Vallés AM, Jouanneau J, Yamada KM, Thiery JP: Alternative splicing in fibroblast growth factor receptor 2 is associated with induced epithelial-mesenchymal transition in rat bladder carcinoma cells. Mol Biol Cell. 1994, 5: 851-862.PubMedCentralCrossRefPubMed Savagner P, Vallés AM, Jouanneau J, Yamada KM, Thiery JP: Alternative splicing in fibroblast growth factor receptor 2 is associated with induced epithelial-mesenchymal transition in rat bladder carcinoma cells. Mol Biol Cell. 1994, 5: 851-862.PubMedCentralCrossRefPubMed
46.
Zurück zum Zitat Arman E, Haffner-Krausz R, Gorivodsky M, Lonai P: Fgfr2 is required for limb outgrowth and lung-branching morphogenesis. Proc Natl Acad Sci U S A. 1999, 96: 11895-11899.PubMedCentralCrossRefPubMed Arman E, Haffner-Krausz R, Gorivodsky M, Lonai P: Fgfr2 is required for limb outgrowth and lung-branching morphogenesis. Proc Natl Acad Sci U S A. 1999, 96: 11895-11899.PubMedCentralCrossRefPubMed
47.
Zurück zum Zitat Chaffer CL, Brennan JP, Slavin JL, Blick T, Thompson EW, Williams ED: Mesenchymal-to-epithelial transition facilitates bladder cancer metastasis: role of fibroblast growth factor receptor-2. Cancer Res. 2006, 66: 11271-11278.CrossRefPubMed Chaffer CL, Brennan JP, Slavin JL, Blick T, Thompson EW, Williams ED: Mesenchymal-to-epithelial transition facilitates bladder cancer metastasis: role of fibroblast growth factor receptor-2. Cancer Res. 2006, 66: 11271-11278.CrossRefPubMed
48.
Zurück zum Zitat Scott MS, Ono M, Yamada K, Endo A, Barton GJ, Lamond AI: Human box C/D snoRNA processing conservation across multiple cell types. Nucleic Acids Res. 2012, 40: 3676-3688.PubMedCentralCrossRefPubMed Scott MS, Ono M, Yamada K, Endo A, Barton GJ, Lamond AI: Human box C/D snoRNA processing conservation across multiple cell types. Nucleic Acids Res. 2012, 40: 3676-3688.PubMedCentralCrossRefPubMed
49.
Zurück zum Zitat Zöller M: CD44: can a cancer-initiating cell profit from an abundantly expressed molecule?. Nat Rev Cancer. 2011, 11: 254-267.CrossRefPubMed Zöller M: CD44: can a cancer-initiating cell profit from an abundantly expressed molecule?. Nat Rev Cancer. 2011, 11: 254-267.CrossRefPubMed
50.
Zurück zum Zitat Bennett KL, Modrell B, Greenfield B, Bartolazzi A, Stamenkovic I, Peach R, Jackson DG, Spring F, Aruffo A: Regulation of CD44 binding to hyaluronan by glycosylation of variably spliced exons. J Cell Biol. 1995, 131: 1623-1633.CrossRefPubMed Bennett KL, Modrell B, Greenfield B, Bartolazzi A, Stamenkovic I, Peach R, Jackson DG, Spring F, Aruffo A: Regulation of CD44 binding to hyaluronan by glycosylation of variably spliced exons. J Cell Biol. 1995, 131: 1623-1633.CrossRefPubMed
51.
Zurück zum Zitat Hong RL, Pu YS, Chu JS, Lee WJ, Chen YC, Wu CW: Correlation of expression of CD44 isoforms and E-cadherin with differentiation in human urothelial cell lines and transitional cell carcinoma. Cancer Lett. 1995, 89: 81-87.CrossRefPubMed Hong RL, Pu YS, Chu JS, Lee WJ, Chen YC, Wu CW: Correlation of expression of CD44 isoforms and E-cadherin with differentiation in human urothelial cell lines and transitional cell carcinoma. Cancer Lett. 1995, 89: 81-87.CrossRefPubMed
52.
Zurück zum Zitat Brown RL, Reinke LM, Damerow MS, Perez D, Chodosh LA, Yang J, Cheng C: CD44 splice isoform switching in human and mouse epithelium is essential for epithelial-mesenchymal transition and breast cancer progression. J Clin Invest. 2011, 121: 1064-1074.PubMedCentralCrossRefPubMed Brown RL, Reinke LM, Damerow MS, Perez D, Chodosh LA, Yang J, Cheng C: CD44 splice isoform switching in human and mouse epithelium is essential for epithelial-mesenchymal transition and breast cancer progression. J Clin Invest. 2011, 121: 1064-1074.PubMedCentralCrossRefPubMed
53.
Zurück zum Zitat Schmucker D, Chen B: Dscam and DSCAM: complex genes in simple animals, complex animals yet simple genes. Genes Dev. 2009, 23: 147-156.CrossRefPubMed Schmucker D, Chen B: Dscam and DSCAM: complex genes in simple animals, complex animals yet simple genes. Genes Dev. 2009, 23: 147-156.CrossRefPubMed
55.
Zurück zum Zitat Grosheva I, Shtutman M, Elbaum M, Bershadsky AD: p120 catenin affects cell motility via modulation of activity of Rho-family GTPases: a link between cell-cell contact formation and regulation of cell locomotion. J Cell Sci. 2001, 114: 695-707.PubMed Grosheva I, Shtutman M, Elbaum M, Bershadsky AD: p120 catenin affects cell motility via modulation of activity of Rho-family GTPases: a link between cell-cell contact formation and regulation of cell locomotion. J Cell Sci. 2001, 114: 695-707.PubMed
56.
Zurück zum Zitat Ohkubo T, Ozawa M: The transcription factor Snail downregulates the tight junction components independently of E-cadherin downregulation. J Cell Sci. 2004, 117: 1675-1685.CrossRefPubMed Ohkubo T, Ozawa M: The transcription factor Snail downregulates the tight junction components independently of E-cadherin downregulation. J Cell Sci. 2004, 117: 1675-1685.CrossRefPubMed
57.
Zurück zum Zitat Menke A, Giehl K: Regulation of adherens junctions by Rho GTPases and p120-catenin. Arch Biochem Biophys. 2012, 524: 48-55.CrossRefPubMed Menke A, Giehl K: Regulation of adherens junctions by Rho GTPases and p120-catenin. Arch Biochem Biophys. 2012, 524: 48-55.CrossRefPubMed
58.
Zurück zum Zitat Yanagisawa M, Huveldt D, Kreinest P, Lohse CM, Cheville JC, Parker AS, Copland JA, Anastasiadis PZ: A p120 catenin isoform switch affects Rho activity, induces tumor cell invasion, and predicts metastatic disease. J Biol Chem. 2008, 283: 18344-18354.PubMedCentralCrossRefPubMed Yanagisawa M, Huveldt D, Kreinest P, Lohse CM, Cheville JC, Parker AS, Copland JA, Anastasiadis PZ: A p120 catenin isoform switch affects Rho activity, induces tumor cell invasion, and predicts metastatic disease. J Biol Chem. 2008, 283: 18344-18354.PubMedCentralCrossRefPubMed
59.
Zurück zum Zitat Krause M, Dent EW, Bear JE, Loureiro JJ, Gertler FB: Ena/VASP proteins: regulators of the actin cytoskeleton and cell migration. Annu Rev Cell Dev Biol. 2003, 19: 541-564.CrossRefPubMed Krause M, Dent EW, Bear JE, Loureiro JJ, Gertler FB: Ena/VASP proteins: regulators of the actin cytoskeleton and cell migration. Annu Rev Cell Dev Biol. 2003, 19: 541-564.CrossRefPubMed
60.
Zurück zum Zitat Goswami S, Philippar U, Sun D, Patsialou A, Avraham J, Wang W, Di Modugno F, Nistico P, Gertler FB, Condeelis JS: Identification of invasion specific splice variants of the cytoskeletal protein Mena present in mammary tumor cells during invasion in vivo. Clin Exp Metastasis. 2009, 26: 153-159.PubMedCentralCrossRefPubMed Goswami S, Philippar U, Sun D, Patsialou A, Avraham J, Wang W, Di Modugno F, Nistico P, Gertler FB, Condeelis JS: Identification of invasion specific splice variants of the cytoskeletal protein Mena present in mammary tumor cells during invasion in vivo. Clin Exp Metastasis. 2009, 26: 153-159.PubMedCentralCrossRefPubMed
61.
Zurück zum Zitat Shapiro IM, Cheng AW, Flytzanis NC, Balsamo M, Condeelis JS, Oktay MH, Burge CB, Gertler FB: An EMT-driven alternative splicing program occurs in human breast cancer and modulates cellular phenotype. PLoS Genet. 2011, 7: e1002218-PubMedCentralCrossRefPubMed Shapiro IM, Cheng AW, Flytzanis NC, Balsamo M, Condeelis JS, Oktay MH, Burge CB, Gertler FB: An EMT-driven alternative splicing program occurs in human breast cancer and modulates cellular phenotype. PLoS Genet. 2011, 7: e1002218-PubMedCentralCrossRefPubMed
62.
Zurück zum Zitat Warzecha CC, Shen S, Xing Y, Carstens RP: The epithelial splicing factors ESRP1 and ESRP2 positively and negatively regulate diverse types of alternative splicing events. RNA Biol. 2009, 6: 546-562.PubMedCentralCrossRefPubMed Warzecha CC, Shen S, Xing Y, Carstens RP: The epithelial splicing factors ESRP1 and ESRP2 positively and negatively regulate diverse types of alternative splicing events. RNA Biol. 2009, 6: 546-562.PubMedCentralCrossRefPubMed
63.
Zurück zum Zitat Braun JE, Huntzinger E, Izaurralde E: A molecular link between miRISCs and deadenylases provides new insight into the mechanism of gene silencing by microRNAs. Cold Spring Harb Perspect Biol. 2012, 4: 12-CrossRef Braun JE, Huntzinger E, Izaurralde E: A molecular link between miRISCs and deadenylases provides new insight into the mechanism of gene silencing by microRNAs. Cold Spring Harb Perspect Biol. 2012, 4: 12-CrossRef
64.
Zurück zum Zitat Loeb GB, Khan AA, Canner D, Hiatt JB, Shendure J, Darnell RB, Leslie CS, Rudensky AY: Transcriptome-wide miR-155 binding map reveals widespread noncanonical microRNA targeting. Mol Cell. 2012, 48: 760-770.PubMedCentralCrossRefPubMed Loeb GB, Khan AA, Canner D, Hiatt JB, Shendure J, Darnell RB, Leslie CS, Rudensky AY: Transcriptome-wide miR-155 binding map reveals widespread noncanonical microRNA targeting. Mol Cell. 2012, 48: 760-770.PubMedCentralCrossRefPubMed
65.
Zurück zum Zitat Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 2004, 116: 281-297.CrossRefPubMed Bartel DP: MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell. 2004, 116: 281-297.CrossRefPubMed
66.
Zurück zum Zitat Wright JA, Richer JK, Goodall GJ: microRNAs and EMT in mammary cells and breast cancer. J Mammary Gland Biol Neoplasia. 2010, 15: 213-223.CrossRefPubMed Wright JA, Richer JK, Goodall GJ: microRNAs and EMT in mammary cells and breast cancer. J Mammary Gland Biol Neoplasia. 2010, 15: 213-223.CrossRefPubMed
67.
Zurück zum Zitat Tonevitsky AG, Maltseva DV, Abbasi A, Samatov TR, Sakharov DA, Shkurnikov MU, Lebedev AE, Galatenko VV, Grigoriev AI, Northoff H: Dynamically regulated miRNA-mRNA networks revealed by exercise. BMC Physiol. 2013, 13: 9-10.1186/1472-6793-13-9.PubMedCentralCrossRefPubMed Tonevitsky AG, Maltseva DV, Abbasi A, Samatov TR, Sakharov DA, Shkurnikov MU, Lebedev AE, Galatenko VV, Grigoriev AI, Northoff H: Dynamically regulated miRNA-mRNA networks revealed by exercise. BMC Physiol. 2013, 13: 9-10.1186/1472-6793-13-9.PubMedCentralCrossRefPubMed
68.
Zurück zum Zitat Turchinovich A, Samatov TR, Tonevitsky AG, Burwinkel B: Circulating miRNAs: cell-cell communication function?. Front Genet. 2013, 4: 119-doi:10.3389/fgene.2013.00119PubMedCentralCrossRefPubMed Turchinovich A, Samatov TR, Tonevitsky AG, Burwinkel B: Circulating miRNAs: cell-cell communication function?. Front Genet. 2013, 4: 119-doi:10.3389/fgene.2013.00119PubMedCentralCrossRefPubMed
69.
Zurück zum Zitat Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, Vadas MA, Khew-Goodall Y, Goodall GJ: The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008, 10: 593-601.CrossRefPubMed Gregory PA, Bert AG, Paterson EL, Barry SC, Tsykin A, Farshid G, Vadas MA, Khew-Goodall Y, Goodall GJ: The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting ZEB1 and SIP1. Nat Cell Biol. 2008, 10: 593-601.CrossRefPubMed
70.
Zurück zum Zitat Park S-M, Gaur AB, Lengyel E, Peter ME: The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008, 22: 894-907.PubMedCentralCrossRefPubMed Park S-M, Gaur AB, Lengyel E, Peter ME: The miR-200 family determines the epithelial phenotype of cancer cells by targeting the E-cadherin repressors ZEB1 and ZEB2. Genes Dev. 2008, 22: 894-907.PubMedCentralCrossRefPubMed
72.
Zurück zum Zitat Howe EN, Cochrane DR, Richer JK: Targets of miR-200c mediate suppression of cell motility and anoikis resistance. Breast Cancer Res. 2011, 13: R45-PubMedCentralCrossRefPubMed Howe EN, Cochrane DR, Richer JK: Targets of miR-200c mediate suppression of cell motility and anoikis resistance. Breast Cancer Res. 2011, 13: R45-PubMedCentralCrossRefPubMed
73.
Zurück zum Zitat Howe EN, Cochrane DR, Cittelly DM, Richer JK: miR-200c targets a NF-κB up-regulated TrkB/NTF3 autocrine signaling loop to enhance anoikis sensitivity in triple negative breast cancer. PLoS One. 2012, 7: e49987-PubMedCentralCrossRefPubMed Howe EN, Cochrane DR, Cittelly DM, Richer JK: miR-200c targets a NF-κB up-regulated TrkB/NTF3 autocrine signaling loop to enhance anoikis sensitivity in triple negative breast cancer. PLoS One. 2012, 7: e49987-PubMedCentralCrossRefPubMed
74.
Zurück zum Zitat Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, Spaderna S, Brabletz T: A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep. 2008, 9: 582-589.PubMedCentralCrossRefPubMed Burk U, Schubert J, Wellner U, Schmalhofer O, Vincan E, Spaderna S, Brabletz T: A reciprocal repression between ZEB1 and members of the miR-200 family promotes EMT and invasion in cancer cells. EMBO Rep. 2008, 9: 582-589.PubMedCentralCrossRefPubMed
75.
Zurück zum Zitat Xia H, Ng SS, Jiang S, Cheung WK, Sze J, Bian XW, Kung HF, Lin MC: miR-200a-mediated downregulation of ZEB2 and CTNNB1 differentially inhibits nasopharyngeal carcinoma cell growth, migration and invasion. Biochem Biophys Res Commun. 2010, 391: 535-541.CrossRefPubMed Xia H, Ng SS, Jiang S, Cheung WK, Sze J, Bian XW, Kung HF, Lin MC: miR-200a-mediated downregulation of ZEB2 and CTNNB1 differentially inhibits nasopharyngeal carcinoma cell growth, migration and invasion. Biochem Biophys Res Commun. 2010, 391: 535-541.CrossRefPubMed
76.
Zurück zum Zitat Du J, Yang S, An D, Hu F, Yuan W, Zhai C, Zhu T: BMP-6 inhibits microRNA-21 expression in breast cancer through repressing deltaEF1 and AP-1. Cell Res. 2009, 19: 487-496.CrossRefPubMed Du J, Yang S, An D, Hu F, Yuan W, Zhai C, Zhu T: BMP-6 inhibits microRNA-21 expression in breast cancer through repressing deltaEF1 and AP-1. Cell Res. 2009, 19: 487-496.CrossRefPubMed
77.
Zurück zum Zitat Ma L, Teruya-Feldstein J, Weinberg RA: Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature. 2007, 449: 682-688.CrossRefPubMed Ma L, Teruya-Feldstein J, Weinberg RA: Tumour invasion and metastasis initiated by microRNA-10b in breast cancer. Nature. 2007, 449: 682-688.CrossRefPubMed
78.
Zurück zum Zitat Kong W, Yang H, He L, Zhao JJ, Coppola D, Dalton WS, Cheng JQ: MicroRNA-155 is regulated by the transforming growth factor beta/Smad pathway and contributes to epithelial cell plasticity by targeting RhoA. Mol Cell Biol. 2008, 28: 6773-6784.PubMedCentralCrossRefPubMed Kong W, Yang H, He L, Zhao JJ, Coppola D, Dalton WS, Cheng JQ: MicroRNA-155 is regulated by the transforming growth factor beta/Smad pathway and contributes to epithelial cell plasticity by targeting RhoA. Mol Cell Biol. 2008, 28: 6773-6784.PubMedCentralCrossRefPubMed
80.
Zurück zum Zitat Gebeshuber CA, Zatloukal K, Martinez J: miR-29a suppresses tristetraprolin, which is a regulator of epithelial polarity and metastasis. EMBO Rep. 2009, 10: 400-405.PubMedCentralCrossRefPubMed Gebeshuber CA, Zatloukal K, Martinez J: miR-29a suppresses tristetraprolin, which is a regulator of epithelial polarity and metastasis. EMBO Rep. 2009, 10: 400-405.PubMedCentralCrossRefPubMed
81.
Zurück zum Zitat Ma L, Young J, Prabhala H, Pan E, Mestdagh P, Muth D, Teruya-Feldstein J, Reinhardt F, Onder TT, Valastyan S, Westermann F, Speleman F, Vandesompele J, Weinberg RA: miR-9, a MYC/MYCN-activated microRNA, regulates E-cadherin and cancer metastasis. Nat Cell Biol. 2010, 12: 247-256.PubMedCentralPubMed Ma L, Young J, Prabhala H, Pan E, Mestdagh P, Muth D, Teruya-Feldstein J, Reinhardt F, Onder TT, Valastyan S, Westermann F, Speleman F, Vandesompele J, Weinberg RA: miR-9, a MYC/MYCN-activated microRNA, regulates E-cadherin and cancer metastasis. Nat Cell Biol. 2010, 12: 247-256.PubMedCentralPubMed
82.
Zurück zum Zitat Madhavan D, Zucknick M, Wallwiener M, Cuk K, Modugno C, Scharpff M, Schott S, Heil J, Turchinovich A, Yang R, Benner A, Riethdorf S, Trumpp A, Sohn C, Pantel K, Schneeweiss A, Burwinkel B: Circulating miRNAs as surrogate markers for circulating tumor cells and prognostic markers in metastatic breast cancer. Clin Cancer Res. 2012, 18: 5972-5982.CrossRefPubMed Madhavan D, Zucknick M, Wallwiener M, Cuk K, Modugno C, Scharpff M, Schott S, Heil J, Turchinovich A, Yang R, Benner A, Riethdorf S, Trumpp A, Sohn C, Pantel K, Schneeweiss A, Burwinkel B: Circulating miRNAs as surrogate markers for circulating tumor cells and prognostic markers in metastatic breast cancer. Clin Cancer Res. 2012, 18: 5972-5982.CrossRefPubMed
85.
Zurück zum Zitat Ying L, Chen Q, Wang Y, Zhou Z, Huang Y, Qiu F: Upregulated MALAT-1 contributes to bladder cancer cell migration by inducing epithelial-to-mesenchymal transition. Mol Biosyst. 2012, 8: 2289-2294.CrossRefPubMed Ying L, Chen Q, Wang Y, Zhou Z, Huang Y, Qiu F: Upregulated MALAT-1 contributes to bladder cancer cell migration by inducing epithelial-to-mesenchymal transition. Mol Biosyst. 2012, 8: 2289-2294.CrossRefPubMed
86.
Zurück zum Zitat Ishii Y, Fujimoto S, Fukuda T: Gefitinib prevents bleomycin-induced lung fibrosis in mice. Am J Respir Crit Care Med. 2006, 174: 550-556.CrossRefPubMed Ishii Y, Fujimoto S, Fukuda T: Gefitinib prevents bleomycin-induced lung fibrosis in mice. Am J Respir Crit Care Med. 2006, 174: 550-556.CrossRefPubMed
87.
Zurück zum Zitat Kim Y, Mio T, Mishima M: Gefitinib for non-small cell lung cancer patients with liver cirrhosis. Intern Med. 2009, 48: 1677-1679.CrossRefPubMed Kim Y, Mio T, Mishima M: Gefitinib for non-small cell lung cancer patients with liver cirrhosis. Intern Med. 2009, 48: 1677-1679.CrossRefPubMed
88.
Zurück zum Zitat Mejias M, Garcia-Pras E, Tiani C, Miquel R, Bosch J, Fernandez M: Beneficial effects of sorafenib on splanchnic, intrahepatic, and portocollateral circulations in portal hypertensive and cirrhotic rats. Hepatology. 2009, 49: 1245-1256.CrossRefPubMed Mejias M, Garcia-Pras E, Tiani C, Miquel R, Bosch J, Fernandez M: Beneficial effects of sorafenib on splanchnic, intrahepatic, and portocollateral circulations in portal hypertensive and cirrhotic rats. Hepatology. 2009, 49: 1245-1256.CrossRefPubMed
89.
Zurück zum Zitat Tugues S, Fernandez-Varo G, Muñoz-Luque J, Ros J, Arroyo V, Rodés J, Friedman SL, Carmeliet P, Jiménez W, Morales-Ruiz M: Antiangiogenic treatment with sunitinib ameliorates inflammatory infiltrate, fibrosis, and portal pressure in cirrhotic rats. Hepatology. 2007, 46: 1919-1926.CrossRefPubMed Tugues S, Fernandez-Varo G, Muñoz-Luque J, Ros J, Arroyo V, Rodés J, Friedman SL, Carmeliet P, Jiménez W, Morales-Ruiz M: Antiangiogenic treatment with sunitinib ameliorates inflammatory infiltrate, fibrosis, and portal pressure in cirrhotic rats. Hepatology. 2007, 46: 1919-1926.CrossRefPubMed
90.
Zurück zum Zitat Nagai T, Arao T, Furuta K, Sakai K, Kudo K, Kaneda H, Tamura D, Aomatsu K, Kimura H, Fujita Y, Matsumoto K, Saijo N, Kudo M, Nishio K: Sorafenib inhibits the hepatocyte growth factor-mediated epithelial mesenchymal transition in hepatocellular carcinoma. Mol Cancer Ther. 2011, 10: 169-177.CrossRefPubMed Nagai T, Arao T, Furuta K, Sakai K, Kudo K, Kaneda H, Tamura D, Aomatsu K, Kimura H, Fujita Y, Matsumoto K, Saijo N, Kudo M, Nishio K: Sorafenib inhibits the hepatocyte growth factor-mediated epithelial mesenchymal transition in hepatocellular carcinoma. Mol Cancer Ther. 2011, 10: 169-177.CrossRefPubMed
91.
Zurück zum Zitat Park CY, Kim DK, Sheen YY: EW-7203, a novel small molecule inhibitor of transforming growth factor-β (TGF-β) type I receptor/activin receptor-like kinase-5, blocks TGF-β1-mediated epithelial-to-mesenchymal transition in mammary epithelial cells. Cancer Sci. 2011, 102: 1889-1896.CrossRefPubMed Park CY, Kim DK, Sheen YY: EW-7203, a novel small molecule inhibitor of transforming growth factor-β (TGF-β) type I receptor/activin receptor-like kinase-5, blocks TGF-β1-mediated epithelial-to-mesenchymal transition in mammary epithelial cells. Cancer Sci. 2011, 102: 1889-1896.CrossRefPubMed
92.
Zurück zum Zitat Park CY, Son JY, Jin CH, Nam JS, Kim DK, Sheen YY: EW-7195, a novel inhibitor of ALK5 kinase inhibits EMT and breast cancer metastasis to lung. Eur J Cancer. 2011, 47: 2642-2653.CrossRefPubMed Park CY, Son JY, Jin CH, Nam JS, Kim DK, Sheen YY: EW-7195, a novel inhibitor of ALK5 kinase inhibits EMT and breast cancer metastasis to lung. Eur J Cancer. 2011, 47: 2642-2653.CrossRefPubMed
93.
Zurück zum Zitat Huber MA, Maier HJ, Alacakaptan M, Wiedemann E, Braunger J, Boehmelt G, Madwed JB, Young ER, Marshall DR, Pehamberger H, Wirth T, Kraut N, Beug H: BI 5700, a Selective Chemical Inhibitor of IκB Kinase 2, Specifically Suppresses Epithelial-Mesenchymal Transition and Metastasis in Mouse Models of Tumor Progression. Genes Cancer. 2010, 1: 101-114.PubMedCentralCrossRefPubMed Huber MA, Maier HJ, Alacakaptan M, Wiedemann E, Braunger J, Boehmelt G, Madwed JB, Young ER, Marshall DR, Pehamberger H, Wirth T, Kraut N, Beug H: BI 5700, a Selective Chemical Inhibitor of IκB Kinase 2, Specifically Suppresses Epithelial-Mesenchymal Transition and Metastasis in Mouse Models of Tumor Progression. Genes Cancer. 2010, 1: 101-114.PubMedCentralCrossRefPubMed
94.
Zurück zum Zitat Ma Q, Guin S, Padhye SS, Zhou YQ, Zhang RW, Wang MH: Ribosomal protein S6 kinase (RSK)-2 as a central effector molecule in RON receptor tyrosine kinase mediated epithelial to mesenchymal transition induced by macrophage-stimulating protein. Mol Cancer. 2011, 10: 66-PubMedCentralCrossRefPubMed Ma Q, Guin S, Padhye SS, Zhou YQ, Zhang RW, Wang MH: Ribosomal protein S6 kinase (RSK)-2 as a central effector molecule in RON receptor tyrosine kinase mediated epithelial to mesenchymal transition induced by macrophage-stimulating protein. Mol Cancer. 2011, 10: 66-PubMedCentralCrossRefPubMed
95.
Zurück zum Zitat Ghosh B, Benyumov AO, Ghosh P, Jia Y, Avdulov S, Dahlberg PS, Peterson M, Smith K, Polunovsky VA, Bitterman PB, Wagner CR: Nontoxic chemical interdiction of the epithelial-to-mesenchymal transition by targeting cap-dependent translation. ACS Chem Biol. 2009, 4: 367-377.PubMedCentralCrossRefPubMed Ghosh B, Benyumov AO, Ghosh P, Jia Y, Avdulov S, Dahlberg PS, Peterson M, Smith K, Polunovsky VA, Bitterman PB, Wagner CR: Nontoxic chemical interdiction of the epithelial-to-mesenchymal transition by targeting cap-dependent translation. ACS Chem Biol. 2009, 4: 367-377.PubMedCentralCrossRefPubMed
96.
Zurück zum Zitat Chua KN, Sim WJ, Racine V, Lee SY, Goh BC, Thiery JP: A cell-based small molecule screening method for identifying inhibitors of epithelial-mesenchymal transition in carcinoma. PLoS One. 2012, 7: e33183-PubMedCentralCrossRefPubMed Chua KN, Sim WJ, Racine V, Lee SY, Goh BC, Thiery JP: A cell-based small molecule screening method for identifying inhibitors of epithelial-mesenchymal transition in carcinoma. PLoS One. 2012, 7: e33183-PubMedCentralCrossRefPubMed
Metadaten
Titel
Epithelial-mesenchymal transition: focus on metastatic cascade, alternative splicing, non-coding RNAs and modulating compounds
verfasst von
Timur R Samatov
Alexander G Tonevitsky
Udo Schumacher
Publikationsdatum
01.12.2013
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2013
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-12-107

Weitere Artikel der Ausgabe 1/2013

Molecular Cancer 1/2013 Zur Ausgabe

Mammakarzinom: Brustdichte beeinflusst rezidivfreies Überleben

26.05.2024 Mammakarzinom Nachrichten

Frauen, die zum Zeitpunkt der Brustkrebsdiagnose eine hohe mammografische Brustdichte aufweisen, haben ein erhöhtes Risiko für ein baldiges Rezidiv, legen neue Daten nahe.

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

Das sind die führenden Symptome junger Darmkrebspatienten

Darmkrebserkrankungen in jüngeren Jahren sind ein zunehmendes Problem, das häufig längere Zeit übersehen wird, gerade weil die Patienten noch nicht alt sind. Welche Anzeichen Ärzte stutzig machen sollten, hat eine Metaanalyse herausgearbeitet.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.