TGF-beta Rev 2019
TGF-beta Rev 2019
TGF-beta Rev 2019
Molecular Sciences
Review
TGF-β-Mediated Epithelial-Mesenchymal Transition
and Cancer Metastasis
Yang Hao, David Baker and Peter ten Dijke *
Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center,
Einthovenweg 20, 2300 RC Leiden, The Netherlands; Y.Hao@lumc.nl (Y.H.); D.A.Baker@lumc.nl (D.B.)
* Correspondence: p.ten_dijke@lumc.nl; Tel.: +31-71-526-9271
Received: 18 April 2019; Accepted: 24 May 2019; Published: 5 June 2019
Abstract: Transforming growth factor β (TGF-β) is a secreted cytokine that regulates cell proliferation,
migration, and the differentiation of a plethora of different cell types. Consistent with these findings,
TGF-β plays a key role in controlling embryogenic development, inflammation, and tissue repair,
as well as in maintaining adult tissue homeostasis. TGF-β elicits a broad range of context-dependent
cellular responses, and consequently, alterations in TGF-β signaling have been implicated in many
diseases, including cancer. During the early stages of tumorigenesis, TGF-β acts as a tumor suppressor
by inducing cytostasis and the apoptosis of normal and premalignant cells. However, at later stages,
when cancer cells have acquired oncogenic mutations and/or have lost tumor suppressor gene
function, cells are resistant to TGF-β-induced growth arrest, and TGF-β functions as a tumor
promotor by stimulating tumor cells to undergo the so-called epithelial-mesenchymal transition
(EMT). The latter leads to metastasis and chemotherapy resistance. TGF-β further supports cancer
growth and progression by activating tumor angiogenesis and cancer-associated fibroblasts and
enabling the tumor to evade inhibitory immune responses. In this review, we will consider the role of
TGF-β signaling in cell cycle arrest, apoptosis, EMT and cancer cell metastasis. In particular, we will
highlight recent insights into the multistep and dynamically controlled process of TGF-β-induced
EMT and the functions of miRNAs and long noncoding RNAs in this process. Finally, we will discuss
how these new mechanistic insights might be exploited to develop novel therapeutic interventions.
Keywords: EMT; lncRNA; metastasis; miRNA; SMAD; TGF-β; targeted therapy; tumor microenvironment
1. Introduction
Cancer treatments have been refined over a period spanning several thousand years, culminating in
the primary modern approaches of surgery, chemotherapy and radiation therapy [1]. In recent decades,
more targeted and personalized treatments have gained prominence. The fundamental aim of these
therapies is to specifically kill tumor cells while leaving healthy tissues intact [2]. Despite demonstrable
progress, these strategies frequently deliver relatively modest improvements in disease outcomes
owing to acquired resistance and excessive toxicity [3]. These findings indicate the need for greater
optimization of current treatments as well as the identification of alternative targets for the development
of novel cancer remedies.
Cancer is a complex disease in which tumor cell heterogeneity and the reciprocal interplay between
tumor cells and surrounding stromal cells and extracellular matrix are key determinants in tumor
progression and therapy response. Tumorigenesis shows similarity to subverted normal embryogenic
developmental processes in which communication between cells is controlled by cytokines that act
in an autocrine, paracrine or juxtacrine manner. In this light, we will provide a general overview
of the established roles of one such important developmental cancer signaling pathway, namely
the transforming growth factor-β (TGF-β), in tumorigenesis [4]. In particular, we consider how
this essential TGF-β signaling network orchestrates the epithelial to mesenchymal transition (EMT),
the mechanism by which cancer cells lose polarity and separate from each other, adopt the characteristics
of a mesenchymal phenotype, become motile and invade distant sites. Key TGF-β-induced effectors
in this process are the transcriptional repressors of E-cadherin, e.g., SNAIL1, SNAIL2 (also termed
SLUG), ZEB1/2 and TWIST. Moreover, miRNAs and long noncoding RNAs (lncRNAs) are emerging
as potentially quantifiable biomarkers of cancer status and are potential targets for anti-metastatic
therapies [5]. In this review, we will highlight the pivotal role of these molecules in regulating TGF-β
signaling and epithelial-mesenchymal transition (EMT).
90
91 Figure 1.1. TGF-β/SMAD
Figure TGF-β/SMAD and and non-SMAD
non-SMAD signaling.
signaling. TGF-β
TGF-β elicits
elicits its
its cellular
cellular responses
responses by
by forming
forming
92 ligand-induced
ligand-induced complex formation of TGF-β type I and type II cell surface receptors (i.e.(i.e.,
complex formation of TGF-β type I and type II cell surface receptors TβRITβRI
and
93 and TβRII) that are endowed with serine/threonine kinase activity. The extracellular signal
TβRII) that are endowed with serine/threonine kinase activity. The extracellular signal is transduced is
transduced across the plasma membrane through the action of the constitutively active TβRII kinase that
94 across the plasma membrane through the action of the constitutively active TβRII kinase that
phosphorylates specific serine and threonine residues in the intracellular juxtamembrane GS domain
95 phosphorylates specific serine and threonine residues in the intracellular juxtamembrane GS domain
of TβRI. Intracellular signaling is then initiated when the activated TβRI kinase phosphorylates or
96 of TβRI. Intracellular signaling is then initiated when the activated TβRI kinase phosphorylates or
activates intracellular signal mediators. In the case of the canonical SMAD pathway, TβRI recruits and
97 activates intracellular signal mediators. In the case of the canonical SMAD pathway, TβRI recruits
phosphorylates specific R-SMADs, e.g., SMAD2 and SMAD3, which can form heteromeric complexes
98 and phosphorylates specific R-SMADs, e.g., SMAD2 and SMAD3, which can form heteromeric
with SMAD4. These transcription factor complexes then translocate into the nucleus and cooperate with
99 complexes with SMAD4. These transcription factor complexes then translocate into the nucleus and
other transcription regulators to regulate target gene expression. In the non-SMAD pathway, TGF-β
100 cooperate with other transcription regulators to regulate target gene expression. In the non-SMAD
receptors activate other pathways, including various branches of MAPK pathways, RHO-like GTPase
101 pathway, TGF-β receptors activate other pathways, including various branches of MAPK pathways,
signaling pathways and phosphatidylinositol-3-kinase (PI3K)/AKT pathways. Inhibitory signals are
102 RHO-like GTPase signaling pathways and phosphatidylinositol-3-kinase (PI3K)/AKT pathways.
indicated with inhibitory red arrows; Stimulatory signals are indicated with green arrows.
103 Inhibitory signals are indicated with inhibitory red arrows; Stimulatory signals are indicated with
104 greenR-
The arrows.
and Co-SMADs have two conserved domains, termed Mad homology (MH)1 and
MH2 domains at the amino-terminal (MH1) and carboxy-terminal (MH2) ends of the proteins.
105 The R- and Co-SMADs have two conserved domains, termed Mad homology (MH)1 and MH2
MH1 and MH2 are separated by a flexible linker region. R-SMAD phosphorylation by type I receptors
106 domains at the amino-terminal (MH1) and carboxy-terminal (MH2) ends of the proteins. MH1 and
occurs on two serine residues, which comprise the SXS sequence motif, at the C-termini. SMAD3 and
107 MH2 are separated by a flexible linker region. R-SMAD phosphorylation by type I receptors occurs
SMAD4, but not SMAD2, bind directly to the consensus 50 -CAGA-30 DNA motif. Heteromeric complex
108 on two serine residues, which comprise the SXS sequence motif, at the C-termini. SMAD3 and
formation between R-SMADs and SMAD4 is mediated by MH2 domains. Heteromeric complex
109 SMAD4, but not SMAD2, bind directly to the consensus 5′-CAGA-3′ DNA motif. Heteromeric
formation of R-SMADs and SMAD4 exposes nuclear import signals and shields nuclear export signals,
110 complex formation between R-SMADs and SMAD4 is mediated by MH2 domains. Heteromeric
resulting in their nuclear accumulation. In the nucleus, they can act as transcription factors in concert
111 complex formation of R-SMADs and SMAD4 exposes nuclear import signals and shields nuclear
with other DNA binding transcription factors, coactivators and repressors and chromatin remodeling
112 export signals, resulting in their nuclear accumulation. In the nucleus, they can act as transcription
factors, which enable diverse transcriptional responses depending upon the particular combination
113 factors in concert with other DNA binding transcription factors, coactivators and repressors and
of proteins [19]. I-SMAD7, which has a carboxy terminal region with homology to R-SMADs and
114 chromatin remodeling factors, which enable diverse transcriptional responses depending upon the
SMAD4 MH2 domains, antagonizes the activation of TGF-β receptor/SMAD signaling. SMAD7 achieves
115 particular combination of proteins [19]. I-SMAD7, which has a carboxy terminal region with
this via multiple mechanisms, including the recruitment of the E3 ubiquitin ligase SMURF2 to the
116 homology to R-SMADs and SMAD4 MH2 domains, antagonizes the activation of TGF-β
activated receptor, thereby targeting the TGF-β receptor for proteasomal and lysosomal degradation.
117 receptor/SMAD signaling. SMAD7 achieves this via multiple mechanisms, including the recruitment
SMAD7 can also attenuate signaling by recruiting phosphatases to the activated TGF-β receptor, which
118 of the E3 ubiquitin ligase SMURF2 to the activated receptor, thereby targeting the TGF-β receptor for
mediate receptor inactivation by the dephosphorylation of specific amino acid residues [20].
119 proteasomal and lysosomal degradation. SMAD7 can also attenuate signaling by recruiting
120 phosphatases to the activated TGF-β receptor, which mediate receptor inactivation by the
121 dephosphorylation of specific amino acid residues [20].
Int. J. Mol. Sci. 2019, 20, 2767 4 of 34
TGF-β can also signal via non-SMAD pathways. This process can occur directly or indirectly [21].
An example of an indirect mechanism is TGF-β/SMAD-stimulated expression of growth factors, such
as platelet-derived growth factor (PDGF) and epidermal growth factor (EGF), which thereafter initiate
non-SMAD responses. Non-SMAD signaling can also be initiated directly downstream of the TGF-β
receptor by activation of various branches of the MAP kinase (MAPK) pathway, Rho-like GTPase
signaling pathways and the phosphatidylinositol-3-kinase (PI3K)/AKT pathway [22]. For example,
activated TβRI can induce tyrosine phosphorylation of SHCA, which associates with GRB2, and recruits
a GRB2/SOS complex, thereby triggering activation of the RAS/RAF/MAP kinase signaling cascade [23].
Activated TGF-β receptor complexes also induce K63-linked polyubiquitination of TRAF4/6, leading
to the recruitment of TAK1 and triggering its activation, thus allowing TAK1 to activate JNK signaling
through MKK4 or P38 MAPK signaling via MKK3/6 [18,24,25]. At tight junctions, TβRII phosphorylates
PAR6 at serine residue 345. This phosphorylated PAR6 recruits SMURF1 to the activated TβRI-TβRII
receptor complex. The PAR6-SMURF1 complex subsequently mediates localized ubiquitination
and turnover of RHOA at cellular protrusions [26]. TGF-β can promote the interaction between
CDC42 GTPase/RAC and p21-activated kinase (PAK) 2, an interaction that is dependent on SMAD7 [27].
TGF-β receptors interact with the regulatory p85 subunit of PI3K, resulting in activation of the
PI3K/AKT pathway, which controls translational responses through mammalian target of rapamycin
(mTOR) [28]. Activation of non-SMAD signaling occurs in a context-dependent manner, and these
pathways also crosstalk with the canonical SMAD pathway.
and plays an important role in cell death by mediating delayed TGF-β-induced p38 MAP kinase
activation [42]. Here, the effects of TGF-β in proapoptotic signaling occur in a context-dependent
manner. Additionally, the TGF-β signaling pathway can be coupled to the cell death machinery through
the induction of reactive oxygen species (ROS) [43], apoptosis genes (SHIP and TIEG), modulation
of epigenetic regulators (DNMTs) [44], H3K79me3 and H2BK120me1 [45], and telomere shortening
through regulating human telomerase reverse transcriptase (hTERT) in the breast cancer MCF-7 cell
line
Int. J.[46]. These
Mol. Sci. 2019,effects of PEER
20, x FOR TGF-β in proapoptotic signaling occur in a context-dependent manner.
REVIEW 5 of 37
158
159 Figure
Figure 2. TGF-β-induced growth
2. TGF-β-induced growth inhibition
inhibition and
and apoptosis. The TGF-β/SMAD
apoptosis. The TGF-β/SMAD pathway pathway can
can arrest
arrest cells
cells
160 in
in the
the G1
G1phase
phaseofofthe
thecell
cellcycle
cyclebybymodulating
modulatingthe theexpression
expression ofof
specific genes,
specific genes,including
including induction
inductionof
161 4EBP1,
of 4EBP1,p15INK4b,
p15INK4b,p16INK4A,
p16INK4A, p21WAF1
p21WAF1 andand
p57KIP2
p57KIP2andand
repression of Idofproteins,
repression E2F, E2F,
Id proteins, c-MYC and
c-MYC
162 CDC25a
and CDC25a genes. The The
genes. TGF-β/SMAD
TGF-β/SMAD pathway can can
pathway induce cellcell
induce apoptosis by inducing
apoptosis by inducingthethe
expression
expressionof
163 proapoptotic genes, such as BCL-XL, BIM, BIK, SHIP and TIEG. The TGF-β activation
of proapoptotic genes, such as BCL-XL, BIM, BIK, SHIP and TIEG. The TGF-β activation of TAK-1 of TAK-1 occurs
164 via TRAF6
occurs via and
TRAF6the and
adaptor XIAP-mediated
the adaptor TAB/TAK-1
XIAP-mediated complex and
TAB/TAK-1 GADD45β.
complex ActivationActivation
and GADD45β. of NF-κB
165 by the TAKbypathway
of NF-κB the TAKstimulates
pathway p38/JNK
stimulatesphosphorylation, which has been
p38/JNK phosphorylation, reported
which to lead
has been to apoptosis.
reported to lead
166 Additionally, the TGF-β signaling pathway can be coupled to the cell death machinery
to apoptosis. Additionally, the TGF-β signaling pathway can be coupled to the cell death machinery through ROS,
167 autophagy activation (ATG-5/-6/-7), induction of DAPK expression, epigenetic changes
through ROS, autophagy activation (ATG-5/-6/-7), induction of DAPK expression, epigenetic changes and shortening
168 of
andtelomere length
shortening (regulating
of telomere hTERT).
length Inhibitory
(regulating hTERT).signals are indicated
Inhibitory with
signals are inhibitory
indicated with red arrows;
inhibitory
169 Stimulatory signals are indicated with green arrows. SMAD-mediated transcriptional
red arrows; Stimulatory signals are indicated with green arrows. SMAD-mediated transcriptional events are
indicated with the black arrow.
170 events are indicated with the black arrow.
The TGF-β signaling pathway can inhibit tumor growth by multiple other mechanisms, including
171 In addition to cell cycle arrest, TGF-β can induce cell apoptosis in the early phase of
activating autophagy in certain human cancer cells. For example, in human hepatocellular carcinoma
172 tumorigenesis [29]. However, the precise mechanisms by which TGF-β induces this effect in different
cell lines, TGF-β induced the accumulation of autophagosomes and increased the expression levels of
173 cell types remain unclear. The expression of some apoptotic regulators (such as growth arrest and
the autophagy markers Autophagy-related 5 (ATG5), Beclin1, ATG7 and death-associated protein kinase
174 DNA damage (GADD) 45, Bcl-2-like protein 11 (BIM), BCL-2 interacting killer (BIK), death associated
(DAPK) [47]. Moreover, siRNA-mediated silencing of autophagy genes attenuated TGF-β-mediated
175 protein kinase (DAPK), FAS, and B-cell lymphoma-extra large (BCL-XL)) was shown to be regulated
growth inhibition and induction of the proapoptotic genes BIM and BMF in human hepatocellular
176 by the TGF-β/SMAD signaling pathway [36]. For example, BIM was found to be a key mediator of
carcinoma cells [48].
177 TGF-β-induced apoptosis in intestinal adenoma cells [37] and in hepatocarcinoma cells [38]. TGF-β1-
Because of its central role in tumor suppression, TGF-β signaling components were found to
178 associated regulatory SMAD proteins bind to the BIK (also known as NBK) promoter, which encodes
be mutated and functionally inactivated in various cancers [49]. The first example was SMAD4,
179 a proapoptotic sensitizer protein in B cells [39]. After TGF-β1 treatment, researchers found that TGF-
which is frequently mutated in gastrointestinal cancers [50]. Subsequently, other TGF-β signaling
180 β induced SMAD-dependent binding between the proapoptotic effector BIM and BCL-XL in gastric
181 carcinoma cell lines [40] and a decrease in BCL-XL expression followed by activation of the apoptosis
182 proteins caspase-9 and caspase-3 in human hepatoma cells (HuH-7) [41]. TGF-β can activate the
183 TAK1-p38/JNK pathway, which has been reported to lead to apoptosis in HEK 293T cells [25]. This
184 molecule also promoted the expression of SMAD-dependent GADD45β in hepatocytes and plays an
Int. J. Mol. Sci. 2019, 20, 2767 6 of 34
components, e.g., TGF-β receptors [51] and SMADs (SMAD2 and SMAD3), were found to be mutated
in various cancers, including bladder, colon, breast, esophageal, stomach, brain, liver, and lung
cancers [52]. Loss of tumor suppressor function and epigenome and microenvironmental changes can
also affect the tumor-promoting activity of the TGF-β receptor/SMAD pathway [53]. For example,
in gastrointestinal tumors, TβR1 activity was decreased due to the methylation status of the TβR1
promoter [54]. In turn, TGF-β/SMAD can affect the epigenome of genes involved in cancer processes.
TGF-β and SMAD2/3 show oncogenic activities, such as promoting glioma cell proliferation, by affecting
the methylation status of the platelet-derived growth factor-β (PDGF-B) gene and autocrine PDGF-B
signaling within tumor microenvironments [55]. TGF-β stimulated myofibroblast percent and invasion
rate in tumor-associated fibroblasts (CAFs) that increase tumor invasion [56].
235
236 Figure
Figure 3.3. TGF-β mediates EMT.
TGF-β mediates EMT. TGF-β
TGF-β isis aa strong
strong promoter
promoter of of EMT,
EMT, which
which is
is characterized
characterized by
by aa loss
loss
237 of epithelial and gain of mesenchymal markers. Polar epithelial cells remodel into highly
of epithelial and gain of mesenchymal markers. Polar epithelial cells remodel into highly migratory migratory
238 mesenchymal
mesenchymal cells,
cells, followed by decreased
followed by decreased adhesion of cells
adhesion of cells and
and loss
loss of
of polarity
polarity and
and tight
tight junctions.
junctions.
239 TGF-β via SMAD or non-SMAD signaling pathways can induce the expression of
TGF-β via SMAD or non-SMAD signaling pathways can induce the expression of several EMT-TFs, several EMT-TFs,
240 such
such asas SNAIL1,
SNAIL1, SNAIL2,
SNAIL2, ZEB1/2
ZEB1/2 and
and TWIST.
TWIST. The The migratory
migratory andand invasive
invasive mesenchymal-like
mesenchymal-like tumor
tumor
241 cells
cells extravasate from primary lesions into blood or lymphatic vessels and then intravasate
extravasate from primary lesions into blood or lymphatic vessels and then intravasate to
to distant
distant
242 sites
sites via,
via, where
where they
they form
form metastatic
metastatic colonies
colonies upon
upon MET.
MET.
5.2. Molecular Mechanisms in TGF-β-induced EMT
243 Increasing data have shown that EMT is not a single, stereotypical program but instead has a
244 SMAD
multistep levels/activities
process that passes mediate
through TGF-β-induced EMT by
intermediate hybrid inducing
states theEMT
(partial expression of E-cadherin
state, P-EMT) during
245 transcriptional
the transition fromrepressors, such
epithelial as SNAIL, ZEBcells
to mesenchymal and[66].
TWIST,
The which cooperatetransition
TGF-β-induced with otherfromtranscription
epithelial
246 regulators
to P-EMT in the nucleuswhereas
is reversible, [58]. Several additional
the transition from lines of evidence
P-EMT argue forcells
to mesenchymal a roleis of SMADs
potentially
247 in TGF-β-induced
irreversible dependingEMT,on and someofexamples
the type cell that isare provided
involved [59].below. Knockdown
Studies have found thatof SMAD4
there are in
at
248 MDA-MB-231
least seven tumor breast cancer cells robustly
subpopulations attenuated
associated bone metastasis
with different EMT stagesin nude miceand
in skin andbreast
significantly
cancer
249 prolonged
tissues, whichsurvival of the treated
contributes animals [70].
to intratumor SMAD3 and
heterogeneity. SMAD4
These EMT interact to form adisplayed
subpopulations complex
250 with SNAIL1
differences that targets
in cellular the tight-junction
plasticity, invasiveness, protein (CAR) potential,
and metastatic and E-cadherin
and tumorduring
cellsTGF-β-driven
with an early
251 EMT
stage of EMT were most likely to metastasize [67]. An interesting paper reported that TGF-βinhibited
in breast epithelial cells. Conversely, co-silencing of SNAIL1 and SMAD4 by siRNA activates
252 repression
scleroderma ofepithelial
CAR andcells
occludin
to the during
P-EMT EMT
process[71]. In addition,
in fibrotic SMAD3/SMAD4-mediated
skin [68]. Related to this, single cellSNAILRNA-
253 transcription
seq showed that contributed to EMT
P-EMT plays an during skinrole
important carcinogenesis, whilecancer,
in head and neck SMAD2 loss
and significantly
further in vitroincreased
analyses
254 this effect [72].
suggested thatMoreover, SMAD7, the
TGF-β dynamically transcriptional
controls target between
the transition and negativeP-EMTregulator of TGF-β signaling,
and non-P-EMT states in
255 upregulated
cells [69]. TGF-β and inducing SMAD7 transcription prevented TGF-β-induced EMT and invasion
of cancer cells [73]. Additionally, ubiquitin ligases that promote poly-ubiquitination and proteasomal
256 5.2. MolecularofMechanisms
degradation in TGF-β-induced
SMADs affect EMT is illustrated by E3 ubiquitin ligase RNF8, which
EMT. This finding
activates TWIST via K63-linked ubiquitination to promote EMT and cancer stem cell (CSC) self-renewal,
257 SMAD levels/activities mediate TGF-β-induced EMT by inducing the expression of E-cadherin
resulting in enhanced metastasis and chemoresistance in breast cancer [74].
258 transcriptional repressors, such as SNAIL, ZEB and TWIST, which cooperate with other transcription
TGF-β can also induce EMT in a non-SMAD-dependent fashion, for example, by promoting cytoskeletal
259 regulators in the nucleus [58]. Several additional lines of evidence argue for a role of SMADs in TGF-
remodeling, which leads to activation of ERK [75]. The ERK required for cytoskeletal remodeling interacts
260 β-induced EMT, and some examples are provided below. Knockdown of SMAD4 in MDA-MB-231
with SHC or GRB2 to form an SHC-GRB2-ERK complex, which is a key component of TGF-β-induced
261 breast cancer cells robustly attenuated bone metastasis in nude mice and significantly prolonged
tumor invasion and metastasis [76]. ERK substrates, AP-1 family members, enhance SMAD transcriptional
262 survival of the treated animals [70]. SMAD3 and SMAD4 interact to form a complex with SNAIL1
263 that targets the tight-junction protein (CAR) and E-cadherin during TGF-β-driven EMT in breast
Int. J. Mol. Sci. 2019, 20, 2767 8 of 34
activity to regulate gene expression and TGF-β-induced EMT [77]. However, the RHO-like GTPases,
including RHOA, RAC and CDC42, are also involved in TGF-β-induced EMT. TGF-β regulates cytoskeletal
changes via mediating RHO GTPase to achieve the dissolution of tight junctions among cells. TGF-β
mediates the RHOA activity level and promotes the activation of LIM kinase (LIMK) by Rho-related kinase
(ROCK) and phosphorylated myosin light chain (MLC) to inhibit cofilin [78]. In addition, TGF-β affects
tight junctions through SMAD7-dependent CDC42-PAK1 (p21-activated kinase) and filopodia formation.
The TRAF6-TAK1-JNK/P38 pathway and PI3K-AKT-mTOR signaling are also necessary non-SMAD
pathways for TGF-β-mediated EMT [79,80]. Scientists have shown that PI3K/AKT signaling promotes
tumor metastasis by inducing TWIST1 phosphorylation, via a crosstalk between AKT/PKB and TGF-β
signaling [81]. Twist also had a significant effect on AKT signaling pathway activation by inducing
expression of miR-10b in gastric cancer cells, and the miR-10b induced by TWIST increased the expression
of a well-characterized pro-metastatic gene, RHOC [82]. Moreover, in an orthotopic syngeneic mouse tumor
model, metastasis caused by EMT was attenuated in mice treated with the p38 inhibitor SB203580 [83].
TRAF6 knockdown inhibited the migration and invasion caused by EMT of SCCHN (squamous cell
carcinoma of head and neck) cells [84].
In addition to these SMAD/non-SMAD pathways, TGF-β affects the activities of other EMT
trigger signaling pathways (NOTCH, WNT, INTEGRIN, etc.) by several complexes, such as ZEB1/2,
the SNAIL1-SMAD3/4 complex, the β-catenin-SMAD2 complex, the LEF1-SMAD3/4 complex and
the SMAD3-AP1-1 complex [85]. As early as 20 years ago, scientists discovered that SMAD4 could
form a complex with β-catenin and LEF1/TCF (lymphoid enhancer factor1), which are downstream
components of the Wnt signaling cascade in vivo [86]. SMAD signaling subsequently stimulates the
formation of β-catenin/LEF1 and SNAIL-LEF1 complexes, which promote EMT by inhibiting the
expression of E-cadherin [87,88]. TGF-β can promote EMT associated with WNT-11 signals through
the WNT-11 receptor FZD8 in prostate cancer [89]. Moreover, WNT-11 signaling mediates the nuclear
entry process of TAK1 (TGF-β-activated kinase) [90]. The TGF-β and NOTCH pathways coregulate
a large cohort of genes in human cancer, such as renal cell carcinoma [91]. R-SMAD activates the
NOTCH ligand Jagged1 to release Notch intracellular domain (ICN) and then binds to CLS (an acronym
for CBF-1/RBPJ-κ in Homo sapiens/Mus musculus respectively, Suppressor of Hairless in Drosophila
melanogaster, Lag-1 in Caenorhabditis elegans). This ICN-CLS complex induces the binding of the
transcription factor SNAIL or HEYl to the E-cadherin E-box to reduce E-cadherin expression and
initiate the EMT process [92]. Moreover, SMAD signaling and MAPK/JNK signaling converge at
AP1-binding promoter sites by SMAD3 and SMAD4, which cooperate with c-JUN/c-FOS [93], and the
RAS-ERK MAP kinase pathways are likely to act synergistically with TGF-β and contribute to multiple
aspects of the EMT, including the pro-invasive and pro-metastatic behavior of tumor cells of diverse
tissue origins [94]. TGF-β increases the level of SNAIL and promotes EMT with the cooperation of
oncogenic RAS [57] and the transcription factor nuclear factor κB (NF-κB) [95]. In addition, TGF-β
upregulates receptors and ligands of PDGF, leading to phosphorylation of PI3K and activation of the
SRC/STAT3 pathway, thereby triggering the EMT process [96].
321
322 Figure 4. MicroRNAs in TGF-β-induced EMT. At the heart of TGF-β-induced EMT, there are two
Figure 4. MicroRNAs in TGF-β-induced EMT. At the heart of TGF-β-induced EMT, there are two
323 mainmain
double-negative
double-negative feedback
feedback regulatory
regulatoryloops
loops of
of miRNAs, e.g.,the
miRNAs, e.g., theSNAIL1/miR-34
SNAIL1/miR-34 family
family and and
324 ZEB/miR-200 family and the autocrine TGF-β/miR-200 negative feedback loop.
ZEB/miR-200 family and the autocrine TGF-β/miR-200 negative feedback loop. Specifically, TGF-β Specifically, TGF-β
325 downregulates miR-200 family members, thereby increasing ZEB1 and ZEB2 mRNA
downregulates miR-200 family members, thereby increasing ZEB1 and ZEB2 mRNA levels indirectly, levels indirectly,
326 and ZEB binds
and ZEB to promoters
binds to promoters ofofthethemiR-200
miR-200 members
members to torepress
represstheir
their expression,
expression, thus
thus constituting
constituting a a
327 double-negative
double-negative regulatory
regulatory loop.
loop. TheThesame
samesituation
situation occurs
occursininSNAIL1
SNAIL1 and
andmiR-34,
miR-34,which are directly
which are directly
328 linked
linked to status.
to p53 p53 status.
For For
the the autocrine
autocrine TGF-β/miR-200
TGF-β/miR-200 system,autocrine
system, autocrineTGF-β
TGF-β positively
positively regulates
regulates the
329 the expression
expression of SNAIL1of SNAIL1
and then and then increases
increases ZEB mRNA
ZEB mRNA and protein
and protein levels,levels, further
further downregulating
downregulating miR-200.
330 miR-200.
Inhibitory Inhibitory
signals signals are
are indicated with indicated with
inhibitory inhibitory
(dashed) red(dashed)
arrows; red arrows; Stimulatory
Stimulatory signals are signals
indicatedarewith
331 indicated with green arrows.
green arrows.
High mobility group protein A2 (HMGA2) has been shown to promote lung cancer progression
in mouse and human cells by competing with TGF-β type III receptor for the let-7 microRNA (miRNA)
family [107], while decreased let-7g levels influence the TGF-β pathway by targeting TβR1 and
SMAD2 gene expression [108]. The overexpression of miR-10b induced TGF-β-driven EMT in breast
cancer [109]. In contrast, silencing of miR-10b markedly suppressed the formation of lung metastases
by inhibiting its target gene HOXD10 in a mouse mammary tumor model [110].
TGF-β upregulates certain miRNAs, such as miR-182, which prolong NF-κB activation by
directly suppressing an NF-κB negative regulator (cylindromatosis, CYLD) [111]. Overexpression of
miR-182 restrained SMAD7 expression and promoted breast tumor invasion and TGF-β-induced
osteoclastogenesis and bone metastasis [73]. MiR-181a, another miRNA upregulated by TGF-β, promoted
TGF-β-mediated EMT and metastasis in breast cancer [112] and via repression of SMAD7 in ovarian cancer
progression [113]. TGF-β activates miR-1269 by SOX4 and thereby enhances TGF-β signaling by targeting
SMAD7 and HOXD10. This positive feedback loop significantly increased the ability of colorectal cancer
cells to invade and metastasize in vivo [114]. Overexpression of miR-216a/217 activated the PI3K/AKT and
TGF-β pathways by targeting PTEN, and SMAD7 underlies hepatocarcinogenesis and tumor recurrence
of hepatocarcinoma [115]. TGF-β induces the expression and promoter activity of miR-155 through
SMAD4. This change reduces RHOA protein and disrupts tight junction formation, leading to EMT [116].
Other research has shown that miR-206 inhibits autocrine production of TGF-β as well as downstream
neuropilin-1 (NRP1) and SMAD2 expression, resulting in decreased migration, invasion, and EMT in
breast cancer cells [117]. Several other miRNAs, such as miR-373, miR-655, miR206, miR-155, miR-140-5p,
miR-494, miR-125a/b, and miR-375, have been implicated in EMT [118–122]. However, for many of these
factors, their specific functions remain to be elucidated.
396 that a lncRNA activated by TGF-β, lncRNA-ATB, induces EMT and invasion by competitively
397Int. J. binding miR-200
Mol. Sci. 2019, family members, which promoted organ-specific metastasis by binding IL-11
20, 2767 11 of 34
398 mRNA. This competitive binding increased IL-11 mRNA stability, which caused autocrine induction
399 of IL-11 and subsequent activation of STAT3 signaling [131]. These findings suggest that lncRNA-
400activityATB,ofamiR-205
mediatorduring
of TGF-β EMT. Moreover,
signaling, couldelevated
predispose expression
HCC patients of lncRNA-PNUTS
to metastasis [132]. was correlated
Another
401withstudy showed that
upregulated lncRNA-PNUTS,
levels of ZEB mRNAs which is highly
[133]. LncRNA expressed
MEG3incan mesenchymal
modulate breast tumor of
the activity cells,
TGF-β
402genescompetitive binds to and neutralizes the activity of miR-205 during
by binding to distal regulatory elements [134]. Another lncRNA, DNM3OS, was associated withEMT. Moreover, elevated
403overexpression
expression of oflncRNA-PNUTS was correlated
TWIST1 and specifically with upregulated
contributed to EMT inlevels
ovarianof ZEB mRNAs
cancer [135].[133]. LncRNA
Recently, a paper
404showed MEG3 can modulate the activity of TGF-β genes by binding to distal
that lncRNA-MUF can directly activate WNT/β-catenin signaling and EMT by binding regulatory elements [134]. to
405 Another lncRNA, DNM3OS, was associated with overexpression of TWIST1 and specifically
ANNEXIN 2A. LncRNA-MUF can also indirectly promote EMT by competitively binding to miR-34a
406 contributed to EMT in ovarian cancer [135]. Recently, a paper showed that lncRNA-MUF can directly
and upregulating SNAIL1 expression [136].
407 activate WNT/β-catenin signaling and EMT by binding to ANNEXIN 2A. LncRNA-MUF can also
408 Reduced promote
indirectly lncRNAEMT H19 byexpression in hepatocarcinogenesis
competitively binding to miR-34a and (HCG) tissues from
upregulating SNAIL1patients with the
expression
409epithelial
[136]. TGF-β gene signature [137] but increased H19 expression promoted tumor metastasis after
410TGF-β treatment in Hep3B cells [138]. In a mouse model of spontaneous
Reduced lncRNA H19 expression in hepatocarcinogenesis (HCG) tissues from patients with the metastatic breast cancer,
411lncRNA H19 TGF-β
epithelial mediatedgeneEMT and [137]
signature METbut byincreased
differentially binding to
H19 expression the microRNAs
promoted miR-200b/c
tumor metastasis after and
412let-7bTGF-β[139].treatment
LncRNA in H19
Hep3Bcancells [138].
also In a mouse
interact modeland/or
with SLUG of spontaneous
EZH2, whichmetastatic breast E-cadherin
regulates cancer,
413expression
lncRNA[140].
H19 mediated
Lnc-Spry1 EMT is and MET by differentially
downregulated by TGF-β binding to the microRNAs
and plays miR-200b/c
a direct regulatory roleandin the
414earlylet-7b [139]. LncRNA H19 can also interact with SLUG and/or EZH2, which
stage of TGF-β-induced EMT, thus affecting cell invasion and migration. This molecule also regulates E-cadherin
415controlsexpression [140]. Lnc-Spry1 is downregulated by TGF-β and plays a direct regulatory role in the early
gene and protein expression levels through an interaction with the splicing factor U2AF65 [141].
416 stage of TGF-β-induced EMT, thus affecting cell invasion and migration. This molecule also controls
LncRNA-KRTAP5-AS1 and lncRNA-TUBB2A control the function of CLAUDIN-4 and thereby influence
417 gene and protein expression levels through an interaction with the splicing factor U2AF65 [141].
418EMTLncRNA-KRTAP5-AS1
in gastric cancer [142]. LncRNA HOTAIR (for HOX Transcript antisense intergenic RNA) acts as a
and lncRNA-TUBB2A control the function of CLAUDIN-4 and thereby
419crucial player EMT
influence during in EMT
gastricbycancer
mediating
[142]. aLncRNA
physicalHOTAIR
interaction
(for between SNAIL antisense
HOX Transcript and EZH2, which form
intergenic
420an enzymatic
RNA) actssubunit of the
as a crucial polycomb
player during repressive complex
EMT by mediating 2, the main
a physical writer of
interaction chromatin-repressive
between SNAIL and
421marks EZH2, which form an enzymatic subunit of the polycomb repressive complex 2, the mainsignaling
[143]. Together, these findings suggest that lncRNAs can be mediators of TGF-β writer of and
422maychromatin-repressive
serve as a potentialmarks target[143].
for anti-metastatic therapies.
Together, these findings The that
suggest mechanisms
lncRNAs can by be
which lncRNAs
mediators
423regulateof TGF-β
TGF-β signaling and are
signaling maylargely
serve asunknown,
a potential and
targethow
for anti-metastatic
they affect TGF-β therapies. The mechanisms
pathway components in
424cancer by metastasis
which lncRNAsremains regulate TGF-β signaling
to be discovered are 5largely
(Figure and Tableunknown,
1). and how they affect TGF-β
425 pathway components in cancer metastasis remains to be discovered (Figure 5 and Table 1).
426
Figure 5. LncRNAs in TGF-β signaling and TGF-β-induced EMT. LncRNAs associated with TGF-β
signaling and TGF-β-induced EMT in various cancer cell types. LncRNAs with high expression in
tumor tissues (in red) and low expression in tumor tissues (in green).
Int. J. Mol. Sci. 2019, 20, 2767 12 of 34
Table 1. LncRNAs involved in TGF-β signaling, focusing on those that impact TGF-β-induced EMT.
LncRNAs with high expression in tumor tissues (in red) and low expression in tumor tissues (in green).
Cancer Type LncRNA Function and Mechanism of Action Example of Key Findings or Experiments
Knockdown results in a morphological change of
Functions as a sponge of miR-141-3p, breast cancer cells from spindle-like to round shape
lncRNA-ATB [144]
increasing ZEB1 and ZEB2 expression. and in a remarkable inhibition of cell migration
and invasion.
Functions as an enhancer RNA (eRNA) to
activate Slug gene transcription in the Knockdown of either AC026904.1 or
nucleus, whereas UCA1 exerts a UCA1 prolongs survival time of the nude mice
AC026904.1 and UCA1 [145] competitive endogenous RNA (ceRNA) for bearing D3H2LN mammary tumors, these two
titrating miR-1 and miR-203a to promote genes exert critical roles in TGF-β-induced EMT
Slug expression at the post-transcriptional and promote invasion in metastatic breast cancer.
level in the cytoplasm.
Suppresses TGF-β-induced EMT by Overexpression reduces TGF-β-induced tumor
NKILA [146]
blocking NF-κB signaling metastasis in vivo.
Functions as a downstream effector Ectopic expression partly attenuates the
molecule, down-regulated by TGF-β1, TGF-β1-induced EMT and knockdown promotes
ANCR [147]
and is essential for TGF-β1-induced EMT TGF-β1-induced EMT and metastasis in breast
by decreasing RUNX2 expression. cancer.
Functions as an immediate-early regulator
of EMT that is downregulated by TGF-β,
Knockdown promotes a mesenchymal-like
affecting the expression of TGF-β-regulated
Lnc-Spry1 [141] phenotype and results in increased cell migration
gene targets; alternative splicing by
and invasion.
U2AF65 splicing factor; isoform switching
Breast cancer of fibroblast growth factor receptors.
Ectopic expression disrupts tight junction Knockdown results in decrease of cell migration,
lncRNA-HIT [148]
by targeting E-cadherin. invasion, tumor growth, and metastasis.
Functions as a sponge of miR-145 and
therefore upregulate the expression of Regulates the cancer stem cell phenotype in
linc-ROR [149,150] ARF6, which regulates adhesion and Triple-negative Breast Cancer, which plays a critical
invasion properties of breast tumor cells role in drug resistance and metastasis.
through E-cadherin.
Functions as an endogenous sponge of
miR-520c-3p, and controls the expression of Knockdown inhibits the progression of breast
HOXA-AS2 [151]
miR-520c-3p target genes, TβR2 and RELA, cancer cells in vitro and in vivo.
in breast cancer cells.
Knockdown causes cell cycle arrested in
G0/G1 phase, promotes cell apoptosis and
Down-regulation inhibits the proliferation,
CCAT2 [152] downregulates the protein expression levels
invasion and migration in breast cancer cells.
of TGF-β, Smad2 and α-SMA in breast
cancer cells.
Regulates the TGF-β pathway genes
through formation of RNA-DNA triplex Ectopic expression inhibits in vivo tumorigenesis
MEG3 [134,153,154] structures, and downregulates AKT and and angiogenesis in a nude mouse xenograft
functions as a sponge of miR-421 to model.
regulate E-cadherin expression.
LINC00978 (known as Knockdown inhibits the activation of Knockdown inhibits the proliferation, migration
MIR4435-2HG and TGF-β/SMAD signaling pathway and EMT and invasion and decreases the in vivo
AK001796) [155] in GC cells. tumorigenicity of GC cells in mice.
Knockdown inhibits TGF-β1-induced-EMT A potential oncogenic factor by regulating GC cells
UCA1 [156] process and the effect could be partly proliferation, invasion, and metastasis under
restored by TGF-β1 treatment. TGF-β1 induction.
Gastric cancer
A novel biomarker of lncRNA, correlated with
Induced by TGF-β and functions as a
lncRNA-ATB [157–159] increased invasion depth, more distant metastasis
ceRNA of miR-141-3p or miR-200s.
and advanced tumor-node-metastasis stage.
Functions as a competing endogenous
XIST [160] lncRNA (ceRNA) to regulate TGF-β1 by sh-XIST inhibited GC development in vitro.
sponging miR-185 in GC.
Int. J. Mol. Sci. 2019, 20, 2767 13 of 34
Table 1. Cont.
Cancer Type LncRNA Function and Mechanism of Action Example of Key Findings or Experiments
Regulates EMT process and the expression Overexpression inhibits cell viability, migration
LncRNA-LET [161] of TIMP2 and activates the Wnt/β-catenin and EMT process, and increases apoptosis in KGN
and Notch signaling pathways. cells
Functions by competing with miR-370-3p to Knockdown suppresses TGF-β-induced EMT,
H19 [162] regulate TGF-β-induced EMT in ovarian while H19 overexpression promotes
cancer. TGF-β-induced EMT.
Interrupts the interaction between
miR-200c/MALAT1 decreases the invasive capacity
TGF-β increases its expression by inhibiting
of EEC cells and EMT in vitro and inhibits EEC
miR-200c; MALAT1 interacts with
growth and EMT-associated protein expression
MALAT1 [163,164] MARCH7, which regulates
in vivo; This LncRNA plays an important role in
TGF-β-smad2/3 pathway by interacting
TβR2-Smad2/3-MALAT1/MARCH7/ATG7 feedback
with TβR2, via miR-200a as a ceRNA.
loop mediated autophagy, migration and invasion
Ovarian in ovarian cancer.
carcinomas Overexpression increases expression of TGF-β1 in
TGF-β1 treatment has no effects on LINK-A ovarian carcinoma cells and promotes cell
LINK-A [165] cervical cancer expression, and there is no clear migration and invasion and this effect can be
mechanism. attenuated by TGF-β1; Plasma levels are correlated
with distant tumor metastasis but not tumor size.
A promising prognostic marker that correlates with
lncRNA-ATB [166] No clear mechanism. the malignant phenotypes and poor prognosis of
cervical cancer
A mediator of TGF-β signaling.
Functions by competing with miR-25 and
Upregulation induces elevated SNAI2, which in
affecting SNAI2 to regulate the expression
PTAF [167] turn promoted OvCa cell EMT and invasion;
of many EMT-related protein-coding genes
knockdown inhibits tumor progression and
in OvCa.
metastasis in an orthotopic mouse model of OvCa.
Knockdown reverses CAF-CM-induced EMT and
Induced by TGF-β, and can regulate EMT
lncRNA-ZEB2NAT [168] invasion of cancer cells, as well as reduces the
process by affecting ZEB2 protein level.
ZEB2 protein level.
Overexpression is significantly correlated with
Induced by TGF-β and regulates EMT by poor survival in patients with bladder cancer.
negatively correlated E-cadherin, Inhibition of malat1 or suz12 suppresses the
MALAT1 [169]
N-cadherin and fibronectin expression by migratory and invasive properties induced by
zeste 12 (suz12) in vitro and in vivo. TGF-β and inhibits tumor metastasis in animal
models.
Its BMP-9-induced expression associates with
increased proliferation and migration of bladder
Induced by BMP9 through phosphorylated cancer cells. The promoting effect of BMP9 is
UCA1 [170]
Bladder cancer AKT and there are no clear mechanism. rescued after interfering with UCA1 in
BMP9 overexpressed bladder cancer cells both
in vitro and in vivo.
Is upregulated by TGF-β, acting as a
Its overexpression significantly promotes cell
lncRNA-ATB [171] molecular sponge of miR-126 and regulate
viability, migration, and invasion in T24 cells.
the direct target of miR-126 (KRAS).
Regulates the cell cycle, cyclin-D1 and EMT
PlncRNA-1 [172] in prostate cancer cells through the Functions as an oncogene.
TGF-β1 pathway.
Knockdown can reverse
High expression is associated with poor prognosis
TGF-β1-induced-EMT phenotype in
ROR [173] in gallbladder cancer patients and knockdown
SGC-996 and Noz cells. However, there are
inhibits cell proliferation, migration, and invasion.
no clear mechanism.
Regulates let-7a/TGF-β1/Smad signaling Overexpression promotes the proliferation and
ANRIL [174]
pathway. migration of prostate cancer cells.
Prostate cancer Upregulated by TGF-β, stimulates EMT Overexpression promotes, and knockdown of
associated with ZEB1 and lncRNA-ATB inhibits the growth of prostate cancer
lncRNA-ATB [175]
ZNF217 expression levels via ERK and cells via regulations of cell cycle regulatory protein
PI3K/AKT signaling pathways. expression levels.
Promotes the progression of pituitary tumors,
Activates the TGFBR2/SMAD3 and
ectopic expression of lnc-SNHG1 promotes cell
RAB11A/Wnt/β-catenin pathways in
lnc-SNHG1 [176] proliferation, migration, and invasion, as well as
pituitary tumor cells via sponging
the EMT, by affecting the cell cycle and cell
miR-302/372/373/520.
apoptosis in vitro and tumor growth in vivo.
Brain tumor
Functions as a ceRNA of miR-1 and
miR-203a to promote Slug expression, Knockdown attenuates EMT and stemness
UCA1 [177]
which underlies TGF-β-induced EMT and processes and their enhancement by TGF-β.
stemness of glioma cells.
Int. J. Mol. Sci. 2019, 20, 2767 14 of 34
Table 1. Cont.
Cancer Type LncRNA Function and Mechanism of Action Example of Key Findings or Experiments
Inhibits TGF-β-induced EMT and thereby
TGF-β1 inhibits its transcription in a
lncRNA-LINP1 [178] controlling cancer cell migration, invasion,
SMAD4-dependent manner.
and stemless in lung cancer cells.
Induced by TGF-β and functions via
Promotes non-small cell lung cancer progression
TBILA [179] cis-regulating HGAL and activating
in vitro and in vivo.
S100A7/JAB1 signaling.
Overexpression inhibits proliferation and
Functions as an endogenous sponge by
TGF-β1-induced EMT in A549 and H1299 cells,
directly binding to miR-137, negatively
XIST [180,181] regulating proliferation and TGF-β1-induced EMT
regulating its expression and regulating
in NSCLC, which could be involved in NSCLC
Notch gene expression.
progression.
Affects the physical interaction of its
binding partner (importin β1) with Smad3, Stimulates TGF-β signaling and regulates
NORAD [182]
and then inhibits the nuclear accumulation TGF-β-induced EMT-like phenotype in A549 cells.
of Smad complexes in response to TGF-β.
Regulates EMT by down-regulating
Overexpression promotes proliferation, migration,
miR-494 in A549 cells, which in turn
lncRNA-ATB [183] and invasion of A549 cells. In contract, ATB silence
increases phosphorylated levels of AKT,
shows the opposite influence.
JAK1, and STAT3.
Functions as a sponge of miR-150-5p and
Inhibition attenuates the tumorigenesis ability of
linc00673 [184] indirectly modulates the expression of key
A549 cells in vivo.
EMT regulator ZEB1.
Expression is regulated by TGF-β and Inhibits migration, invasion and viability of
Lung cancer regulates EMT process by inhibiting the NSCLC cells. Lower NKILA expression are
NKILA [185]
phosphorylation of IκBα and NF-κB correlated with lymph node metastasis and
activation to attenuate Snail expression. advanced TNM stage.
Knockdown inhibits TGF-β-mediated changes in
Associates with JARID2 and the regulatory
cell morphology and cell motility characteristic of
regions of target genes to recruit the
MEG3 [186] EMT and counteracts TGF-β-dependent changes in
complex by epigenetic regulation
the expression of EMT-related genes; In contrast,
(PRC2/JARID2/ H3K27 axis).
overexpression enhances these effects.
ls highly enriched in TGF-β-mediated
exosomes and might function by increasing Knockdown affects lung cancer invasion and
lnc-MMP2-2 [187]
the expression of MMP2 through its vascular permeability.
enhancer activity.
Inhibits NSCLC cell migration and invasion Low expression level indicates shorter
by downregulating TGF-β1 expression, postoperative survival time of NSCLC patients,
however TGF-β1 treatment shows no whereas, ectopic expression inhibits NSCLC cell
ANCR [188]
significant effects on ANCR expression but migration, invasion and downregulated
promotes NSCLC cell migration and TGF-β1 expression, and this effect can be
invasion. attenuated by TGF- β1.
Functions as a mediator of TGF-β signaling,
Knockdown promotes cell migration and invasion,
LINC01186 [189] is down-regulated by TGF-β1 regulating
whereas, overexpression prevents cell metastasis.
EMT by Smad3
Regulates the expression of various
Its expression may determine the overall survival
pathways and genes, especially
HOXA11-AS [190] and disease-free survival of lung adenocarcinoma
DOCK8 and TGF-β pathway, however,
patients in TCGA.
there is no clear mechanism.
Knockdown inhibits the proliferation, migration
Functions as a ceRNA of miR-140-5p and
lncRNA SBF2-AS1 [191] and invasion of HCC cells and attenuate the
regulates the expression of TβR1.
development of HCC tumor in vivo.
Ectopic expression in HCC cells promotes cell
Functions as a ceRNA of miR-26a/b and
proliferation and induces drug resistance, whereas
thereby modulates the expression of
SNHG6-003 [166] knockdown promotes apoptosis. High expression
transforming growth factor-β-activated
of SNHG6-003 closely correlated with tumor
kinase 1 (TAK1).
progression and shorter survival in HCC patients.
Liver cancer
Interacts with KRT19, as a sponge of Knockdown inhibits cell proliferation and invasion
LINC00974 [192] miR-642, activating the Notch and TGF-β with an activation of apoptosis and cell cycle arrest
pathways. both in vitro and in vitro.
Upregulated by TGF-b and can induce EMT
and invasion by acting as ceRNA of
miR-200 family and increasing ZEB1/2; Promotes the invasion-metastasis cascade in
lncRNA-ATB [132]
promotes organ colonization by binding hepatocellular carcinoma.
IL-11 mRNA, autocrine induction of IL-11,
and triggering STAT3 signaling.
Int. J. Mol. Sci. 2019, 20, 2767 15 of 34
Table 1. Cont.
Cancer Type LncRNA Function and Mechanism of Action Example of Key Findings or Experiments
Acts as an oncogene in pancreatic cancer,
Regulates EMT process via TGF-β1/Smad
PVT1 [193] knockdown of PVT1 inhibits viability, adhesion,
signaling.
migration and invasion.
MEG8, which shares the DLK1-DIO3 locus
with MEG3, can induce the recruitment of
EZH2 protein to miR-34a and
miR-203 genes for histone H3 methylation
Plays critical role in TGF-β-induced EMT in
MEG8 [194] and transcriptional repression, inducing
A549 lung cancer and Panc1 pancreatic cancer cells.
EMT-related cell morphological changes
and increases cell motility in the absence of
TGF-β by activating the gene expression
Pancreatic program required for EMT.
cancer Overexpression increases cell proliferation and
Regulates EMT process though
TUG1 [195] migration capacities, enhancing the proliferation
TGF-β/Smad pathway
and migration of pancreatic cancer cells
Induced by TGF-β, and this gene contains
miR-100, miR-125b and let-7a in its intron,
through SMAD2/3. These miRNAs regulate Plays prominent role in metastasis of pancreatic
MIR100HG [196]
a multitude of genes involved in the cancer.
inhibition of p53 and DNA damage
response pathways.
Low expression levels are correlated with lymph
lncRNA-ATB [197] No clear mechanism. node metastases neural invasion, and clinical stage
and worse overall survival prognoses of patients.
Upregulation reduces migration, invasion,
BX357664 [197,198] Blocks the TGF-β1/p38/HSP27 pathway.
and proliferation capabilities in RCC cells.
Its expression is correlated with metastases and
Renal cancer promotes cell migration and invasion in renal cell
carcinoma. Knockdown could inhibit cell
lncRNA-ATB [199] No clear mechanism.
proliferation, trigger apoptosis, reduce
epithelial-to-mesenchymal transition program and
suppress cell migration and invasion.
High expression is significantly associated with
Upregulated by TGF-β, and suppresses
greater tumor size, depth of tumor invasion,
lncRNA-ATB [199–201] E-cadherin expression and promoting EMT
lymphatic invasion, vascular invasion, and lymph
process.
node metastasis.
Colorectal Downregulated by TGF-β and its
cancer expression is positively correlated with
Inhibits EMT and metastasis in colorectal cancer
E-cadherin, and negatively correlated with
LINC01133 [202] (CRC) cells and low LIINC01133 expression in
Vimentin. Directly interacting with SRSF6,
tumors with poor survival in CRC samples.
which promotes EMT and metastasis in
CRC cells.
Represses Notch and TGF-β signaling
pathway by inhibiting Notch1, Hes1, Overexpression represses cell proliferation and
MEG3 [203]
TGF-β and N-cadherin expression, migration ability.
and increasing E-cadherin.
Induced by TGF-β and its overexpression
Overexpression promotes cell metastasis,
decreases E-cadherin level, however, this
Bone tumor MALAT1 [204] a potential diagnostic and prognostic factor in
effect was partially reversed by
osteosarcoma.
EZH2 knockdown.
Inhibits tumor suppressor miR-34s signals
and the targets of miR-34a, including
Knockdown delays the tumor growth in
SNHG7 [205] proliferation-related Notch1,
osteosarcoma tissues.
apoptosis-related BCL-2, cell cycle-related
CDK6, and EMT-related SMAD4.
Induced by TGF-β and supports a role for Functions as an oncogene and as a tumor
MALAT1 [206]
MALAT1 in EMT in thyroid tumors. suppressor in different types of thyroid tumors.
Functions as a competing endogenous RNA Expression is induced by TGF-β in cells
ROR [207]
(ceRNA) of sponging miR-145. undergoing EMT.
A novel prognostic factor, which correlates with
Knockdown increases the levels of
poor prognosis and exhibits that silenced
Thyroid cancer SPRY4-IT1 [208] TGF-β1 and p-Smad2/3 and this effect could
SPRY4-IT1 inhibited the proliferative and
be rescued by the interference of TGF-β1.
migratory abilities of TC cells.
Promotes invasion and metastasis of TC cells,
Reduces p15INK4B expression through
ANRIL [209] and the silencing of ANRIL inhibits the invasion
inhibiting TGF-β/Smad signaling pathway.
and metastasis of TPC-1 cells.
Int. J. Mol. Sci. 2019, 20, 2767 16 of 34
459
460 Figure
Figure 6. TGF-β mediates
6. TGF-β mediates metastasis.
metastasis. TGF-βTGF-βproduced
producedbyby cancer
cancer cells
cells can
can alter
alter the
the bone
bone
461 microenvironment by inducing the expression of osteolytic factors like PTHrP and
microenvironment by inducing the expression of osteolytic factors like PTHrP and IL11. The IL11. The osteoclast
462 bone resorption
osteoclast bone via RANKLvia
resorption production
RANKL by osteoblasts
production byresults in more
osteoblasts release
results in of TGF-β,
more which
release in turn
of TGF-β,
463 acts on tumor cells thereby creating a positive feedback loop called a “vicious cycle”. Chemokine
which in turn acts on tumor cells thereby creating a positive feedback loop called a “vicious cycle”. receptor
464 CXCL4 and receptor
Chemokine angiogenesis
CXCL4 inducer connective tissue
and angiogenesis growth
inducer factor (CTGF)
connective are alsofactor
tissue growth key modulators
(CTGF) are
465 induced
also key modulators induced by TGF-β in this process. Another important factor of this is
by TGF-β in this process. Another important factor of this metastatic preference the Wnt
metastatic
antagonist Dickkopf 1 (DKK1); cells that highly secrete DKK1 tend to metastasize to bone, while low
466 preference is the Wnt antagonist Dickkopf 1 (DKK1); cells that highly secrete DKK1 tend to
DKK1-secreting tumor cells tend to metastasize to the lung. TGF-β leads to transcriptional upregulation
467 metastasize to bone, while low DKK1-secreting tumor cells tend to metastasize to the lung. TGF-β
of proangiogenic factors, including CTGF, matrix metalloprotease (MMP)2, MMP-9, and MMP10,
468 leads to transcriptional upregulation of proangiogenic factors, including CTGF, matrix
or inhibition of TIMP to mediate the formation of new blood vessels. TGF-β inhibits the proliferation
469 metalloprotease (MMP)2, MMP-9, and MMP10, or inhibition of TIMP to mediate the formation of new
of T cells and B cells and inhibits the production of immune factors by B lymphocytes. In addition,
470 blood vessels. TGF-β inhibits the proliferation of T cells and B cells and inhibits the production of
TGF-β-induced angiopoietin-like 4 (ANGPTL4) via the SMAD signaling pathway is proangiogenic and
471 immune factors by B lymphocytes. In addition, TGF-β-induced angiopoietin-like 4 (ANGPTL4) via
can disrupt lung capillary walls and seed pulmonary metastases. Inhibitory signals are indicated with
472 the SMAD signaling pathway is proangiogenic and can disrupt lung capillary walls and seed
inhibitory red arrows; Stimulatory signals are indicated with green arrows.
473 pulmonary metastases. Inhibitory signals are indicated with inhibitory red arrows; Stimulatory
474 signalsPromotes
6.2. TGF-β are indicated with green arrows.
Angiogenesis
475 Regardless
Many specific of the primary
studies haveor secondary
analyzed thetumor,
role of angiogenesis
TGF-β in lungoccurs once
and liver the tumor In
metastases. is amore than
previous
476 1–2 mm in diameter; therefore, a rich blood supply is necessary to
study, increased TGF-β levels were shown to lead to lung metastases in the MMTV/PyVmT provide nutrients and oxygen for
477 tumor growth
transgenic andofmetastasis
model metastatic[224].
breast Tumor
cancercells secrete
[218]. a varietyinofbreast
In addition, growth factors,
cancer including
cells, TGF-β,
TGF-β induced
478 to accelerate the development of cancer by inducing angiogenesis (Figure
ANGPTL4 via the SMAD signaling pathway, and this cytokine could disrupt lung capillary walls 6B) [225].
479 On endothelial
and seed pulmonarycells TGF-β can
metastases [219].bind to the there
Because type Iare
receptor, activin
inherent receptor-like
differences kinase 1 (ALK-1),
in the microvasculature
480 prompting downstream signaling involving intracellular and nuclear
of these two tissues, lung metastasis requires robust extravasation functions provided by proteins (SMADs andANGPTl4
Id1) and
481 leading to a proangiogenic response [226]. Furthermore, TGF-β1 and hypoxia
and other factors and additional lung colonizing functions achieved by ID1/ID3 [220]. Therefore, the are potent inducers
482 of vascular endothelial
vasculature growth factor
disruptive mechanism (VEGF)
provides expression
a selective in tumor
invasive cells, and
advantage oncogenes,
in the lung butespecially
not bone.
483 RAS,
Another study showed that the WNT signaling inhibitor Dickkopf 1 (DKK1) is a key for
can also combine with the tumor microenvironment, providing the foundation factortumor cell
for this
484 invasion and angiogenesis [227]. Using a mouse mammary carcinoma model,
metastatic preference; it reduces the recruitment of macrophages and neutrophils by the WNT/PCP- researchers confirmed
485 that VEGF expression
RAC1-JNK pathway andin peri-necrotic
inhibits theareas
levelisofsynergized
tumor-derivedby both hypoxia
TGF-β and TGF-β1,
to inhibit further showing
lung metastasis. Thus,
486 that
tumor cells that highly secrete DKK1 tend to metastasize to bone, while those with associating
this cooperation is achieved through hypoxia-inducible factor (HIF)-1α physically low DKK1
487 with SMAD3
secretion tend[228]. For example,
to metastasize to theTGF-β increases the expression of VEGF-C by coordinating with
lung [221].
488 sine oculis
Statistical analysis of gene expressiontumor
homeobox homolog 1 (SIX1) in cells,revealed
profiles promoting thattumor
TGF-β lymph angiogenesis
signaling is the mostand
489 significant gene pathway in liver metastases of colorectal cancer [222]. Exosomes (small membrane
490 vesicles with a size ranging from 40 to 100 nm) from pancreatic cancer induce Kupffer cells to release
Int. J. Mol. Sci. 2019, 20, 2767 18 of 34
lymph node metastasis [229]. Likewise, TGF-β1 could promote macrophages to secrete more VEGF
via the TβRII/SMAD3 signaling pathway in oral squamous cell carcinoma [230]. TGF-β and VEGF
form a feedback loop through the Semaphorin3A/NEM axis; in general, abrogated VEGF inhibits
the endothelial cell paracrine TGF-β1 and endothelial SMAD2/3 activation; in turn, TGF-β1 further
stimulates endothelial Semaphorin3A expression [231]. Studies have shown that in RAS-transformed
epithelial tumors, TGF-β significantly increases the expression of VEGF/VEGF-R, which has a powerful
effect on capillary formation and migration of endothelial cells, thereby promoting angiogenesis in
tumor cells [232]. TGF-β mediates the formation of new blood vessels by promoting connective tissue
growth factor (CTGF) and angiogenic regulatory enzymes, such as matrix metalloproteinases (MMP-2,
MMP-9, MMP-10, etc.) or by inhibiting tissue inhibitor of metalloproteinases (TIMP) [233].
602
Figure 7. Targeting the TGF-β signaling pathway in cancer. Various molecular mechanisms by
603 Figure TGF-β
which 7. Targeting the TGF-β
signaling signaling
is targeted for pathway
therapeuticin cancer. Various
gain are molecular
depicted. TGF-βmechanisms by which
inhibitory targeting
604 TGF-β signaling is targeted for therapeutic gain are depicted. TGF-β inhibitory targeting agents
agents (Table 1), including TβRI kinase inhibitors, AON targeting ligand or receptor gene expression,(Table 1),
605 including TβRI kinase inhibitors, AON targeting ligand or receptor gene expression, and antibodies
and antibodies interfering with ligand-receptor interactions, have been developed to curtail excessive
606 interfering
TGF-β with activation.
pathway ligand-receptor interactions,
Inhibitory have
signals are been with
indicated developed to curtail
inhibitory excessive
red arrows; TGF-β
Stimulatory
607 pathway
signals areactivation.
indicated Inhibitory
with greensignals
arrows.are indicated with inhibitory red arrows; Stimulatory signals
608 are indicated with green arrows.
8. Concluding Remarks
609 8. Concluding Remarks
TGF-β acts as a tumor suppressor during the early phase of cancer progression and as a tumor
610 TGF-β
promotor in acts as a tumor
advanced suppressor
stages. The latter during
role, in the early phase
particular, of cancer
has been progression
targeted and therapeutic
as a potential as a tumor
611 promotor toininhibiting
approach advancedtumorstages. The and
growth latterdissemination.
role, in particular,
However,hasdeveloping
been targeted as atreatments
effective potential
612 therapeutic
is approach by
severely hampered to inhibiting tumor
the biphasic role growth
of TGF-β andin dissemination. However,
cancer and its function indeveloping effective
many physiological
613 treatments including
processes, is severelythehampered by the system.
cardiovascular biphasicThere
role ofhas
TGF-β
beenin cancer
some and itsinfunction
progress in many
the treatment of
614 physiological processes, including the cardiovascular system. There has been some progress
liver cancer and other types of cancer, but there is a pressing need for a greater understanding of in the
615 treatment ofTGF-β
pathological liver mechanisms
cancer and other
as welltypes of cancer,
as greater but there protocols
clarity regarding is a pressing needadministration
of therapy for a greater
616 understanding
and of pathological TGF-β mechanisms as well as greater clarity regarding protocols of
patient selection.
617 therapy
Withadministration
the success of and patient
immune selection.inhibitors for cancer therapy and considering the potent
checkpoint
immune suppressive effects of TGF-β, it may be of particular interest to see whether TGF-β targeting
agents can increase the efficiency and range of immune therapeutic agents. Such trials are underway,
and the results are eagerly awaited. As highlighted in this review, other approaches could include the
targeting of E3 ubiquitin ligases and deubiquitinating enzymes and miRNAs and lncRNAs, which
control the stability of TGF-β receptors and SMAD proteins.
Author Contributions: Writing—original draft preparation, Y.H.; writing—review and editing, D.B.; supervision,
P.t.D.
Funding: Our studies on TGF-β signaling in cancer are supported by Cancer Genomics Centre Netherlands
(CGC.nl). YH is supported by CSC scholarship.
Int. J. Mol. Sci. 2019, 20, 2767 21 of 34
Acknowledgments: Our studies on TGF-β signaling in cancer are supported by Cancer Genomics Centre
Netherlands (CGC.nl). YH is supported by CSC scholarship.
Conflicts of Interest: The authors declare no conflict of interest.
References
1. Faguet, G.B. A brief history of cancer: Age-old milestones underlying our current knowledge database.
Int. J. Cancer 2015, 136, 2022–2036. [CrossRef] [PubMed]
2. Hu, Y.; Fu, L. Targeting cancer stem cells: A new therapy to cure cancer patients. Am. J. Cancer Res. 2012, 2,
340–356. [PubMed]
3. Farkona, S.; Diamandis, E.P.; Blasutig, I.M. Cancer immunotherapy: The beginning of the end of cancer?
BMC Med. 2016, 14, 73. [CrossRef] [PubMed]
4. David, C.J.; Massague, J. Contextual determinants of TGF-β action in development, immunity and cancer.
Nat. Rev. Mol. Cell Biol. 2018, 19, 419–435. [CrossRef] [PubMed]
5. Exposito-Villen, A.; Aranega, E.A.; Franco, D. Functional role of non-coding RNAs during
epithelial-To-mesenchymal transition. Noncoding RNA 2018, 4, 14. [CrossRef]
6. Derynck, R.; Budi, E.H. Specificity, versatility, and control of TGF-β family signaling. Sci. Signal 2019, 12,
eaav5183. [CrossRef] [PubMed]
7. De Larco, J.E.; Todaro, G.J. Growth factors from murine sarcoma virus-transformed cells. Proc. Natl. Acad.
Sci. USA 1978, 75, 4001–4005. [CrossRef]
8. Derynck, R.; Jarrett, J.A.; Chen, E.Y.; Eaton, D.H.; Bell, J.R.; Assoian, R.K.; Roberts, A.B.; Sporn, M.B.;
Goeddel, V.D. Human transforming growth factor-β complementary DNA sequence and expression in
normal and transformed cells. Nature 1985, 316, 701–705. [CrossRef]
9. Sha, X.; Yang, L.; Gentry, E.L. Identification and analysis of discrete functional domains in the pro region of
pre-pro-transforming growth factor β 1. J. Cell Biol. 1991, 114, 827–839. [CrossRef]
10. Shi, M.; Zhu, J.; Wang, R.; Chen, X.; Mi, L.; Walz, T.; Springer, T.A. Latent TGF-β structure and activation.
Nature 2011, 474, 343–349. [CrossRef]
11. Cheifetz, S.; Hernandez, H.; Laiho, M.; ten Dijke, P.; Iwata, K.K.; Massague, J. Distinct transforming growth
factor-β (TGF-β) receptor subsets as determinants of cellular responsiveness to three TGF-β isoforms.
J. Biol. Chem. 1990, 265, 20533–20538. [PubMed]
12. Dong, X.; Zhao, B.; Iacob, R.E.; Zhu, J.; Koksal, A.C.; Lu, C.; Engen, J.R.; Springer, T.A. Force interacts with
macromolecular structure in activation of TGF-β. Nature 2017, 542, 55–59. [CrossRef] [PubMed]
13. Massague, J. Receptors for the TGF-β family. Cell 1992, 69, 1067–1070. [CrossRef]
14. Heldin, C.H.; Moustakas, A. Signaling receptors for TGF-β family members. Cold Spring Harb. Perspect. Biol.
2016, 8, a022053. [CrossRef] [PubMed]
15. Upadhyay, A.; Moss-Taylor, L.; Kim, M.J.; Ghosh, A.C.; O’Connor, M.B. TGF-β family signaling in drosophila.
Cold Spring Harb. Perspect. Biol. 2017, 9. [CrossRef] [PubMed]
16. Savage-Dunn, C.; Padgett, R.W. The TGF-β family in caenorhabditis elegans. Cold Spring Harb. Perspect. Biol.
2017, 9, a022178. [CrossRef] [PubMed]
17. Derynck, R.; Gelbart, W.M.; Harland, R.M.; Heldin, C.H.; Kern, S.E.; Massagué, J.; Melton, D.A.; Mlodzik, M.;
Padgett, R.W.; Roberts, A.B.; et al. Nomenclature: Vertebrate mediators of TGF-β family signals. Cell 1996,
87, 173. [CrossRef]
18. Wrana, J.L. Signaling by the TGF-β superfamily. Cold Spring Harb. Perspect. Biol. 2013, 5, a011197. [CrossRef]
[PubMed]
19. Hill, C.S. Transcriptional control by the SMADs. Cold Spring Harb. Perspect. Biol. 2016, 8, a022079. [CrossRef]
20. Miyazawa, K.; Miyazono, K. Regulation of TGF-β family signaling by inhibitory SMADs. Cold Spring Harb.
Perspect. Biol. 2017, 9, a022095. [CrossRef]
21. Zhang, Y.E. Non-SMAD signaling pathways of the TGF-β family. Cold Spring Harb. Perspect. Biol. 2017, 9,
a022129. [CrossRef] [PubMed]
22. Zhang, Y.E. Non-SMAD pathways in TGF-β signaling. Cell Res. 2009, 19, 128–139. [CrossRef] [PubMed]
23. Ravichandran, K.S. Signaling via Shc family adapter proteins. Oncogene 2001, 20, 6322–6330. [CrossRef]
[PubMed]
Int. J. Mol. Sci. 2019, 20, 2767 22 of 34
24. Yamashita, M.; Fatyol, K.; Jin, C.; Wang, X.; Liu, Z.; Zhang, Y.E. TRAF6 mediates SMAD-independent
activation of JNK and p38 by TGF-β. Mol. Cell. 2008, 31, 918–924. [CrossRef] [PubMed]
25. Sorrentino, A.; Thakur, N.; Grimsby, S.; Marcusson, A.; von Bulow, V.; Schuster, N.; Zhang, S.; Heldin, C.H.;
Landstrom, M. The type I TGF-β receptor engages TRAF6 to activate TAK1 in a receptor kinase-independent
manner. Nat. Cell Biol. 2008, 10, 1199–1207. [CrossRef] [PubMed]
26. Ozdamar, B.; Bose, R.; Barrios-Rodiles, M.; Wang, H.R.; Zhang, Y.; Wrana, J.L. Regulation of the polarity
protein Par6 by TGF-β receptors controls epithelial cell plasticity. Science 2005, 307, 1603–1609. [CrossRef]
[PubMed]
27. Wilkes, M.C.; Murphy, S.J.; Garamszegi, N.; Leof, E.B. Cell-type-specific activation of PAK2 by transforming
growth factor β independent of SMAD2 and SMAD3. Mol. Cell Biol. 2003, 23, 8878–8889. [CrossRef]
28. Bakin, A.V.; Tomlinson, A.K.; Bhowmick, N.A.; Moses, H.L.; Arteaga, C.L. Phosphatidylinositol 3-kinase
function is required for transforming growth factor β-mediated epithelial to mesenchymal transition and
cell migration. J. Biol. Chem. 2000, 275, 36803–36810. [CrossRef]
29. Zhang, Y.; Alexander, P.B.; Wang, X.F. TGF-β family signaling in the control of cell proliferation and survival.
Cold Spring Harb. Perspect. Biol. 2017, 9, a022145. [CrossRef]
30. Azar, R.; Alard, A.; Susini, C.; Bousquet, C.; Pyronnet, S. 4E-BP1 is a target of SMAD4 essential for
TGF-β-mediated inhibition of cell proliferation. EMBO J. 2009, 28, 3514–3522. [CrossRef]
31. Baghdassarian, N.; Ffrench, M. Cyclin-dependent kinase inhibitors (CKIs) and hematological malignancies.
Hematol. Cell Ther. 1996, 38, 313–323. [CrossRef] [PubMed]
32. Xu, J.; Acharya, S.; Sahin, O.; Zhang, Q.; Saito, Y.; Yao, J.; Wang, H.; Li, P.; Zhang, L.; Lowery, F.J.; et al.
14-3-3zeta turns TGF-β’s function from tumor suppressor to metastasis promoter in breast cancer by
contextual changes of SMAD partners from p53 to Gli2. Cancer Cell. 2015, 27, 177–192. [CrossRef] [PubMed]
33. Iavarone, A.; Massague, J. Repression of the CDK activator Cdc25A and cell-cycle arrest by cytokine TGF-β
in cells lacking the CDK inhibitor p15. Nature 1997, 387, 417–422. [CrossRef] [PubMed]
34. Ling, M.T.; Wang, X.; Tsao, S.W.; Wong, Y.C. Down-regulation of Id-1 expression is associated with
TGF-β1-induced growth arrest in prostate epithelial cells. Biochim. Biophys. Acta 2002, 1570, 145–152.
[CrossRef]
35. Chen, C.R.; Kang, Y.; Siegel, P.M.; Massague, J. E2F4/5 and p107 as SMAD cofactors linking the TGF-β
receptor to c-myc repression. Cell 2002, 110, 19–32. [CrossRef]
36. Ozaki, I.; Hamajima, H.; Matsuhashi, S.; Mizuta, T. Regulation of TGF-β1-induced pro-apoptotic signaling
by growth factor receptors and extracellular matrix receptor integrins in the liver. Front. Physiol. 2011, 2, 78.
[CrossRef] [PubMed]
37. Wiener, Z.; Band, A.M.; Kallio, P.; Hogstrom, J.; Hyvonen, V.; Kaijalainen, S.; Ritvos, O.; Haglund, C.;
Kruuna, O.; Robine, S.; et al. Oncogenic mutations in intestinal adenomas regulate Bim-mediated apoptosis
induced by TGF-β. Proc. Natl. Acad. Sci. USA 2014, 111, E2229–E2236. [CrossRef]
38. Zhao, X.; Liu, Y.; Du, L.; He, L.; Ni, B.; Hu, J.; Zhu, D.; Chen, Q. Threonine 32 (Thr32) of FoxO3 is critical for
TGF-β-induced apoptosis via Bim in hepatocarcinoma cells. Protein Cell 2015, 6, 127–138. [CrossRef]
39. Spender, L.C.; O’Brien, D.I.; Simpson, D.; Dutt, D.; Gregory, C.D.; Allday, M.J.; Clark, L.J.; Inman, G.J. TGF-β
induces apoptosis in human B cells by transcriptional regulation of BIK and BCL-XL. Cell Death Differ. 2009,
16, 593–602. [CrossRef]
40. Ohgushi, M.; Kuroki, S.; Fukamachi, H.; O’Reilly, L.A.; Kuida, K.; Strasser, A.; Yonehara, S.
Transforming growth factor β-dependent sequential activation of SMAD, Bim, and caspase-9 mediates
physiological apoptosis in gastric epithelial cells. Mol. Cell. Biol. 2005, 25, 10017–10028. [CrossRef]
41. Shima, Y.; Nakao, K.; Nakashima, T.; Kawakami, A.; Nakata, K.; Hamasaki, K.; Kato, Y.; Eguchi, K.; Ishii, N.
Activation of caspase-8 in transforming growth factor-β-induced apoptosis of human hepatoma cells.
Hepatology 1999, 30, 1215–1222. [CrossRef] [PubMed]
42. Takekawa, M.; Tatebayashi, K.; Itoh, F.; Adachi, M.; Imai, K.; Saito, H. SMAD-dependent GADD45β
expression mediates delayed activation of p38 MAP kinase by TGF-β. EMBO J. 2002, 21, 6473–6482.
[CrossRef] [PubMed]
43. Yan, F.; Wang, Y.; Wu, X.; Peshavariya, H.M.; Dusting, G.J.; Zhang, M.; Jiang, F. Nox4 and redox signaling
mediate TGF-β-induced endothelial cell apoptosis and phenotypic switch. Cell Death Dis. 2014, 5, e1010.
[CrossRef] [PubMed]
Int. J. Mol. Sci. 2019, 20, 2767 23 of 34
44. Cardenas, H.; Vieth, E.; Lee, J.; Segar, M.; Liu, Y.; Nephew, K.P.; Matei, D. TGF-β induces global changes in
DNA methylation during the epithelial-to-mesenchymal transition in ovarian cancer cells. Epigenetics 2014,
9, 1461–1472. [CrossRef] [PubMed]
45. Evanno, E.; Godet, J.; Piccirilli, N.; Guilhot, J.; Milin, S.; Gombert, J.M.; Fouchaq, B.; Roche, J. Tri-methylation of
H3K79 is decreased in TGF-β1-induced epithelial-to-mesenchymal transition in lung cancer. Clin. Epigenetics
2017, 9, 80. [CrossRef] [PubMed]
46. Cassar, L.; Nicholls, C.; Pinto, A.R.; Chen, R.; Wang, L.; Li, H.; Liu, J.P. TGF-β receptor mediated telomerase
inhibition, telomere shortening and breast cancer cell senescence. Protein Cell 2017, 8, 39–54. [CrossRef]
[PubMed]
47. Kiyono, K.; Suzuki, H.I.; Matsuyama, H.; Morishita, Y.; Komuro, A.; Kano, M.R.; Sugimoto, K.; Miyazono, K.
Autophagy is activated by TGF-β and potentiates TGF-β-mediated growth inhibition in human hepatocellular
carcinoma cells. Cancer Res. 2009, 69, 8844–8852. [CrossRef]
48. Suzuki, H.I.; Kiyono, K.; Miyazono, K. Regulation of autophagy by transforming growth factor-β (TGF-β)
signaling. Autophagy 2010, 6, 645–647. [CrossRef]
49. Korkut, A.; Zaidi, S.; Kanchi, R.S.; Rao, S.; Gough, N.R.; Schultz, A.; Li, X.; Lorenzi, P.L.; Berger, A.C.;
Robertson, G.; et al. A pan-cancer analysis reveals high-frequency genetic alterations in mediators of
signaling by the TGF-β superfamily. Cell Syst. 2018, 7, 422–437.e7. [CrossRef]
50. Hahn, S.A.; Schutte, M.; Hoque, A.T.; Moskaluk, C.A.; da Costa, L.T.; Rozenblum, E.; Weinstein, C.L.;
Fischer, A.; Yeo, C.J.; Hruban, R.H.; et al. DPC4, a candidate tumor suppressor gene at human chromosome
18q21.1. Science 1996, 271, 350–353. [CrossRef]
51. Markowitz, S.; Wang, J.; Myeroff, L.; Parsons, R.; Sun, L.; Lutterbaugh, J.; Fan, R.S.; Zborowska, E.;
Kinzler, K.W.; Vogelstein, B.; et al. Inactivation of the type II TGF-β receptor in colon cancer cells with
microsatellite instability. Science 1995, 268, 1336–1338. [CrossRef] [PubMed]
52. Macias-Silva, M.; Abdollah, S.; Hoodless, P.A.; Pirone, R.; Attisano, L.; Wrana, J.L. MADR2 is a substrate of
the TGF-β receptor and its phosphorylation is required for nuclear accumulation and signaling. Cell 1996, 87,
1215–1224. [CrossRef]
53. Yang, L.; Pang, Y.; Moses, H.L. TGF-β and immune cells: An important regulatory axis in the tumor
microenvironment and progression. Trends Immunol. 2010, 31, 220–227. [CrossRef] [PubMed]
54. Pinto, M.; Oliveira, C.; Cirnes, L.; Machado, J.C.; Ramires, M.; Nogueira, A.; Carneiro, F.; Seruca, R.
Promoter methylation of TGF-β receptor I and mutation of TGF-β receptor II are frequent events in MSI
sporadic gastric carcinomas. J. Pathol. 2003, 200, 32–38. [CrossRef]
55. Bruna, A.; Darken, R.S.; Rojo, F.; Ocana, A.; Penuelas, S.; Arias, A.; Paris, R.; Tortosa, A.; Mora, J.; Baselga, J.;
et al. High TGF-β-SMAD activity confers poor prognosis in glioma patients and promotes cell proliferation
depending on the methylation of the PDGF-B gene. Cancer Cell 2007, 11, 147–160. [CrossRef] [PubMed]
56. Caja, L.; Dituri, F.; Mancarella, S.; Caballero-Diaz, D.; Moustakas, A.; Giannelli, G.; Fabregat, I. TGF-β and
the tissue microenvironment: Relevance in fibrosis and cancer. Int. J. Mol. Sci. 2018, 19, 1294. [CrossRef]
[PubMed]
57. Peinado, H.; Quintanilla, M.; Cano, A. Transforming growth factor β-1 induces snail transcription factor in
epithelial cell lines: Mechanisms for epithelial mesenchymal transitions. J. Biol. Chem. 2003, 278, 21113–21123.
[CrossRef]
58. Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial-mesenchymal transition. Nat. Rev. Mol.
Cell Biol. 2014, 15, 178–196. [CrossRef]
59. Nieto, M.A.; Huang, R.Y.; Jackson, R.A.; Thiery, J.P. Emt: 2016. Cell 2016, 166, 21–45. [CrossRef]
60. Tsai, J.H.; Yang, J. Epithelial-mesenchymal plasticity in carcinoma metastasis. Genes Dev. 2013, 27, 2192–2206.
[CrossRef]
61. van Staalduinen, J.; Baker, D.; Dijke, P.t.; van Dam, H. Epithelial-mesenchymal-transition-inducing
transcription factors: New targets for tackling chemoresistance in cancer? Oncogene 2018, 37, 6195–6211.
[CrossRef] [PubMed]
62. Jolly, M.K.; Ware, K.E.; Gilja, S.; Somarelli, J.A.; Levine, H. EMT and MET: Necessary or permissive for
metastasis? Mol. Oncol. 2017, 11, 755–769. [CrossRef] [PubMed]
63. Zheng, X.; Carstens, J.L.; Kim, J.; Scheible, M.; Kaye, J.; Sugimoto, H.; Wu, C.C.; LeBleu, V.S.; Kalluri, R.
Epithelial-to-mesenchymal transition is dispensable for metastasis but induces chemoresistance in pancreatic
cancer. Nature 2015, 527, 525–530. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2019, 20, 2767 24 of 34
64. Fischer, K.R.; Durrans, A.; Lee, S.; Sheng, J.; Li, F.; Wong, S.T.; Choi, H.; el Rayes, T.; Ryu, S.; Troeger, J.; et al.
Epithelial-to-mesenchymal transition is not required for lung metastasis but contributes to chemoresistance.
Nature 2015, 527, 472–476. [CrossRef] [PubMed]
65. Aiello, N.M.; Brabletz, T.; Kang, Y.; Nieto, M.A.; Weinberg, R.A.; Stanger, B.Z. Upholding a role for EMT in
pancreatic cancer metastasis. Nature 2017, 547, E7–E8. [CrossRef] [PubMed]
66. Li, W.; Kang, Y. Probing the fifty shades of EMT in metastasis. Trends Cancer 2016, 2, 65–67. [CrossRef]
[PubMed]
67. Pastushenko, I.; Brisebarre, A.; Sifrim, A.; Fioramonti, M.; Revenco, T.; Boumahdi, S.; van Keymeulen, A.;
Brown, D.; Moers, V.; Lemaire, S.; et al. Identification of the tumour transition states occurring during EMT.
Nature 2018, 556, 463–468. [CrossRef]
68. Nikitorowicz-Buniak, J.; Denton, C.P.; Abraham, D.; Stratton, R. Partially evoked epithelial-mesenchymal
transition (EMT) is associated with increased TGF-β signaling within lesional scleroderma skin. PLoS ONE
2015, 10, e0134092.
69. Puram, S.V.; Parikh, A.S.; Tirosh, I. Single cell RNA-seq highlights a role for a partial EMT in head and neck
cancer. Mol. Cell Oncol. 2018, 5, e1448244. [CrossRef]
70. Deckers, M.; van Dinther, M.; Buijs, J.; Que, I.; Lowik, C.; van der Pluijm, G.; Dijke, P.t. The tumor suppressor
SMAD4 is required for transforming growth factor β-induced epithelial to mesenchymal transition and bone
metastasis of breast cancer cells. Cancer Res. 2006, 66, 2202–2209. [CrossRef]
71. Vincent, T.; Neve, E.P.; Johnson, J.R.; Kukalev, A.; Rojo, F.; Albanell, J.; Pietras, K.; Virtanen, I.; Philipson, L.;
Leopold, P.L.; et al. A SNAIL1-SMAD3/4 transcriptional repressor complex promotes TGF-β mediated
epithelial-mesenchymal transition. Nat. Cell Biol. 2009, 11, 943–950. [CrossRef] [PubMed]
72. Hoot, K.E.; Lighthall, J.; Han, G.; Lu, S.L.; Li, A.; Ju, W.; Kulesz-Martin, M.; Bottinger, E.; Wang, X.J.
Keratinocyte-specific SMAD2 ablation results in increased epithelial-mesenchymal transition during skin
cancer formation and progression. J. Clin. Investig. 2008, 118, 2722–2732. [CrossRef] [PubMed]
73. Yu, J.; Lei, R.; Zhuang, X.; Li, X.; Li, G.; Lev, S.; Segura, M.F.; Zhang, X.; Hu, G. MicroRNA-182 targets
SMAD7 to potentiate TGF-β-induced epithelial-mesenchymal transition and metastasis of cancer cells.
Nat. Commun. 2016, 7, 13884. [CrossRef] [PubMed]
74. Kuang, J.; Li, L.; Guo, L.; Su, Y.; Wang, Y.; Xu, Y.; Wang, X.; Meng, S.; Lei, L.; Xu, L.; et al. RNF8 promotes
epithelial-mesenchymal transition of breast cancer cells. J. Exp. Clin. Cancer Res. 2016, 35, 88. [CrossRef]
[PubMed]
75. Zavadil, J.; Bitzer, M.; Liang, D.; Yang, Y.C.; Massimi, A.; Kneitz, S.; Piek, E.; Bottinger, E.P. Genetic programs
of epithelial cell plasticity directed by transforming growth factor-β. Proc. Natl. Acad. Sci. USA 2001, 98,
6686–6691. [CrossRef] [PubMed]
76. Lee, M.K.; Pardoux, C.; Hall, M.C.; Lee, P.S.; Warburton, D.; Qing, J.; Smith, S.M.; Derynck, R. TGF-β activates
Erk MAP kinase signalling through direct phosphorylation of ShcA. EMBO J. 2007, 26, 3957–3967. [CrossRef]
[PubMed]
77. Davies, M.; Robinson, M.; Smith, E.; Huntley, S.; Prime, S.; Paterson, I. Induction of an epithelial to
mesenchymal transition in human immortal and malignant keratinocytes by TGF-β1 involves MAPK, SMAD
and AP-1 signalling pathways. J. Cell Biochem. 2005, 95, 918–931. [CrossRef] [PubMed]
78. Lin, T.; Zeng, L.; Liu, Y.; DeFea, K.; Schwartz, M.A.; Chien, S.; Shyy, J.Y. Rho-ROCK-LIMK-cofilin pathway
regulates shear stress activation of sterol regulatory element binding proteins. Circ. Res. 2003, 92, 1296–1304.
[CrossRef] [PubMed]
79. Landstrom, M. The TAK1-TRAF6 signalling pathway. Int. J. Biochem. Cell Biol. 2010, 42, 585–589. [CrossRef]
80. Song, J.; Landstrom, M. TGF-β activates PI3K-AKT signaling via TRAF6. Oncotarget 2017, 8, 99205–99206.
[CrossRef]
81. Xue, G.; Restuccia, D.F.; Lan, Q.; Hynx, D.; Dirnhofer, S.; Hess, D.; Ruegg, C.; Hemmings, B.A.
Akt/PKB-mediated phosphorylation of TWIST1 promotes tumor metastasis via mediating cross-talk between
PI3K/Akt and TGF-β signaling axes. Cancer Dis. 2012, 2, 248–259. [CrossRef] [PubMed]
82. Ma, L.; Teruya-Feldstein, J.; Weinberg, R.A. Tumour invasion and metastasis initiated by microRNA-10b in
breast cancer. Nature 2007, 449, 682–688. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2019, 20, 2767 25 of 34
83. Werden, S.J.; Sphyris, N.; Sarkar, T.R.; Paranjape, A.N.; LaBaff, A.M.; Taube, J.H.; Hollier, B.G.;
Ramirez-Pena, E.Q.; Soundararajan, R.; den Hollander, P.; et al. Phosphorylation of serine 367 of FOXC2 by
p38 regulates ZEB1 and breast cancer metastasis, without impacting primary tumor growth. Oncogene 2016,
35, 5977–5988. [CrossRef] [PubMed]
84. Chen, L.; Li, Y.C.; Wu, L.; Yu, G.T.; Zhang, W.F.; Huang, C.F.; Sun, Z.J. TRAF6 regulates tumour metastasis
through EMT and CSC phenotypes in head and neck squamous cell carcinoma. J. Cell. Mol. Med. 2018, 22,
1337–1349. [CrossRef] [PubMed]
85. Gonzalez, D.M.; Medici, D. Signaling mechanisms of the epithelial-mesenchymal transition. Sci. Signal 2014,
7, re8. [CrossRef] [PubMed]
86. Nishita, M.; Hashimoto, M.K.; Ogata, S.; Laurent, M.N.; Ueno, N.; Shibuya, H.; Cho, K.W. Interaction between
Wnt and TGF-β signalling pathways during formation of Spemann’s organizer. Nature 2000, 403, 781–785.
[CrossRef] [PubMed]
87. Medici, D.; Hay, E.D.; Goodenough, D.A. Cooperation between snail and LEF-1 transcription factors is
essential for TGF-β1-induced epithelial-mesenchymal transition. Mol. Biol. Cell. 2006, 17, 1871–1879.
[CrossRef] [PubMed]
88. Jamora, C.; Lee, P.; Kocieniewski, P.; Azhar, M.; Hosokawa, R.; Chai, Y.; Fuchs, E. A signaling pathway
involving TGF-β2 and snail in hair follicle morphogenesis. PLoS Biol. 2005, 3, e11. [CrossRef]
89. Murillo-Garzon, V.; Gorrono-Etxebarria, I.; Akerfelt, M.; Puustinen, M.C.; Sistonen, L.; Nees, M.; Carton, J.;
Waxman, J.; Kypta, R.M. Frizzled-8 integrates Wnt-11 and transforming growth factor-β signaling in prostate
cancer. Nat. Commun. 2018, 9, 1747. [CrossRef]
90. Kanei-Ishii, C.; Ninomiya-Tsuji, J.; Tanikawa, J.; Nomura, T.; Ishitani, T.; Kishida, S.; Kokura, K.; Kurahashi, T.;
Ichikawa-Iwata, E.; Kim, Y.; et al. Wnt-1 signal induces phosphorylation and degradation of c-Myb protein
via TAK1, HIPK2, and NLK. Genes Dev. 2004, 18, 816–829. [CrossRef]
91. Sjolund, J.; Bostrom, A.K.; Lindgren, D.; Manna, S.; Moustakas, A.; Ljungberg, B.; Johansson, M.; Fredlund, E.;
Axelson, H. The notch and TGF-β signaling pathways contribute to the aggressiveness of clear cell renal cell
carcinoma. PLoS ONE 2011, 6, e23057. [CrossRef] [PubMed]
92. Zavadil, J.; Cermak, L.; Soto-Nieves, N.; Bottinger, E.P. Integration of TGF-β/SMAD and Jagged1/Notch
signalling in epithelial-to-mesenchymal transition. EMBO J. 2004, 23, 1155–1165. [CrossRef] [PubMed]
93. Zhang, Y.; Feng, X.H.; Derynck, R. SMAD3 and SMAD4 cooperate with c-Jun/c-Fos to mediate TGF-β-induced
transcription. Nature 1998, 394, 909–913. [CrossRef] [PubMed]
94. Huber, M.A.; Kraut, N.; Beug, H. Molecular requirements for epithelial-mesenchymal transition during
tumor progression. Curr. Opin. Cell Biol. 2005, 17, 548–558. [CrossRef] [PubMed]
95. Olsen, S.N.; Wronski, A.; Castano, Z.; Dake, B.; Malone, C.; de Raedt, T.; Enos, M.; DeRose, Y.S.; Zhou, W.;
Guerra, S.; et al. Loss of RasGAP tumor suppressors underlies the aggressive nature of luminal b breast
cancers. Cancer Discov. 2017, 7, 202–217. [CrossRef] [PubMed]
96. Bowman, T.; Broome, M.A.; Sinibaldi, D.; Wharton, W.; Pledger, W.J.; Sedivy, J.M.; Irby, R.; Yeatman, T.;
Courtneidge, S.A.; Jove, R. Stat3-mediated Myc expression is required for Src transformation and
PDGF-induced mitogenesis. Proc. Natl. Acad. Sci. USA 2001, 98, 7319–7324. [CrossRef] [PubMed]
97. Lamouille, S.; Subramanyam, D.; Blelloch, R.; Derynck, R. Regulation of epithelial-mesenchymal and
mesenchymal-epithelial transitions by microRNAs. Curr. Opin. Cell Biol. 2013, 25, 200–207. [CrossRef]
[PubMed]
98. Gregory, P.A.; Bert, A.G.; Paterson, E.L.; Barry, S.C.; Tsykin, A.; Farshid, G.; Vadas, M.A.; Khew-Goodall, Y.;
Goodall, G.J. The miR-200 family and miR-205 regulate epithelial to mesenchymal transition by targeting
ZEB1 and SIP1. Nat. Cell Biol. 2008, 10, 593–601. [CrossRef]
99. Diepenbruck, M.; Tiede, S.; Saxena, M.; Ivanek, R.; Kalathur, R.K.R.; Luond, F.; Meyer-Schaller, N.;
Christofori, G. miR-1199-5p and Zeb1 function in a double-negative feedback loop potentially coordinating
EMT and tumour metastasis. Nat. Commun. 2017, 8, 1168. [CrossRef]
100. Kim, N.H.; Kim, H.S.; Li, X.Y.; Lee, I.; Choi, H.S.; Kang, S.E.; Cha, S.Y.; Ryu, J.K.; Yoon, D.; Fearon, E.R.; et al.
A p53/miRNA-34 axis regulates Snail1-dependent cancer cell epithelial-mesenchymal transition. J. Cell. Biol.
2011, 195, 417–433. [CrossRef]
101. Hahn, S.; Jackstadt, R.; Siemens, H.; Hunten, S.; Hermeking, H. SNAIL and miR-34a feed-forward regulation
of ZNF281/ZBP99 promotes epithelial-mesenchymal transition. EMBO J. 2013, 32, 3079–3095. [CrossRef]
[PubMed]
Int. J. Mol. Sci. 2019, 20, 2767 26 of 34
102. Majid, S.; Dar, A.A.; Saini, S.; Shahryari, V.; Arora, S.; Zaman, M.S.; Chang, I.; Yamamura, S.; Tanaka, Y.;
Chiyomaru, T.; et al. miRNA-34b inhibits prostate cancer through demethylation, active chromatin
modifications, and AKT pathways. Clin. Cancer Res. 2013, 19, 73–84. [CrossRef] [PubMed]
103. Siemens, H.; Neumann, J.; Jackstadt, R.; Mansmann, U.; Horst, D.; Kirchner, T.; Hermeking, H. Detection of
miR-34a promoter methylation in combination with elevated expression of c-Met and β-catenin predicts
distant metastasis of colon cancer. Clin. Cancer Res. 2013, 19, 710–720. [CrossRef] [PubMed]
104. Genovese, G.; Ergun, A.; Shukla, S.A.; Campos, B.; Hanna, J.; Ghosh, P.; Quayle, S.N.; Rai, K.; Colla, S.;
Ying, H.; et al. microRNA regulatory network inference identifies miR-34a as a novel regulator of TGF-β
signaling in glioblastoma. Cancer Discov. 2012, 2, 736–749. [CrossRef] [PubMed]
105. Tian, X.J.; Zhang, H.; Xing, J. Coupled reversible and irreversible bistable switches underlying TGF-β-induced
epithelial to mesenchymal transition. Biophys. J. 2013, 105, 1079–1089. [CrossRef] [PubMed]
106. Gregory, P.A.; Bracken, C.P.; Smith, E.; Bert, A.G.; Wright, J.A.; Roslan, S.; Morris, M.; Wyatt, L.; Farshid, G.;
Lim, Y.Y.; et al. An autocrine TGF-β/ZEB/miR-200 signaling network regulates establishment and maintenance
of epithelial-mesenchymal transition. Mol. Biol. Cell 2011, 22, 1686–1698. [CrossRef] [PubMed]
107. Kumar, M.S.; Armenteros-Monterroso, E.; East, P.; Chakravorty, P.; Matthews, N.; Winslow, M.M.;
Downward, J. HMGA2 functions as a competing endogenous RNA to promote lung cancer progression.
Nature 2014, 505, 212–217. [CrossRef]
108. Liao, Y.C.; Wang, Y.S.; Guo, Y.C.; Lin, W.L.; Chang, M.H.; Juo, S.H. Let-7g improves multiple endothelial
functions through targeting transforming growth factor-β and SIRT-1 signaling. J. Am. Coll. Cardiol. 2014, 63,
1685–1694. [CrossRef]
109. Han, X.; Yan, S.; Weijie, Z.; Feng, W.; Liuxing, W.; Mengquan, L.; Qingxia, F. Critical role of miR-10b in
transforming growth factor-β1-induced epithelial-mesenchymal transition in breast cancer. Cancer Gene Ther.
2014, 21, 60–67. [CrossRef]
110. Ma, L.; Reinhardt, F.; Pan, E.; Soutschek, J.; Bhat, B.; Marcusson, E.G.; Teruya-Feldstein, J.; Bell, G.W.;
Weinberg, R.A. Therapeutic silencing of miR-10b inhibits metastasis in a mouse mammary tumor model.
Nat. Biotechnol. 2010, 28, 341–347. [CrossRef]
111. Song, L.; Liu, L.; Wu, Z.; Li, Y.; Ying, Z.; Lin, C.; Wu, J.; Hu, B.; Cheng, S.Y.; Li, M.; et al. TGF-β induces
miR-182 to sustain NF-kappaB activation in glioma subsets. J. Clin. Investig. 2012, 122, 3563–3578. [CrossRef]
[PubMed]
112. Taylor, M.A.; Sossey-Alaoui, K.; Thompson, C.L.; Danielpour, D.; Schiemann, W.P. TGF-β upregulates
miR-181a expression to promote breast cancer metastasis. J. Clin. Investig. 2013, 123, 150–163. [CrossRef]
[PubMed]
113. Parikh, A.; Lee, C.; Joseph, P.; Marchini, S.; Baccarini, A.; Kolev, V.; Romualdi, C.; Fruscio, R.; Shah, H.;
Wang, F.; et al. microRNA-181a has a critical role in ovarian cancer progression through the regulation of the
epithelial-mesenchymal transition. Nat. Commun. 2014, 5, 2977. [CrossRef] [PubMed]
114. Bu, P.; Wang, L.; Chen, K.Y.; Rakhilin, N.; Sun, J.; Closa, A.; Tung, K.L.; King, S.; Varanko, A.K.; Xu, Y.; et al.
miR-1269 promotes metastasis and forms a positive feedback loop with TGF-β. Nat. Commun. 2015, 6, 6879.
[CrossRef] [PubMed]
115. Xia, H.; Ooi, L.L.; Hui, K.M. MicroRNA-216a/217-induced epithelial-mesenchymal transition targets PTEN
and SMAD7 to promote drug resistance and recurrence of liver cancer. Hepatology 2013, 58, 629–641.
[CrossRef] [PubMed]
116. Kong, W.; Yang, H.; He, L.; Zhao, J.J.; Coppola, D.; Dalton, W.S.; Cheng, J.Q. MicroRNA-155 is regulated by
the transforming growth factor β/SMAD pathway and contributes to epithelial cell plasticity by targeting
RhoA. Mol. Cell. Biol. 2008, 28, 6773–6784. [CrossRef]
117. Yin, K.; Yin, W.; Wang, Y.; Zhou, L.; Liu, Y.; Yang, G.; Wang, J.; Lu, J. MiR-206 suppresses epithelial
mesenchymal transition by targeting TGF-β signaling in estrogen receptor positive breast cancer cells.
Oncotarget 2016, 7, 24537–24548. [CrossRef] [PubMed]
118. Suzuki, H.I. MicroRNA Control of TGF-β Signaling. Int. J. Mol. Sci. 2018, 19, 1901. [CrossRef]
119. Zaravinos, A. The regulatory role of MicroRNAs in EMT and cancer. J. Oncol. 2015, 2015, 865816. [CrossRef]
120. Romano, G.; Kwong, L.N. miRNAs, melanoma and microenvironment: An intricate network. Int. J. Mol. Sci.
2017, 18, 2354. [CrossRef]
Int. J. Mol. Sci. 2019, 20, 2767 27 of 34
121. Musavi Shenas, M.H.; Eghbal-Fard, S.; Mehrisofiani, V.; Yazdani, N.A.; Farzam, O.R.; Marofi, F.; Yousefi, M.
MicroRNAs and signaling networks involved in epithelial-mesenchymal transition. J. Cell Physiol. 2019, 234,
5775–5785. [CrossRef]
122. Lin, C.W.; Kao, S.H.; Yang, P.C. The miRNAs and epithelial-mesenchymal transition in cancers.
Curr. Pharm. Des. 2014, 20, 5309–5318. [CrossRef]
123. Shields, E.J.; Petracovici, A.F.; Bonasio, R. lncRedibly versatile: Biochemical and biological functions of long
noncoding RNAs. Biochem. J. 2019, 476, 1083–1104. [CrossRef]
124. Gutschner, T.; Diederichs, S. The hallmarks of cancer: A long non-coding RNA point of view. RNA Biol.
2012, 9, 703–719. [CrossRef]
125. Schmitt, A.M.; Chang, H.Y. Long noncoding RNAs in cancer pathways. Cancer Cell 2016, 29, 452–463.
[CrossRef]
126. Lu, H.; Yang, D.; Zhang, L.; Lu, S.; Ye, J.; Li, M.; Hu, W. Linc-pint inhibits early stage pancreatic ductal
adenocarcinoma growth through TGF-β pathway activation. Oncol. Lett. 2019, 17, 4633–4639. [CrossRef]
127. Shin, T.J.; Lee, K.H.; Cho, H.M.; Cho, J.Y. Concise approach for screening long non-coding RNAs functionally
linked to human breast cancer associated genes. Exp. Mol. Pathol. 2019, 108, 89–96. [CrossRef]
128. Shi, T.; Gao, G.; Cao, Y. Long noncoding RNAs as novel biomarkers have a promising future in cancer
diagnostics. Dis. Markers 2016, 2016, 9085195. [CrossRef]
129. Sakai, S.; Ohhata, T.; Kitagawa, K.; Uchida, C.; Aoshima, T.; Niida, H.; Suzuki, T.; Inoue, Y.; Miyazawa, K.;
Kitagawa, M. Long noncoding RNA ELIT-1 acts as a SMAD3 cofactor to facilitate TGF-β/SMAD signaling
and promote epithelial-mesenchymal transition. Cancer Res. 2019.
130. He, L.; Thomson, J.M.; Hemann, M.T.; Hernando-Monge, E.; Mu, D.; Goodson, S.; Powers, S.;
Cordon-Cardo, C.; Lowe, S.W.; Hannon, G.J.; et al. A microRNA polycistron as a potential human
oncogene. Nature 2005, 435, 828–833. [CrossRef]
131. Li, W.; Kang, Y. A new Lnc in metastasis: Long noncoding RNA mediates the prometastatic functions of
TGF-β. Cancer Cell 2014, 25, 557–559. [CrossRef]
132. Yuan, J.H.; Yang, F.; Wang, F.; Ma, J.Z.; Guo, Y.J.; Tao, Q.F.; Liu, F.; Pan, W.; Wang, T.T.; Zhou, C.C.; et al.
A long noncoding RNA activated by TGF-β promotes the invasion-metastasis cascade in hepatocellular
carcinoma. Cancer Cell 2014, 25, 666–681. [CrossRef]
133. Grelet, S.; Link, L.A.; Howley, B.; Obellianne, C.; Palanisamy, V.; Gangaraju, V.K.; Diehl, J.A.; Howe, P.H.
A regulated PNUTS mRNA to lncRNA splice switch mediates EMT and tumour progression. Nat. Cell Biol.
2017, 19, 1105–1115. [CrossRef]
134. Mondal, T.; Subhash, S.; Vaid, R.; Enroth, S.; Uday, S.; Reinius, B.; Mitra, S.; Mohammed, A.; James, A.R.;
Hoberg, E.; et al. MEG3 long noncoding RNA regulates the TGF-β pathway genes through formation of
RNA-DNA triplex structures. Nat. Commun. 2015, 6, 7743. [CrossRef]
135. Mitra, R.; Chen, X.; Greenawalt, E.J.; Maulik, U.; Jiang, W.; Zhao, Z.; Eischen, C.M. Decoding critical long
non-coding RNA in ovarian cancer epithelial-to-mesenchymal transition. Nat. Commun. 2017, 8, 1604.
[CrossRef]
136. Yan, X.; Zhang, D.; Wu, W.; Wu, S.; Qian, J.; Hao, Y.; Yan, F.; Zhu, P.; Wu, J.; Huang, G.; et al. Mesenchymal stem
cells promote hepatocarcinogenesis via lncRNA-MUF interaction with ANXA2 and miR-34a. Cancer Res.
2017, 77, 6704–6716. [CrossRef]
137. Zhang, J.; Han, C.; Ungerleider, N.; Chen, W.; Song, K.; Wang, Y.; Kwon, H.; Ma, W.; Wu, T. A transforming
growth factor-β and H19 signaling axis in tumor-initiating hepatocytes that regulates hepatic carcinogenesis.
Hepatology 2018, 69, 1549–1563. [CrossRef]
138. Matouk, I.J.; Raveh, E.; Abu-lail, R.; Mezan, S.; Gilon, M.; Gershtain, E.; Birman, T.; Gallula, J.; Schneider, T.;
Barkali, M.; et al. Oncofetal H19 RNA promotes tumor metastasis. Biochim. Biophys. Acta 2014, 1843,
1414–1426. [CrossRef]
139. Zhou, W.; Ye, X.L.; Xu, J.; Cao, M.G.; Fang, Z.Y.; Li, L.Y.; Guan, G.H.; Liu, Q.; Qian, Y.H.; Xie, D. The lncRNA
H19 mediates breast cancer cell plasticity during EMT and MET plasticity by differentially sponging
miR-200b/c and let-7b. Sci. Signal 2017, 10, eaak9557. [CrossRef]
140. Luo, M.; Li, Z.; Wang, W.; Zeng, Y.; Liu, Z.; Qiu, J. Long non-coding RNA H19 increases bladder cancer
metastasis by associating with EZH2 and inhibiting E-cadherin expression. Cancer Lett. 2013, 333, 213–221.
[CrossRef]
Int. J. Mol. Sci. 2019, 20, 2767 28 of 34
141. Rodriguez-Mateo, C.; Torres, B.; Gutierrez, G.; Pintor-Toro, J.A. Downregulation of Lnc-Spry1 mediates
TGF-β-induced epithelial-mesenchymal transition by transcriptional and posttranscriptional regulatory
mechanisms. Cell Death Differ. 2017, 24, 785–797. [CrossRef]
142. Song, Y.X.; Sun, J.X.; Zhao, J.H.; Yang, Y.C.; Shi, J.X.; Wu, Z.H.; Chen, X.W.; Gao, P.; Miao, Z.F.; Wang, Z.N.
Non-coding RNAs participate in the regulatory network of CLDN4 via ceRNA mediated miRNA evasion.
Nat. Commun. 2017, 8, 289. [CrossRef]
143. Battistelli, C.; Cicchini, C.; Santangelo, L.; Tramontano, A.; Grassi, L.; Gonzalez, F.J.; de Nonno, V.; Grassi, G.;
Amicone, L.; Tripodi, M. The snail repressor recruits EZH2 to specific genomic sites through the enrollment
of the lncRNA HOTAIR in epithelial-to-mesenchymal transition. Oncogene 2017, 36, 942–955. [CrossRef]
144. Zhang, Y.; Li, J.; Jia, S.; Wang, Y.; Kang, Y.; Zhang, W. Down-regulation of lncRNA-ATB inhibits
epithelial-mesenchymal transition of breast cancer cells by increasing miR-141-3p expression. Biochem. Cell Biol.
2018, 97, 85–90. [CrossRef]
145. Li, G.Y.; Wang, W.; Sun, J.Y.; Xin, B.; Zhang, X.; Wang, T.; Zhang, Q.F.; Yao, L.B.; Han, H.; Fan, D.M.; et al.
Long non-coding RNAs AC026904.1 and UCA1: A “one-two punch” for TGF-β-induced SNAI2 activation
and epithelial-mesenchymal transition in breast cancer. Theranostics 2018, 8, 2846–2861. [CrossRef]
146. Wu, W.; Chen, F.; Cui, X.; Yang, L.; Chen, J.; Zhao, J.; Huang, D.; Liu, J.; Yang, L.; Zeng, J.; et al. LncRNA NKILA
suppresses TGF-β-induced epithelial-mesenchymal transition by blocking NF-kappaB signaling in breast
cancer. Int. J. Cancer 2018, 143, 2213–2224. [CrossRef]
147. Li, Z.; Dong, M.; Fan, D.; Hou, P.; Li, H.; Liu, L.; Lin, C.; Liu, J.; Su, L.; Wu, L.; et al. LncRNA ANCR
down-regulation promotes TGF-β-induced EMT and metastasis in breast cancer. Oncotarget 2017, 8,
67329–67343. [CrossRef]
148. Richards, E.J.; Zhang, G.; Li, Z.P.; Permuth-Wey, J.; Challa, S.; Li, Y.; Kong, W.; Dan, S.; Bui, M.M.;
Coppola, D.; et al. Long non-coding RNAs (LncRNA) regulated by transforming growth factor (TGF)
β: LncRNA-hit-mediated TGF-β-induced epithelial to mesenchymal transition in mammary epithelia.
J. Biol. Chem. 2015, 290, 6857–6867. [CrossRef]
149. Pan, Y.; Li, C.; Chen, J.; Zhang, K.; Chu, X.; Wang, R.; Chen, L. The emerging roles of long noncoding RNA
ROR (lincRNA-ROR) and its possible mechanisms in human cancers. Cell Physiol. Biochem. 2016, 40, 219–229.
[CrossRef]
150. Eades, G.; Wolfson, B.; Zhang, Y.; Li, Q.; Yao, Y.; Zhou, Q. lincRNA-RoR and miR-145 regulate invasion in
triple-negative breast cancer via targeting ARF6. Mol. Cancer Res. 2015, 13, 330–338. [CrossRef]
151. Fang, Y.; Wang, J.; Wu, F.; Song, Y.; Zhao, S.; Zhang, Q. Long non-coding RNA HOXA-AS2 promotes
proliferation and invasion of breast cancer by acting as a miR-520c-3p sponge. Oncotarget 2017, 8, 46090–46103.
[CrossRef]
152. Wu, Z.J.; Li, Y.; Wu, Y.Z.; Wang, Y.; Nian, W.Q.; Wang, L.L.; Li, L.C.; Luo, H.L.; Wang, D.L. Long non-coding
RNA CCAT2 promotes the breast cancer growth and metastasis by regulating TGF-β signaling pathway.
Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 706–714.
153. Zhang, C.Y.; Yu, M.S.; Li, X.; Zhang, Z.; Han, C.R.; Yan, B. Overexpression of long non-coding RNA
MEG3 suppresses breast cancer cell proliferation, invasion, and angiogenesis through AKT pathway.
Tumour Biol. 2017, 39, 1010428317701311. [CrossRef]
154. Zhang, W.; Shi, S.; Jiang, J.; Li, X.; Lu, H.; Ren, F. LncRNA MEG3 inhibits cell epithelial-mesenchymal
transition by sponging miR-421 targeting E-cadherin in breast cancer. Biomed. Pharmacother. 2017, 91, 312–319.
[CrossRef]
155. Fu, M.; Huang, Z.; Zang, X.; Pan, L.; Liang, W.; Chen, J.; Qian, H.; Xu, W.; Jiang, P.; Zhang, X. Long noncoding
RNA LINC00978 promotes cancer growth and acts as a diagnostic biomarker in gastric cancer. Cell Prolif.
2018, 51, e12425. [CrossRef]
156. Zuo, Z.K.; Gong, Y.; Chen, X.H.; Ye, F.; Yin, Z.M.; Gong, Q.N.; Huang, J.S. TGF-β1-Induced LncRNA
UCA1 upregulation promotes gastric cancer invasion and migration. DNA Cell Biol. 2017, 36, 159–167.
[CrossRef]
157. Saito, T.; Kurashige, J.; Nambara, S.; Komatsu, H.; Hirata, H.; Ueda, M.; Sakimura, S.; Uchi, R.; Takano, Y.;
Shinden, Y.; et al. A long non-coding RNA activated by transforming growth factor-β is an independent
prognostic marker of gastric cancer. Ann. Surg. Oncol. 2015, 22 (Suppl. 3), S915–S922. [CrossRef]
158. Chen, Y.; Wei, G.; Xia, H.; Tang, Q.; Bi, F. Long noncoding RNAATB promotes cell proliferation, migration
and invasion in gastric cancer. Mol. Med. Rep. 2018, 17, 1940–1946.
Int. J. Mol. Sci. 2019, 20, 2767 29 of 34
159. Lei, K.; Liang, X.; Gao, Y.; Xu, B.; Xu, Y.; Li, Y.; Tao, Y.; Shi, W.; Liu, J. Lnc-ATB contributes to gastric cancer
growth through a MiR-141-3p/TGF-β2 feedback loop. Biochem. Biophys. Res. Commun. 2017, 484, 514–521.
[CrossRef]
160. Zhang, Q.; Chen, B.; Liu, P.; Yang, J. XIST promotes gastric cancer (GC) progression through TGF-β1 via
targeting miR-185. J. Cell Biochem. 2018, 119, 2787–2796. [CrossRef]
161. Han, Q.; Zhang, W.; Meng, J.; Ma, L.; Li, A. LncRNA-LET inhibits cell viability, migration and EMT
while induces apoptosis by up-regulation of TIMP2 in human granulosa-like tumor cell line KGN.
Biomed. Pharmacother. 2018, 100, 250–256. [CrossRef]
162. Li, J.; Huang, Y.; Deng, X.; Luo, M.; Wang, X.; Hu, H.; Liu, C.; Zhong, M. Long noncoding RNA
H19 promotes transforming growth factor-β-induced epithelial-mesenchymal transition by acting as
a competing endogenous RNA of miR-370-3p in ovarian cancer cells. Onco. Targets Ther. 2018, 11, 427–440.
[CrossRef]
163. Li, Q.; Zhang, C.; Chen, R.; Xiong, H.; Qiu, F.; Liu, S.; Zhang, M.; Wang, F.; Wang, Y.; Zhou, X.; et al.
Disrupting MALAT1/miR-200c sponge decreases invasion and migration in endometrioid endometrial
carcinoma. Cancer Lett. 2016, 383, 28–40. [CrossRef]
164. Hu, J.; Zhang, L.; Mei, Z.; Jiang, Y.; Yi, Y.; Liu, L.; Meng, Y.; Zhou, L.; Zeng, J.; Wu, H.; et al.
Interaction of E3 ubiquitin ligase MARCH7 with long noncoding RNA MALAT1 and autophagy-related
protein ATG7 promotes autophagy and invasion in ovarian cancer. Cell Physiol. Biochem. 2018, 47, 654–666.
[CrossRef]
165. Ma, J.; Xue, M. LINK-A lncRNA promotes migration and invasion of ovarian carcinoma cells by activating
TGF-β pathway. Biosci. Rep. 2018, 38, BSR20180936. [CrossRef]
166. Cao, C.; Zhang, T.; Zhang, D.; Xie, L.; Zou, X.; Lei, L.; Wu, D.; Liu, L. The long non-coding RNA, SNHG6-003,
functions as a competing endogenous RNA to promote the progression of hepatocellular carcinoma. Oncogene
2017, 36, 1112–1122. [CrossRef]
167. Liang, H.; Zhao, X.; Wang, C.; Sun, J.; Chen, Y.; Wang, G.; Fang, L.; Yang, R.; Yu, M.; Gu, Y.;
et al. Systematic analyses reveal long non-coding RNA (PTAF)-mediated promotion of EMT and
invasion-metastasis in serous ovarian cancer. Mol. Cancer 2018, 17, 96. [CrossRef]
168. Zhuang, J.; Lu, Q.; Shen, B.; Huang, X.; Shen, L.; Zheng, X.; Huang, R.; Yan, J.; Guo, H. TGF-β1 secreted
by cancer-associated fibroblasts induces epithelial-mesenchymal transition of bladder cancer cells through
lncRNA-ZEB2NAT. Sci. Rep. 2015, 5, 11924. [CrossRef]
169. Fan, Y.; Shen, B.; Tan, M.; Mu, X.; Qin, Y.; Zhang, F.; Liu, Y. TGF-β-induced upregulation of malat1 promotes
bladder cancer metastasis by associating with suz12. Clin. Cancer Res. 2014, 20, 1531–1541. [CrossRef]
170. Gou, L.; Liu, M.; Xia, J.; Wan, Q.; Jiang, Y.; Sun, S.; Tang, M.; Zhou, L.; He, T.; Zhang, Y. BMP9 promotes the
proliferation and migration of bladder cancer cells through up-regulating lncRNA UCA1. Int. J. Mol. Sci.
2018, 19, 1116. [CrossRef]
171. Zhai, X.; Xu, W. Long noncoding RNA ATB promotes proliferation, migration, and invasion in bladder
cancer by suppressing MicroRNA-126. Oncol. Res. 2018, 26, 1063–1072. [CrossRef]
172. Jin, Y.; Cui, Z.; Li, X.; Jin, X.; Peng, J. Upregulation of long non-coding RNA PlncRNA-1 promotes proliferation
and induces epithelial-mesenchymal transition in prostate cancer. Oncotarget 2017, 8, 26090–26099. [CrossRef]
173. Wang, S.H.; Zhang, M.D.; Wu, X.C.; Weng, M.Z.; Zhou, D.; Quan, Z.W. Overexpression of LncRNA-ROR
predicts a poor outcome in gallbladder cancer patients and promotes the tumor cells proliferation, migration,
and invasion. Tumour Biol. 2016, 37, 12867–12875. [CrossRef]
174. Zhao, B.; Lu, Y.L.; Yang, Y.; Hu, L.B.; Bai, Y.; Li, R.Q.; Zhang, G.Y.; Li, J.; Bi, C.W.; Yang, L.B.; et al.
Overexpression of lncRNA ANRIL promoted the proliferation and migration of prostate cancer cells via
regulating let-7a/TGF-β1/ SMAD signaling pathway. Cancer Biomark. 2018, 21, 613–620. [CrossRef]
175. Xu, S.; Yi, X.M.; Tang, C.P.; Ge, J.P.; Zhang, Z.Y.; Zhou, W.Q. Long non-coding RNA ATB promotes growth
and epithelial-mesenchymal transition and predicts poor prognosis in human prostate carcinoma. Oncol. Rep.
2016, 36, 10–22. [CrossRef]
176. Wang, H.; Wang, G.; Gao, Y.; Zhao, C.; Li, X.; Zhang, F.; Jiang, C.; Wu, B. Lnc-SNHG1 activates the
TGFBR2/SMAD3 and RAB11A/Wnt/β-catenin pathway by sponging MiR-302/372/373/520 in invasive
pituitary tumors. Cell Physiol. Biochem. 2018, 48, 1291–1303. [CrossRef]
Int. J. Mol. Sci. 2019, 20, 2767 30 of 34
177. Li, Z.; Liu, H.; Zhong, Q.; Wu, J.; Tang, Z. LncRNA UCA1 is necessary for TGF-β-induced
epithelial-mesenchymal transition and stemness via acting as a ceRNA for SLUG in glioma cells.
FEBS Open Bio. 2018, 8, 1855–1865. [CrossRef]
178. Zhang, C.; Hao, Y.; Wang, Y.; Xu, J.; Teng, Y.; Yang, X. TGF-β/SMAD4-regulated LncRNA-LINP1 inhibits
epithelial-mesenchymal transition in lung cancer. Int. J. Biol. Sci. 2018, 14, 1715–1723. [CrossRef]
179. Lu, Z.; Li, Y.; Che, Y.; Huang, J.; Sun, S.; Mao, S.; Lei, Y.; Li, N.; Sun, N.; He, J. The TGF-β-induced lncRNA
TBILA promotes non-small cell lung cancer progression in vitro and in vivo via cis-regulating HGAL and
activating S100A7/JAB1 signaling. Cancer Lett. 2018, 432, 156–168. [CrossRef]
180. Wang, X.; Zhang, G.; Cheng, Z.; Dai, L.; Jia, L.; Jing, X.; Wang, H.; Zhang, R.; Liu, M.; Jiang, T.; et al.
Knockdown of LncRNA-XIST suppresses proliferation and TGF-β1-induced EMT in NSCLC through the
notch-1 pathway by regulation of miR-137. Genet. Test Mol. Biomark. 2018, 22, 333–342. [CrossRef]
181. Li, C.; Wan, L.; Liu, Z.; Xu, G.; Wang, S.; Su, Z.; Zhang, Y.; Zhang, C.; Liu, X.; Lei, Z.; et al. Long non-coding
RNA XIST promotes TGF-β-induced epithelial-mesenchymal transition by regulating miR-367/141-ZEB2 axis
in non-small-cell lung cancer. Cancer Lett. 2018, 418, 185–195. [CrossRef]
182. Kawasaki, N.; Miwa, T.; Hokari, S.; Sakurai, T.; Ohmori, K.; Miyauchi, K.; Miyazono, K.;
Koinuma, D. Long noncoding RNA NORAD regulates transforming growth factor-β signaling and
epithelial-to-mesenchymal transition-like phenotype. Cancer Sci. 2018, 109, 2211–2220. [CrossRef]
183. Cao, Y.; Luo, X.; Ding, X.; Cui, S.; Guo, C. LncRNA ATB promotes proliferation and metastasis in A549 cells
by down-regulation of microRNA-494. J. Cell Biochem. 2018, 119, 6935–6942. [CrossRef]
184. Lu, W.; Zhang, H.; Niu, Y.; Wu, Y.; Sun, W.; Li, H.; Kong, J.; Ding, K.; Shen, H.M.; Wu, H.; et al.
Long non-coding RNA linc00673 regulated non-small cell lung cancer proliferation, migration, invasion and
epithelial mesenchymal transition by sponging miR-150-5p. Mol. Cancer 2017, 16, 118. [CrossRef]
185. Lu, Z.; Li, Y.; Wang, J.; Che, Y.; Sun, S.; Huang, J.; Chen, Z.; He, J. Long non-coding RNA NKILA inhibits
migration and invasion of non-small cell lung cancer via NF-kappaB/Snail pathway. J. Exp. Clin. Cancer Res.
2017, 36, 54. [CrossRef]
186. Terashima, M.; Tange, S.; Ishimura, A.; Suzuki, T. MEG3 long noncoding RNA contributes to the epigenetic
regulation of epithelial-mesenchymal transition in lung cancer cell lines. J. Biol. Chem. 2017, 292, 82–99.
[CrossRef]
187. Wu, D.M.; Deng, S.H.; Liu, T.; Han, R.; Zhang, T.; Xu, Y. TGF-β-mediated exosomal lnc-MMP2-2 regulates
migration and invasion of lung cancer cells to the vasculature by promoting MMP2 expression. Cancer Med.
2018, 7, 5118–5129. [CrossRef]
188. Wang, S.; Lan, F.; Xia, Y. lncRA ANCR inhibits non-small cell lung cancer cell migration and invasion by
inactivating TGF-β pathway. Med. Sci. Monit. 2018, 24, 6002–6009. [CrossRef]
189. Hao, Y.; Yang, X.; Zhang, D.; Luo, J.; Chen, R. Long noncoding RNA LINC01186, regulated by TGF-β/SMAD3,
inhibits migration and invasion through epithelial-mesenchymal-transition in lung cancer. Gene 2017, 608,
1–12. [CrossRef]
190. Zhang, Y.; He, R.Q.; Dang, Y.W.; Zhang, X.L.; Wang, X.; Huang, S.N.; Huang, W.T.; Jiang, M.T.; Gan, X.N.;
Xie, Y.; et al. Comprehensive analysis of the long noncoding RNA HOXA11-AS gene interaction regulatory
network in NSCLC cells. Cancer Cell Int. 2016, 16, 89. [CrossRef]
191. Li, Y.; Liu, G.; Li, X.; Dong, H.; Xiao, W.; Lu, S. Long non-coding RNA SBF2-AS1 promotes hepatocellular
carcinoma progression through regulation of miR-140-5p-TGFBR1 pathway. Biochem. Biophys. Res. Commun.
2018, 503, 2826–2832. [CrossRef]
192. Tang, J.; Zhuo, H.; Zhang, X.; Jiang, R.; Ji, J.; Deng, L.; Qian, X.; Zhang, F.; Sun, B. A novel biomarker
Linc00974 interacting with KRT19 promotes proliferation and metastasis in hepatocellular carcinoma.
Cell Death Dis. 2014, 5, e1549. [CrossRef]
193. Zhang, X.; Feng, W.; Zhang, J.; Ge, L.; Zhang, Y.; Jiang, X.; Peng, W.; Wang, D.; Gong, A.; Xu, M.
Long noncoding RNA PVT1 promotes epithelialmesenchymal transition via the TGF-β/SMAD pathway in
pancreatic cancer cells. Oncol. Rep. 2018, 40, 1093–1102.
194. Terashima, M.; Ishimura, A.; Wanna-Udom, S.; Suzuki, T. MEG8 long noncoding RNA contributes to epigenetic
progression of the epithelial-mesenchymal transition of lung and pancreatic cancer cells. J. Biol. Chem. 2018,
293, 18016–18030. [CrossRef]
195. Qin, C.F.; Zhao, F.L. Long non-coding RNA TUG1 can promote proliferation and migration of pancreatic
cancer via EMT pathway. Eur. Rev. Med. Pharmacol. Sci. 2017, 21, 2377–2384.
Int. J. Mol. Sci. 2019, 20, 2767 31 of 34
196. Ottaviani, S.; Stebbing, J.; Frampton, A.E.; Zagorac, S.; Krell, J.; de Giorgio, A.; Trabulo, S.M.; Nguyen, V.T.M.;
Magnani, L.; Feng, H.; et al. TGF-β induces miR-100 and miR-125b but blocks let-7a through LIN28B
controlling PDAC progression. Nat. Commun. 2018, 9, 1845. [CrossRef]
197. Qu, S.; Yang, X.; Song, W.; Sun, W.; Li, X.; Wang, J.; Zhong, Y.; Shang, R.; Ruan, B.; Zhang, Z.; et al.
Downregulation of lncRNA-ATB correlates with clinical progression and unfavorable prognosis in pancreatic
cancer. Tumour Biol. 2016, 37, 3933–3938. [CrossRef]
198. Liu, Y.; Qian, J.; Li, X.; Chen, W.; Xu, A.; Zhao, K.; Hua, Y.; Huang, Z.; Zhang, J.; Liang, C.; et al.
Long noncoding RNA BX357664 regulates cell proliferation and epithelial-to-mesenchymal transition via
inhibition of TGF-β1/p38/HSP27 signaling in renal cell carcinoma. Oncotarget 2016, 7, 81410–81422.
199. Xiong, J.; Liu, Y.; Jiang, L.; Zeng, Y.; Tang, W. High expression of long non-coding RNA lncRNA-ATB
is correlated with metastases and promotes cell migration and invasion in renal cell carcinoma. Jpn. J.
Clin. Oncol. 2016, 46, 378–384. [CrossRef]
200. Yue, B.; Qiu, S.; Zhao, S.; Liu, C.; Zhang, D.; Yu, F.; Peng, Z.; Yan, D. LncRNA-ATB mediated E-cadherin
repression promotes the progression of colon cancer and predicts poor prognosis. J. Gastroenterol. Hepatol.
2016, 31, 595–603. [CrossRef]
201. Iguchi, T.; Uchi, R.; Nambara, S.; Saito, T.; Komatsu, H.; Hirata, H.; Ueda, M.; Sakimura, S.; Takano, Y.;
Kurashige, J.; et al. A long noncoding RNA, lncRNA-ATB, is involved in the progression and prognosis of
colorectal cancer. Anticancer Res. 2015, 35, 1385–1388.
202. Kong, J.; Sun, W.; Li, C.; Wan, L.; Wang, S.; Wu, Y.; Xu, E.; Zhang, H.; Lai, M. Long non-coding RNA
LINC01133 inhibits epithelial-mesenchymal transition and metastasis in colorectal cancer by interacting
with SRSF6. Cancer Lett. 2016, 380, 476–484. [CrossRef]
203. Zhang, S.Z.; Cai, L.; Li, B. MEG3 long non-coding RNA prevents cell growth and metastasis of osteosarcoma.
Bratisl Lek Listy 2017, 118, 632–636. [CrossRef]
204. Huo, Y.; Li, Q.; Wang, X.; Jiao, X.; Zheng, J.; Li, Z.; Pan, X. MALAT1 predicts poor survival in osteosarcoma
patients and promotes cell metastasis through associating with EZH2. Oncotarget 2017, 8, 46993–47006.
[CrossRef]
205. Deng, Y.; Zhao, F.; Zhang, Z.; Sun, F.; Wang, M. Long noncoding RNA SNHG7 promotes the tumor
growth and epithelial-to-mesenchymal transition via regulation of mir-34a signals in osteosarcoma.
Cancer Biother. Radiopharm. 2018, 33, 365–372. [CrossRef]
206. Zhang, R.; Hardin, H.; Huang, W.; Chen, J.; Asioli, S.; Righi, A.; Maletta, F.; Sapino, A.; Lloyd, R.V.
MALAT1 long non-coding RNA expression in thyroid tissues: Analysis by in situ hybridization and real-time
PCR. Endocr. Pathol. 2017, 28, 7–12. [CrossRef]
207. Zhang, R.; Hardin, H.; Huang, W.; Buehler, D.; Lloyd, R.V. Long non-coding RNA linc-ROR is upregulated in
papillary thyroid carcinoma. Endocr. Pathol. 2018, 29, 1–8. [CrossRef]
208. Zhou, H.; Sun, Z.; Li, S.; Wang, X.; Zhou, X. LncRNA SPRY4-IT was concerned with the poor prognosis and
contributed to the progression of thyroid cancer. Cancer Gene Ther. 2018, 25, 39–46. [CrossRef]
209. Zhao, J.J.; Hao, S.; Wang, L.L.; Hu, C.Y.; Zhang, S.; Guo, L.J.; Zhang, G.; Gao, B.; Jiang, Y.; Tian, W.G.;
et al. Long non-coding RNA ANRIL promotes the invasion and metastasis of thyroid cancer cells through
TGF-β/SMAD signaling pathway. Oncotarget 2016, 7, 57903–57918. [CrossRef]
210. Lambert, A.W.; Pattabiraman, D.R.; Weinberg, R.A. Emerging biological principles of metastasis. Cell 2017,
168, 670–691. [CrossRef]
211. Chiang, A.C.; Massague, J. Molecular basis of metastasis. N. Engl. J. Med. 2008, 359, 2814–2823. [CrossRef]
212. Jung, H.Y.; Fattet, L.; Yang, J. Molecular pathways: Linking tumor microenvironment to
epithelial-mesenchymal transition in metastasis. Clin. Cancer Res. 2015, 21, 962–968. [CrossRef]
213. Nguyen, D.X.; Bos, P.D.; Massague, J. Metastasis: From dissemination to organ-specific colonization.
Nat. Rev. Cancer 2009, 9, 274–284. [CrossRef]
214. Roodman, G.D. Biology of osteoclast activation in cancer. J. Clin. Oncol. 2001, 19, 3562–3571. [CrossRef]
215. Yin, J.J.; Selander, K.; Chirgwin, J.M.; Dallas, M.; Grubbs, B.G.; Wieser, R.; Massague, J.; Mundy, G.R.;
Guise, T.A. TGF-β signaling blockade inhibits PTHrP secretion by breast cancer cells and bone metastases
development. J. Clin. Investig. 1999, 103, 197–206. [CrossRef]
216. Kang, Y.; Siegel, P.M.; Shu, W.; Drobnjak, M.; Kakonen, S.M.; Cordon-Cardo, C.; Guise, T.A.; Massague, J.
A multigenic program mediating breast cancer metastasis to bone. Cancer Cell 2003, 3, 537–549. [CrossRef]
Int. J. Mol. Sci. 2019, 20, 2767 32 of 34
217. Buijs, J.T.; Stayrook, K.R.; Guise, T.A. The role of TGF-β in bone metastasis: Novel therapeutic perspectives.
Bonekey Rep. 2012, 1, 96. [CrossRef]
218. Biswas, S.; Guix, M.; Rinehart, C.; Dugger, T.C.; Chytil, A.; Moses, H.L.; Freeman, M.L.; Arteaga, C.L.
Inhibition of TGF-β with neutralizing antibodies prevents radiation-induced acceleration of metastatic cancer
progression. J. Clin. Investig. 2007, 117, 1305–1313. [CrossRef]
219. Padua, D.; Zhang, X.H.; Wang, Q.; Nadal, C.; Gerald, W.L.; Gomis, R.R.; Massague, J. TGF-β primes breast
tumors for lung metastasis seeding through angiopoietin-like 4. Cell 2008, 133, 66–77. [CrossRef]
220. Gupta, G.P.; Perk, J.; Acharyya, S.; de Candia, P.; Mittal, V.; Todorova-Manova, K.; Gerald, W.L.; Brogi, E.;
Benezra, R.; Massague, J. ID genes mediate tumor reinitiation during breast cancer lung metastasis. Proc. Natl.
Acad. Sci. USA 2007, 104, 19506–19511. [CrossRef]
221. Zhuang, X.; Zhang, H.; Li, X.; Li, X.; Cong, M.; Peng, F.; Yu, J.; Zhang, X.; Yang, Q.; Hu, G. Differential effects
on lung and bone metastasis of breast cancer by Wnt signalling inhibitor DKK1. Nat. Cell. Biol. 2017, 19,
1274–1285. [CrossRef]
222. Jung, B.; Staudacher, J.J.; Beauchamp, D. Transforming growth factor β superfamily signaling in development
of colorectal cancer. Gastroenterology 2017, 152, 36–52. [CrossRef]
223. Steinbichler, T.B.; Dudas, J.; Riechelmann, H.; Skvortsova, II. The role of exosomes in cancer metastasis.
Semin. Cancer Biol. 2017, 44, 170–181. [CrossRef]
224. Carmeliet, P.; Jain, R.K. Angiogenesis in cancer and other diseases. Nature 2000, 407, 249–257. [CrossRef]
225. Roberts, A.B.; Thompson, N.L.; Heine, U.; Flanders, C.; Sporn, M.B. Transforming growth factor-β: Possible
roles in carcinogenesis. Br. J. Cancer 1988, 57, 594–600. [CrossRef]
226. Goumans, M.J.; Valdimarsdottir, G.; Itoh, S.; Rosendahl, A.; Sideras, P.; Dijke, P.t. Balancing the activation
state of the endothelium via two distinct TGF-β type I receptors. EMBO J. 2002, 21, 1743–1753. [CrossRef]
227. Breier, G.; Blum, S.; Peli, J.; Groot, M.; Wild, C.; Risau, W.; Reichmann, E. Transforming growth factor-β and
RAS regulate the VEGF/VEGF-receptor system during tumor angiogenesis. Int. J. Cancer 2002, 97, 142–148.
[CrossRef]
228. Sanchez-Elsner, T.; Botella, L.M.; Velasco, B.; Corbi, A.; Attisano, L.; Bernabeu, C. Synergistic cooperation
between hypoxia and transforming growth factor-β pathways on human vascular endothelial growth factor
gene expression. J. Biol. Chem. 2001, 276, 38527–38535. [CrossRef]
229. Liu, D.; Li, L.; Zhang, X.X.; Wan, D.Y.; Xi, B.X.; Hu, Z.; Ding, W.C.; Zhu, D.; Wang, X.L.; Wang, W.; et al.
SIX1 promotes tumor lymphangiogenesis by coordinating TGF-β signals that increase expression of VEGF-C.
Cancer Res. 2014, 74, 5597–5607. [CrossRef]
230. Sun, H.; Miao, C.; Liu, W.; Qiao, X.; Yang, W.; Li, L.; Li, C. TGF-β1/TβRII/SMAD3 signaling pathway promotes
VEGF expression in oral squamous cell carcinoma tumor-associated macrophages. Biochem. Biophys.
Res. Commun. 2018, 497, 583–590. [CrossRef]
231. Groppa, E.; Brkic, S.; Bovo, E.; Reginato, S.; Sacchi, V.; di Maggio, N.; Muraro, M.G.; Calabrese, D.;
Heberer, M.; Gianni-Barrera, R.; et al. VEGF dose regulates vascular stabilization through Semaphorin3A
and the Neuropilin-1 + monocyte/TGF-β1 paracrine axis. EMBO Mol. Med. 2015, 7, 1366–1384. [CrossRef]
232. Rak, J.; Mitsuhashi, Y.; Sheehan, C.; Tamir, A.; Viloria-Petit, A.; Filmus, J.; Mansour, S.J.; Ahn, N.G.; Kerbel, R.S.
Oncogenes and tumor angiogenesis: Differential modes of vascular endothelial growth factor up-regulation
in ras-transformed epithelial cells and fibroblasts. Cancer Res. 2000, 60, 490–498.
233. Miyazono, K.; Ehata, S.; Koinuma, D. Tumor-promoting functions of transforming growth factor-β in
progression of cancer. Ups. J. Med. Sci. 2012, 117, 143–152. [CrossRef]
234. Chen, W.; Ten Dijke, P. Immunoregulation by members of the TGF-β superfamily. Nat. Rev. Immunol. 2016,
16, 723–740. [CrossRef]
235. Gorelik, L.; Flavell, R.A. Abrogation of TGF-β signaling in T cells leads to spontaneous T cell differentiation
and autoimmune disease. Immunity 2000, 12, 171–181. [CrossRef]
236. Donkor, M.K.; Sarkar, A.; Savage, P.A.; Franklin, R.A.; Johnson, L.K.; Jungbluth, A.A.; Allison, J.P.; Li, M.O.
T cell surveillance of oncogene-induced prostate cancer is impeded by T cell-derived TGF-β1 cytokine.
Immunity 2011, 35, 123–134. [CrossRef]
237. Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel, E.E., III; Koeppen, H.;
Astarita, J.L.; Cubas, R.; et al. TGF-β attenuates tumour response to PD-L1 blockade by contributing to
exclusion of T cells. Nature 2018, 554, 544–548. [CrossRef]
Int. J. Mol. Sci. 2019, 20, 2767 33 of 34
238. Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.;
Sevillano, M.; Ibiza, S.; Canellas, A.; Hernando-Momblona, X.; et al. TGF-β drives immune evasion in
genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [CrossRef]
239. Gorelik, L.; Flavell, R.A. Immune-mediated eradication of tumors through the blockade of transforming
growth factor-β signaling in T cells. Nat. Med. 2001, 7, 1118–1122. [CrossRef]
240. Thomas, D.A.; Massague, J. TGF-β directly targets cytotoxic T cell functions during tumor evasion of immune
surveillance. Cancer Cell 2005, 8, 369–380. [CrossRef]
241. Fu, S.; Zhang, N.; Yopp, A.C.; Chen, D.; Mao, M.; Chen, D.; Zhang, H.; Ding, Y.; Bromberg, J.S. TGF-β induces
Foxp3 + T-regulatory cells from CD4 + CD25 - precursors. Am. J. Transplant. 2004, 4, 1614–1627. [CrossRef]
242. Worthington, J.J.; Kelly, A.; Smedley, C.; Bauche, D.; Campbell, S.; Marie, J.C.; Travis, M.A.
Integrin alphavβ8-mediated TGF-β activation by effector regulatory T cells is essential for suppression of
T-Cell-mediated inflammation. Immunity 2015, 42, 903–915. [CrossRef]
243. Metelli, A.; Wu, B.X.; Fugle, C.W.; Rachidi, S.; Sun, S.; Zhang, Y.; Wu, J.; Tomlinson, S.; Howe, P.H.; Yang, Y.;
et al. Surface expression of TGF-β docking receptor GARP promotes oncogenesis and immune tolerance in
breast cancer. Cancer Res. 2016, 76, 7106–7117. [CrossRef]
244. Imai, K.; Minamiya, Y.; Koyota, S.; Ito, M.; Saito, H.; Sato, Y.; Motoyama, S.; Sugiyama, T.; Ogawa, J.
Inhibition of dendritic cell migration by transforming growth factor-β1 increases tumor-draining lymph
node metastasis. J. Exp. Clin. Cancer Res. 2012, 31, 3. [CrossRef]
245. Ito, M.; Minamiya, Y.; Kawai, H.; Saito, S.; Saito, H.; Nakagawa, T.; Imai, K.; Hirokawa, M.; Ogawa, J.
Tumor-derived TGF-β-1 induces dendritic cell apoptosis in the sentinel lymph node. J. Immunol. 2006, 176,
5637–5643. [CrossRef]
246. Kobie, J.J.; Wu, R.S.; Kurt, R.A.; Lou, S.; Adelman, M.K.; Whitesell, L.J.; Ramanathapuram, L.V.; Arteaga, C.L.;
Akporiaye, E.T. Transforming growth factor β inhibits the antigen-presenting functions and antitumor
activity of dendritic cell vaccines. Cancer Res. 2003, 63, 1860–1864.
247. Allavena, P.; Sica, A.; Garlanda, C.; Mantovani, A. The Yin-Yang of tumor-associated macrophages in
neoplastic progression and immune surveillance. Immunol. Rev. 2008, 222, 155–161. [CrossRef]
248. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of
tumor-associated neutrophil phenotype by TGF-β: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194.
[CrossRef]
249. Wang, Y.; Chu, J.; Yi, P.; Dong, W.; Saultz, J.; Wang, Y.; Wang, H.; Scoville, S.; Zhang, J.; Wu, L.C.; et al.
SMAD4 promotes TGF-β-independent NK cell homeostasis and maturation and antitumor immunity.
J. Clin. Investig. 2018, 128, 5123–5136. [CrossRef]
250. Li, M.O.; Sanjabi, S.; Flavell, R.A. Transforming growth factor-β controls development, homeostasis,
and tolerance of T cells by regulatory T cell-dependent and -independent mechanisms. Immunity 2006, 25,
455–471. [CrossRef]
251. Akhurst, R.J. Targeting TGF-β signaling for therapeutic gain. Cold Spring Harb. Perspect. Biol. 2017, 9,
a022301. [CrossRef]
252. Colak, S.; Ten Dijke, P. Targeting TGF-β signaling in cancer. Trends Cancer 2017, 3, 56–71. [CrossRef]
253. Zappa, C.; Mousa, S.A. Non-small cell lung cancer: Current treatment and future advances. Transl. Lung
Cancer Res. 2016, 5, 288–300. [CrossRef]
254. Jaschinski, F.; Rothhammer, T.; Jachimczak, P.; Seitz, C.; Schneider, A.; Schlingensiepen, K.H. The antisense
oligonucleotide trabedersen (AP 12009) for the targeted inhibition of TGF-β2. Curr. Pharm. Biotechnol. 2011,
12, 2203–2213. [CrossRef]
255. Fujiwara, Y.; Nokihara, H.; Yamada, Y.; Yamamoto, N.; Sunami, K.; Utsumi, H.; Asou, H.; Takahash, I.O.;
Ogasawara, K.; Gueorguieva, I.; et al. Phase 1 study of galunisertib, a TGF-β receptor I kinase inhibitor,
in Japanese patients with advanced solid tumors. Cancer Chemother. Pharmacol. 2015, 76, 1143–1152.
[CrossRef]
256. Garrison, K.; Hahn, T.; Lee, W.C.; Ling, L.E.; Weinberg, A.D.; Akporiaye, E.T. The small molecule TGF-β
signaling inhibitor SM16 synergizes with agonistic OX40 antibody to suppress established mammary tumors
and reduce spontaneous metastasis. Cancer Immunol. Immunother. 2012, 61, 511–521. [CrossRef]
257. Mohammad, K.S.; Javelaud, D.; Fournier, P.G.; Niewolna, M.; McKenna, C.R.; Peng, X.H.; Duong, V.;
Dunn, L.K.; Mauviel, A.; Guise, T.A. TGF-β RI kinase inhibitor SD-208 reduces the development and
progression of melanoma bone metastases. Cancer Res. 2011, 71, 175–184. [CrossRef]
Int. J. Mol. Sci. 2019, 20, 2767 34 of 34
258. Uhl, M.; Aulwurm, S.; Wischhusen, J.; Weiler, M.; Ma, J.Y.; Almirez, R.; Mangadu, R.; Liu, Y.W.; Platten, M.;
Herrlinger, U.; et al. SD-208, a novel transforming growth factor β receptor I kinase inhibitor, inhibits growth
and invasiveness and enhances immunogenicity of murine and human glioma cells in vitro and in vivo.
Cancer Res. 2004, 64, 7954–7961. [CrossRef]
259. Akbari, A.; Amanpour, S.; Muhammadnejad, S.; Ghahremani, M.H.; Ghaffari, S.H.; Dehpour, A.R.;
Mobini, G.R.; Shidfar, F.; Abastabar, M.; Khoshzaban, A.; et al. Evaluation of antitumor activity of a
TGF-β receptor I inhibitor (SD-208) on human colon adenocarcinoma. Daru 2014, 22, 47. [CrossRef]
260. Doi, T.; Lee, K.H.; Kim, T.M.; Ohtsu, A.; Kim, T.Y.; Ikeda, M.; Yoh, K.; Stampino, C.G.; Hirohashi, T.; Suzuki, A.;
et al. A phase I study of the human anti-activin receptor-like kinase 1 antibody PF-03446962 in Asian patients
with advanced solid tumors. Cancer Med. 2016, 5, 1454–1463. [CrossRef]
261. Morris, J.C.; Tan, A.R.; Olencki, T.E.; Shapiro, G.I.; Dezube, B.J.; Reiss, M.; Hsu, F.J.; Berzofsky, J.A.;
Lawrence, D.P. Phase I study of GC1008 (fresolimumab): A human anti-transforming growth factor-β
(TGF-β) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma.
PLoS ONE 2014, 9, e90353. [CrossRef]
262. Lacouture, M.E.; Morris, J.C.; Lawrence, D.P.; Tan, A.R.; Olencki, T.E.; Shapiro, G.I.; Dezube, B.J.;
Berzofsky, J.A.; Hsu, F.J.; Guitart, J. Cutaneous keratoacanthomas/squamous cell carcinomas associated
with neutralization of transforming growth factor β by the monoclonal antibody fresolimumab (GC1008).
Cancer Immunol. Immunother. 2015, 64, 437–446. [CrossRef]
263. Tolcher, A.W.; Berlin, J.D.; Cosaert, J.; Kauh, J.; Chan, E.; Piha-Paul, S.A.; Amaya, A.; Tang, S.; Driscoll, K.;
Kimbung, R.; et al. A phase 1 study of anti-TGF-β receptor type-II monoclonal antibody LY3022859 in
patients with advanced solid tumors. Cancer Chemother. Pharmacol. 2017, 79, 673–680. [CrossRef]
264. Chang, Z.L.; Lorenzini, M.H.; Chen, X.; Tran, U.; Bangayan, N.J.; Chen, Y.Y. Rewiring T-cell responses to
soluble factors with chimeric antigen receptors. Nat. Chem. Biol. 2018, 14, 317–324. [CrossRef]
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access
article distributed under the terms and conditions of the Creative Commons Attribution
(CC BY) license (http://creativecommons.org/licenses/by/4.0/).