ArticlePDF AvailableLiterature Review

Roles of TGF‑β signalling pathway‑related lncRNAs in cancer (Review)

Authors:

Abstract

Long non-coding RNAs (lncRNAs) are a class of RNAs that are >200 nucleotides in length that do not have the ability to be translated into protein but are associated with numerous diseases, including cancer. The involvement of lncRNAs in the signalling of certain signalling pathways can promote tumour progression; these pathways include the transforming growth factor (TGF)-β signalling pathway, which is related to tumour development. The expression of lncRNAs in various tumour tissues is specific, and their interaction with the TGF-β signalling pathway indicates that they may serve as new tumour markers and therapeutic targets. The present review summarized the role of TGF-β pathway-associated lncRNAs in regulating tumorigenesis in different types of cancer and their effects on the TGF-β signalling pathway.
ONCOLOGY LETTERS 25: 107, 2023
Abstract. Long non‑coding RNAs (lncRNAs) are a class of
RNAs that are >200 nucleotides in length that do not have the
ability to be translated into protein but are associated with
numerous diseases, including cancer. The involvement of
lncRNAs in the signalling of certain signalling pathways can
promote tumour progression; these pathways include the trans‑
forming growth factor (TGF)‑β signalling pathway, which is
related to tumour development. The expression of lncRNAs
in various tumour tissues is specic, and their interaction with
the TGF‑β signalling pathway indicates that they may serve
as new tumour markers and therapeutic targets. The present
review summarized the role of TGF‑β pathway‑associated
lncRNAs in regulating tumorigenesis in different types of
cancer and their effects on the TGF‑β signalling pathway.
Contents
1. Introduction
2. Role of lncRNAs in cancer
3. TGF‑β signalling pathway
4. TGF‑β pathway‑related lncRNAs and CRC
5. TGFβ pathway‑related lncRNAs and hepatocellular
carcinoma (HCC)
6. TGF‑β pathway‑related lncRNAs and GC
7. T G F β pathway‑related lncRNAs and breast cancer
8. TGF‑β pathway‑related lncRNAs and lung cancer
9. T GFβ pathway‑related lncRNAs and other cancer types
10. Conclusions and future perspectives
1. Introduction
Long non‑coding RNAs (lncRNAs) are newly discovered
RNAs that are >200 nucleotides in length and are involved
in a variety of molecular regulatory processes, including
transcriptional and posttranscriptional regulation, protein
localisation and RNA interference (1‑3). Although the full
function of a number of lncRNAs is unknown, their role in
cancer is becoming increasingly clear (4,5).
The transforming growth factor (TGF)‑β signalling
pathway consists of multiple signalling proteins that control
a variety of cell functions, including proliferation, differen
tiation, apoptosis and survival (6). Its inactivation leads to a
variety of pathological states, including malignancy, immune
system disorder and inammatory responses (7). However, the
role of the TGF‑β pathway in carcinogenesis is complex, and it
exerts either tumour‑suppressive or tumour‑promoting effects
depending on the cellular environment (8). The complex
regulatory mechanisms of the TGF‑β pathway in cancer are
cur rently unknown.
There is growing evidence of the interaction between
the TGF‑β signalling pathway and lncRNAs in tumours and
several members of the TGF‑β signalling pathway have been
identied as targets of lncRNAs (9‑11). The present review
summarizes knowledge of crosstalk between the TGF‑β
signalling pathway and lncRNAs in cancer.
2. Role of lncRNAs in cancer
LncRNAs that participate in chromatin remodelling, tran
scriptional control, posttranscriptional processing, protein
modification and RNA degradation (12‑14). After the
discovery of the rst lncRNAs in 1990 (15), lncRNAs have
received increasing attention. Numerous lncRNAs participate
in the pathogenesis of different diseases (16); these include
lncRNA CDC6 in breast cancer (17), lncRNA OIN1 in
ovarian cancer (18) and lncRNA RP11‑567G11.1 in pancre
atic cancer (19). Owing to the development of sequencing
Roles of TGF‑β signalling pathway‑related
lncRNAs in cancer (Review)
ZHIZHONG HU, YITONG LIU, MEIQI LIU, YANG ZHANG* and CHENGKUN WANG*
Cancer Research Institute, Medical School, University of South China, Hengyang, Hunan 421001, P.R. China
Received September 19, 2022; Accepted January 13, 2023
DOI: 10.3892/ol.2023.13693
Correspondence to: Dr Yang Zhang or Dr Chengkun Wang,
Cancer Research Institute, Medical School, University of South
China, 28 Chang Sheng Xi Avenue, Hengyang, Hunan 421001,
P. R . C hi na
E‑mail: yangyang@usc.edu.cn
E‑mail: charleswzy@gmail.com
*Contributed equally
Abbreviations: lncRNA, long non‑coding RNA; TGF‑β,
transforming growth factor β; EMT, epithelial‑mesenchymal
transition; TNF, tumour necrosis factor; TRAF, receptor‑associated
factor; ERK, extracellular signal‑regulated kinase; CRC, colorectal
cancer; HCC, hepatocellular carcinoma; GC, gastric cancer;
NSCLC, non‑small cell lung cancer
Key wo rds: cancer, lncRNA, TGF‑β signalling pathway
HU et al: TGF‑β SIGNAL LI NG PATHWAY‑RE LAT ED lncRNAs I N CANCER
2
technology, lncRNAs have been found to serve an important
role in tumour cell proliferation, apoptosis, differentiation and
invasion (11,20).
lncRNAs are considered to be an important component of
cancer, but they play different roles in different types of cancer.
For instance, lncRNA FGD5‑AS1 accelerates cell proliferation
in pancreatic cancer by regulating the microRNA (miRNA
or miR)‑520a‑3p/KIAA1522 axis (21), high expression levels
of lncRNA PCAT1 are associated with drug resistance in
colorectal cancer (CRC) (22) and lncRNA LNMICC promotes
cervical cancer lymph node metastasis by reprogramming
fatty acid metabolism (23). High or low expression of lncRNAs
in tumours contributes to disease via multiple molecular
mechanisms and they have a variety of unique functions and
characteristics. Guide lncRNAs bind enzymatically active
protein complexes and direct them to target gene promoter
regions or genome‑specic loci (24). Scaffold lncRNAs build
a central platform to which multiple protein complexes attach,
thus guiding them to their designated locations (24). Decoy
lncRNAs activate or silence downstream target genes by
binding and interacting with transcription factors or repres
sors (25). In addition, lncRNAs are associated with a number
of key signalling pathways. Regardless of the position of
these lncRNAs in the signalling pathway, they serve different
functions. For example, Wei et al (26) found that lncRNA
MEG3 inhibits proliferation and metastasis of gastric cancer
(GC) cells via the TP53 (a tumour suppressor gene) signal
ling pathway. High levels of lncRNA p21 in thoracic aortic
aneurysms may be associated with regulating vascular smooth
muscle cell proliferation and apoptosis by activating the
TGF‑β signalling pathway (27).
lncRNAs are known to be involved in cellular physiological
and pathological processes (28). Therefore, lncRNAs are also
relevant for diagnosis, treatment and prognosis evaluation (29).
3. TGF‑β signalling pathway
The TGF‑β superfamily has numerous members, including
TGF‑β isoforms, bone morphogenetic protein, growth differ
entiation factors, activators, inhibitors and nodulins (30,31).
TGF has three receptor ligands, TGF‑β1, 2, and 3, which have
similar, but not identical, biological activities in vitro (32). The
TGF‑β signalling pathway consists of two distinct intracellular
pathways: SMAD‑dependent (known as the classical TGF‑β
pathway) and non‑SMAD‑dependent pathway (known as the
non‑classical TGF‑β pathway; Fig. 1) (32). By contrast with
other signalling pathways, the classical TGF‑β pathway is
widely evolved and distributed in a variety of organisms (from
drosophila and nematodes to mice and humans). Activated
TGF‑β is altered by binding to TGF‑β type II receptor
(TGFβR‑II), which affects its structure, then TGFβR‑II
phosphorylates TGFβR‑I on specific serine and threonine
residues (33). In the classical pathway, the activated receptor
complex phosphorylates receptor‑SMADs (R‑SMADs;
including SMAD2 and SMAD3), which are primarily respon‑
sible for the activation of downstream signalling pathways (34).
The receptor‑activated SMAD anchor recruits R‑SMADs into
the activated receptor complex. Finally, the activated receptor
complex binds to SMAD4 (Co‑SMAD4 or common mediator
SMAD4) in a large complex and enters the nucleus, where it
interacts with transcription factors and coactivators to regulate
expression of target genes (34,35).
SMAD6 and SMAD7, also known as inhibitory SMADs
(I‑SMADs), serve an important role in the inhibition of the
TGF‑β signalling pathway through multiple mechanisms.
Firstly, SMAD6/7 competes with R‑SMADs for recruitment
to type I receptors and prevents activation of R‑SMADs by
phosphorylation (36). SMAD7 induces ubiquitination and
degradation of type I receptors by recruiting the E3 ligases
SMURF1 and SMURF2 (37,38). SMAD7 recruits ubiq
uitin‑conjugated E2 enzyme UbcH7 to stimulate SMURF1/2
activity in the R‑SMAD7SM‑URF1/2 complex (39). SMAD7
induces degradation and inactivation of TGFβR‑I by recruiting
two HECT‑type E3 ligases (WWP1/Tiul1 and NEDD4‑2) (40).
This suggests that I‑SMADs are involved in negative feedback
regulation in the TGF‑β/SMAD pathway. Although SMAD
proteins are the basis of TGF‑β regulation of various cellular
signalling pathways, numerous signalling responses are
stimulated by TGF‑β, which is not regulated by SMADs (32).
For example, in the non‑classical pathway, the activated
TGF‑β receptor complex promotes or inhibits downstream
cell biological processes through a number of other trans
duction factors, such as tumour necrosis factor (TNF), TNF
receptor‑associated factor 4 (TRAF4), TRAF6, p38 MAPK,
Ras homology (Rho), phosphoinositide 3 kinase (PI3K)/AKT,
extracellular signal‑regulated kinase (ERK) and NF‑κB, to
promote or inhibit downstream cell biological processes (32).
TGF‑β serves as both an oncogene and an oncogene
promoter. In normal tissue, TGF‑β promotes tissue stabilisa
tion and suppresses inammatory responses. In premalignant
progression, TGF‑β ser ves as an oncogene to promote apoptosis
and cytostasis and inhibit tumorigenesis. However, in cancer
cells, TGF‑β serves as a pro‑oncogene, promoting tumour
growth and metastasis (41,42). TGF‑β signalling promotes
epithelial‑mesenchymal transition (EMT) by increasing
expression of mesenchymal markers, such as N‑cadherin and
vimentin, and decreasing expression of epithelial markers, such
as E‑cadherin (43,44). Since TGF‑β acts extensively in cells,
blocking TGFβ and its downstream signals is a therapeutic
tool. Therefore, anti‑TGFβ signalling therapy is an additional
therapeutic tool along with the currently used CAR‑T (45) and
anti‑PD‑L1 (46) therapy.
4. TGF‑β pathway‑related lncRNAs and CRC
Aberrant lncRNAs in CRC are hypothesized to contribute
to activation or inactivation of the TGF‑β pathway to regu
late tumour development. TGF‑β pathway‑associated CRC
lncRNAs are discussed here, to explore the roles of lncRNAs
in the progression of CRC.
CRC is the third leading cause of cancer‑associated death
worldwide and there are 1.85 million new cases and 850,000
CRC‑associated deaths each year (47). The majority of CRC
tumours arise from precursor lesions, such as adenoma
transforming to adenocarcinoma (48). Therefore, it is key to
identify useful biomarkers to diagnose CRC at the early stages
of disease. Numerous studies have demonstrated the novel
role and therapeutic potential of lncRNAs in CRC (49,50)
(Fig. 2). lncRNA SNHG6 is upregulated in CRC and binds
UPF1 to activate the downstream TGF‑β/SMAD signalling
ONCOLOGY LETTERS 25: 107, 2023 3
pathway to promote proliferation, migration and invasion
of CRC cells (51). Upregulation of lncRNA LOC646329
promotes CRC cell proliferation by competing for binding
to miR‑29b‑1 (52). In addition, knockdown of lnc00858
reduces the proliferative capacity of CRC cells by inducing
production of p53 and blocking the G0/G1 phase of CRC
cells (53). lnc00858 upregulation is negatively correlated
with miR‑25‑3p and SMAD7 is a downstream target of
miR‑25‑3p (53). Similarly, miR‑93‑5p serves as an competing
endogenous RNA (ceRNA) for lncRNA CTBP1‑AS2 and
activates the TGF‑β/SMAD2/3 pathway to promote prolif
eration, invasion and resistance to apoptosis in colon cancer
cells (54). Shen et al (11) demonstrated that TGF‑β promotes
CRC metastasis via the lncRNA TUG1/TWIST1/EMT signal‑
ling pathway. TGF‑β induces metastasis, and knockdown
of TUG1can inhibit metastasis (11). However, expression of
TGF‑β does not increase after TUG1knockdown, suggesting
that TUG1is located downstream of TGF‑β. TUG1 may serve
as a drug target to inhibit CRC development by suppressing
TGF‑β pathway activation (11). Furthermore, Wu et al (49)
found that lnc00941 promotes EMT by directly competing with
β‑transducin repeats‑containing protein to bind to the MH2
structural domain on SMAD4, thereby preventing SMAD4
protein degradation and activating the TGF‑β/SMAD2/3
signalling pathway. lncRNA CASC9 is upregulated in CRC,
and high expression of CASC9 predicts a low prognosis and an
association with TNM stage I (55). Luo et al (55) demonstrated
that CASC9 enhances the function of the telomerase Reverse
Transcriptase (TERT) complex in CRC cells by regulating
expression of TGF‑β2 mRNA and upregulating levels of
TGF‑β2 and TERT, leading to phosphorylation of SMAD3 and
activation of the TGF‑β signalling pathway, thereby enhancing
its Tumorigenic ability.
Since colon cancer only shows symptoms in the advanced
stages of disease, it is necessary to improve the early detection
rate of CRC. An increasing number of studies have found that
TGF‑β/SMAD signalling pathway involvement with lncRNAs
serves an important role in the development of colon cancer,
which provides a new avenue for early diagnosis and treat
ment (49,51,55).
5. TGF‑β pathway‑related lncRNAs and hepatocellular
carcinoma (HCC)
HCC ranks fourth in the world for cancer‑associated
death (56). HCC is a common cancer with a poor prognosis
and high economic cost and disease burden; the 2019 Global
Cancer Report released by the World Health Organization
(WHO) (57) states that about 705 million people worldwide
currently suffer from liver cancer, with about 700,000 new
liver cancer patients each year (57). lncRNAs are involved in
the physiological and pathological processes of HCC cells (28)
(Fig. 2). Certain lncRNAs, such as lnc01278, SBF2‑AS1,
SNAI3‑AS1 and NORAD, have been shown to promote
proliferation, migration and invasion of HCC by partici
pating in the TGF‑β signalling pathway. Huang et al (58)
Figure 1. Classical and non‑classical TGF‑β signaling pathway. TGF‑β ligands bind to TGFβR‑II to modify its conformation and mediate its action. TGFβR‑II
phosphorylates TGFβR‑I on specic serine and threonine residues. In the classical pathway, the activated receptor complex phosphorylates receptor‑SMADs
(SMAD2 or SMAD3), forms a heterogeneous complex with SMAD4 and translocates to the nucleus, where it interacts with tra nscription factors, coactivators
or co‑repressors to regulate expression of target genes. In the non‑classical pathway, TGF‑β activat es MAPKs, N FκB, Ras, TRAF6, TAK1/p38/JNK and PI3Ks,
leading to biological effects. ERK, extracellular signal‑regulated kinase; TAK1, TGF‑β‑activated kinase 1; TF, transcription factor; TGFβR‑I/II, transforming
growth factorβ receptor type I/II; T NK1, tyrosine kinase non‑receptor 1; TRAF6, TNF receptor‑associated factor 4; p, phosphorylated.
HU et al: TGF‑β SIGNAL LI NG PATHWAY‑RE LAT ED lncRNAs I N CANCER
4
found that the lnc01278/miR‑1258/SMAD2/3 axis promotes
HCC metastasis. lnc01278 promotes the expression of the
SMAD2/3 target gene by silencing expression of miR‑1258.
Similarly, downregulation of lncRNA SBF2‑AS1 inhibits
proliferation and migration of HCC by regulating the
miR‑361‑5p/TG Fβ1 signalling pathway (59). SBF2‑AS1
promotes the expression of TGFβR‑I via sponge adsorption of
miR‑140‑5p and in turn promotes the migration and invasion
of HCC cells (60). In addition, SNAI3‑AS1 promotes prolif
eration and metastasis of HCC cells by regulating UPF1 and
activating the TGF‑β/SMAD pathway (61). Yang et al (62)
found that lncRNA NORAD is upregulated in HCC tissue
and that lncRNA NORAD may serve as a ceRNA to regulate
miR‑202‑5p, which promotes HCC progression by targeting
TGFβRs. TGF‑β1 is a positive upstream regulator of UCA1,
while UCA1 is a positive upstream regulator of HXK2, which
forms the TGF‑β1/UCA1/HXK2 axis to promote prolifera
tion of HCC cells (63). In addition, Dong et al (64) found that
downregulation of lncRNA MEG3 promotes HCC prolif
eration, migration and invasion through the upregulation of
TGF‑β1.
SMAD3 (an R‑SMAD) and SMAD4 (a co‑SMAD)
are key proteins involved in the classical TGF‑β signal
ling pathway. Chen et al (65) found that lnc00261 inhibits
SMAD3 expression and phosphorylation and that SMAD3
may be involved in transcriptional regulation in TGF‑β1
signalling. lnc00261 inhibits EMT in HCC cells by inacti
vating the TGFβ1/SMAD3 signalling pathway. In addition,
lncRNAs also participate in the TGF‑β pathway through
epigenetic modifications. Zhang et al (66) found that
lncRNA 34a recruits DNA methyltransferase 3α through
prohibitin‑2 to methylate promoters of miR‑34a and histone
deacetylase 1 to influence histone modification, thereby
inhibiting miR‑34a expression. miR‑34a targets SMAD4 and
downregulation the expression of downstream genes. In the
immune system, activation of TGF‑β signalling suppresses
recruitment of tumour‑inltrating lymphocytes, leading to
tumour immune escape. Wang et al (67) found that rela
tively high levels of lncRNA NNT‑AS1 are associated with
a decrease in the number of inltrating CD4+ lymphocytes
and that knockdown of lncRNA NNT‑AS1 decreases expres
sion of TGF‑β and TGFβR‑I in HCC cells. In conclusion,
lncRNA NNT‑AS1 impairs CD4+ T cell inltration in HCC
by activating the TGF‑β signalling pathway through a novel
mechanism.
Biomarkers useful for early HCC diagnosis are still
lacking and available serum biomarkers show low sensitivity
and specicity, such as α‑fetoprotein and des‑gamma‑carboxy
Figure 2. Molecula r mechanisms of TGF‑β signalling pathway involved in lncRNAs in hepatocellula r carcinoma, colorectal, gastric and breast cancer.
Oncogenic lncRNAs in hepatocellular carcinoma and colorectal, gastric and breast cancer activate the TGF‑β signalling pathway primarily by degrading and
activating the t hree major targets of the complex, SMAD2, SMAD3, and SMAD7, while cer tain lncRNAs may dir ectly regulate TGF‑β as well as T GFβR‑II and
TGFβR‑II, thereby affecting tumorigenesis. lncRNA, long non‑coding RNA; miR, microRNA; TGFβR‑I/II, transforming growth factor‑β receptor type I/II.
ONCOLOGY LETTERS 25: 107, 2023 5
prothrombin (68). TGF‑β signalling pathway‑associated
lncRNAs are typically upregulated in HCC and may be a novel
target for early screening.
6. TGF‑β pathway‑related lncRNAs and GC
GC is the fth most deadly cancer in the world (69). Globally,
1 million new cases of stomach cancer are diagnosed each
year (70). Oncogenic lncRNAs serve an important role in
regulating TGF‑β and are regulated in multiple ways (Fig. 2).
lnc00665 promotes cell proliferation, invasion and metas
tasis by activating the TGF‑β pathway in GC and silencing
Lnc00665 inhibits EMT and decreases the expression levels
of TGF‑β1, SMAD2 and α‑smooth muscle actin (SMA) (71).
Similarly, knockdown of Lnc00978 inhibits activation of the
TGF‑β/SMAD2 signalling pathway and thus inhibits cell cycle
progression, migration, invasion and proliferation and induces
apoptosis in GC cells (72). The differentiation of regulatory
T cells is associated with the TGF‑β signaling pathway (73).
Xiong et al (74) found that lncRNA POU3F3 activates the
TGF‑β signalling pathway by increasing phosphorylation of
SMAD2/3, thus increasing the number of regulatory T cells in
peripheral blood and leading to the proliferation of GC cells.
Huang et al (75) found that SGO1‑AS1 inhibits EMT and
metastasis by competitively binding TGF‑β1 and TGF‑β2 with
polypyrimidine tract binding protein, leading to a decrease in
TGF‑β. In addition, TGF‑β inhibits SGO1‑AS1 transcription
by forming a negative feedback loop to induce ZEB1 produc‑
tion. This SGO1‑AS1/TGF‑β/ZEB1 axis may provide a novel
means for cancer treatment. LncRNAs can also act as cofac
tors for SMAD. Sakai et al (76) identied the EMT‑associated
lncRNA ELIT1, which enhances SMAD promoter activity
via TGF‑β induction and recruiting SMAD3 to the promoter
region of its target gene. In addition, lncRNA MBNL2‑AS1
forms a ceRNA network with miR‑424‑5p and SMAD7,
inactivating the TGF‑β/EMT pathway and inhibiting GC cell
proliferation, migration and invasion (77).
lncRNAs regulate gene expression at genomic, transcrip
tomic and posttranscriptional levels and are recognized as
biomarkers and therapeutic targets for GC (Fig. 2) (77,78).
The TGF‑β signalling pathway is an important pathway that
promotes development of GC and studying the effect of the
interaction of this pathway with lncRNAs in the development
of GC may provide an important target for early diagnosis (71).
7. TG Fβ pathway‑related lncRNAs and breast cancer
Globally, breast cancer is the most frequently diagnosed cancer
in women and ranks second among causes of cancer‑related
deaths in women (79). Although breast cancer can be diag
nosed early and there are numerous treatments available, it is
typically lethal once it metastasises (80). Therefore, it is key to
nd clinically useful biomarkers present in the early stages of
breast cancer. Certain lncR NAs have been shown to promote the
development of breast cancer via the TGF‑β signalling pathway
(Fig. 2). For example, CASC2 (81) and CCAT2 (82) have been
shown to be tumour therapeutic targets by participating in
TGF‑β/SMAD2 signalling and thus promoting proliferation
and metastasis of breast cancer cells. lncRNA ROR knockdown
inhibits SMAD2 and α‑SMA expression and thus inactivates
the TGF‑β signalling pathway to inhibit tumour growth (83).
ARHGAP5‑AS1 induces a decrease in SMAD7 ubiquitination
and degradation by interacting with SMAD7, leading to a
decrease in SMAD7 binding to SMURF1 and SMURF2 (84).
In addition, ARHGAP5‑AS1 may inhibit the TGF‑β signal
ling pathway by stabilising SMAD7. ADAMTS9‑AS2 has
been shown to target downstream ribosomal protein L22 to
inhibit SMAD2 expression, thereby regulating the TGF‑β
signalling pathway, inhibiting cell cycle arrest in breast cancer
cells in vitro and suppressing tumour growth in vivo (85). Loss
of Merlin in breast cancer cells affects functional cellular
metabolism. Mota et al (86) found that the cooperative activity
of TGF‑β transcriptional effectors results in upregulation of
UCA1, which leads to a decrease in Merlin activity against
STAT3. Similarly, Bo et al (87) predicted that lnc00467 may
be involved in signalling pathways involved in peroxisomal
lipid metabolism and immunity via miR‑23b‑5p targeting
TGF‑β2. LncRNAs can also be involved in drug resistance.
Zhang et al (88) found that knockdown of lnc00894‑002 down‑
regulates miR‑200a‑3p and miR‑200b‑3p, upregulates TGF‑β2
and ZEB1 and is involved in the development of tamoxifen
resistance. LncRNA DCST1‑AS1 enhances TGF‑β/SAMD2
signalling in BT‑549 cells by targeting ANXA1 and promoting
EMT (89). Ren et al (90) discovered that SMAD2/3/4 binds to
the promoter site of HOTAIR and is directly transcribed by
HOTAIR, which provides a novel idea for treatment of breast
ca ncer.
Based on the established role of TGF‑β‑associated
lncRNAs in regulating cell proliferation, cell cycle, apoptosis
and other aspects of cell physiology, future studies should
evaluate the potential of these transcripts as therapeutic targets
for breast cancer.
8. TGF‑β pathway‑related lncRNAs and lung cancer
Lung cancer remains the leading cause of cancer‑associated
deaths worldwide (91). One of the reasons for the high
mortality rate of lung cancer is that it often progresses to
an advanced stage before it is diagnosed (92). Therefore,
it is key to determine the molecular mechanisms of lung
tumours and nd new molecular biomarkers for diagnosis
and treatment. Wang et al (93) found that lncRNA ANCR
inhibits non‑small cell lung cancer (NSCLC) cell migration
and invasion via downregulation of TGF‑β1 expression.
Similarly, Su et al (94) found that upregulation of lncRNA
GASL1 may inhibit tumour growth in NSCLC via down
regulation of TGF‑β1. NKILA expression is regulated by
the upstream TGF‑β signalling pathway and interferes with
the NF‑κB/Snail signalling pathway to inhibit migration
and invasion of NSCLC cells (95). Knockdown of SMASR
in lung cancer promotes phosphorylation of SMAD2/3,
thereby inducing EMT via the TGF‑β signalling pathway and
promoting migration and invasion of lung cancer cells (96).
XIST serves as a sponge to directly adsorb miR‑137 and
negatively regulate its expression. miR‑137 overexpres
sion inhibits proliferation and EMT in A549 and H1299
cells (97). In addition, Notch‑1 has been identied as a direct
gene target of miR‑137 (97). Similarly, lncRNA SOX2OT
overexpression serves as a ceRNA to adsorb miR‑104‑5p,
thereby regulating RAC1 expression and activating the
HU et al: TGF‑β SIGNAL LI NG PATHWAY‑RE LAT ED lncRNAs I N CANCER
6
TGF‑β/parathyroid hormone‑associated protein/RANKL
signalling pathway (98).
lncRNAs act as transcribed molecules. Shi et al (99)
demonstrated that E2F1 activates SNHG3 and promotes
NSCLC cell proliferation and migration via the TGF‑β and
IL‑6/JAK2/STAT3 pathways. Similarly, Zhu et al (100) found
that forkhead box P3 protein increases NSCLC cell stem
ness by activating Lnc01232 and thus regulating TGFβR‑I,
activating the TGF‑β signalling pathway and recruiting
IGF2BP2 to stabilise TGFβR‑I. This may provide a theoretical
basis for lncRNA‑based treatment of NSCLC. Furthermore,
upregulation of TBILA enhances RhoA activation by binding
to the SMAD transcription factor complex, which promotes
expression of human hair centre‑associated lymphoma (101).
Jiang et al (102) found that lncRNA HCP5 is induced by
TGF‑β and transcriptionally regulated by SMAD3 to promote
lung adenocarcinoma tumour growth and metastasis. In addi‑
tion, lncRNA LINP1 inhibits EMT in lung cancer cells by
suppressing the TGF‑β pathway (9).
In the aforementioned lung cancer studies, multiple
differentially expressed lncRNAs have been identied, some
of which activate the TGF‑β pathway to drive tumorigenesis,
while others inactivate the TGF‑β pathway to inhibit tumour
progression (Fig. 3). Further study of the role of lncRNAs in
the TGF‑β pathway may help develop molecular markers for
early diagnosis of lung cancer.
9. TGF‑ β pathway‑related lncRNAs and other cancer types
In thyroid cancer, lncRNA FOXD3‑AS1 serves as a sponge
to adsorb miR‑296‑5p and upregulate miR‑296‑5p expres
sion, which inhibits the migration and invasion of thyroid
cancer cells by inactivating the TGF‑β1/SMAD signalling
pathway (103). Zhao et al (104) found that ANRIL may
decrease expression of cyclin‑dependent kinase 4 by inhibiting
the TGF‑β/SMAD signalling pathway and promoting invasion
and metastasis of thyroid cancer cells. Similarly, silencing
SPRY4‑IT1 inhibits TGF‑β1 and phosphorylated SMAD2/3
levels, thereby inhibiting proliferation and migratory capacity
of thyroid cancer cells; knockdown of SPRY4‑IT1‑mediated
functions can be rescued by interference with TGF‑β1 (105).
In cervical cancer, knockdown of lncRNA NEF decreases
the expression of TGF‑β1, which inhibits the migration and
invasion of cervical cancer cells (106). In addition, miR‑665
serves as a ceRNA for lncRNA DANCR and targets TGFβR‑I
through the ERK/SMAD pathway to suppress the malignant
phenotype of cervical cancer cells, which may provide a
novel therapeutic strategy for cervical cancer treatment (107).
Similarly, lncRNA CTS enhances migration and invasive
ability of cervical cancer cells as well as TGF‑β1‑induce d
EMT (108). The expression of lncRNA CTS has a negative
correlation with miR‑505 expression and ZEB2 may act as the
target of miR‑505 (108). lncRNA CTS promotes cervical cell
migration and invasion via the miR‑505/ZEB2/TGF‑β/SMAD
axis (108).
In lymphoma, knockdown of lncRNA ANRIL may inhibit
proliferation and promote apoptosis of Burkitt's lymphoma
cells by regulating the TGF‑β1 signalling pathway (109).
In glioma, UAC1 promotes Slug expression and thus
participates in TGF‑β‑induced EMT by targeting miR‑1
and miR‑203a (110). In addition, p53 inhibits expression of
PVT1and thus inactivates the TGF‑β/SMAD pathway, inhib
iting the proliferation, migration and invasion of glioma cells,
inducing cell apoptosis and inhibiting tumour growth (111).
In endometr ial cancer, lncRNAs promote t umorigenesis and
metastasis via the MIR210HG/miR‑337‑3p/137‑HMGA2 axis,
which activates the TGF‑β/SMAD3 signalling pathway (112).
In prostate cancer, SNHG16 promotes proliferation
and migration of prostate cancer cells by targeting the
TGF‑βRII/SMAD axis (113).
In pancreatic cancer, knockdown of PVT1 inhibits cell
survival, adhesion, migration and invasion by suppressing
TGF‑β/SMAD2/3 signalling (114). These ndings reveal that
PVT1 may serve an oncogenic role in pancreatic cancer by
Figure 3. Molecular mechanisms of TGF‑β signalling pathway involved in lncRNAs in lung and other cancer types. Oncogenic lncRNAs in lung cancer
and other types of cancer activate the TGF‑β signa lling pathway primarily by degrading and activating the three major targets of the SMAD2, SMAD3 and
SMAD7, while certa in lncRNAs may directly regulate TGF‑β as well as TGFβR‑I and TGFβR‑II, thereby affecting tumor igenesis. lncRNA, long non‑coding
RNA; miR, microRNA; TGF‑β, transforming growth factor β; TGFβR‑I/I I, TGF‑β receptor type I/ II.
ONCOLOGY LETTERS 25: 107, 2023 7
Table I. lncRNAs associated with the TGF‑β signalling pathway in cancer.
Interaction with
First author, Expression TGF‑β
year lncRNA Cancer type pattern signalling Cancer phenotype Molecular mechanism (Refs.)
Wang et al, SNHG6 CRC Activation Promotes proliferation, invasion lncRNA SNHG6, UPF1 (51)
2019 and migration (protein), SMAD2/3
Javanmard et al, LOC646329 CRC Activation Promotes proliferation lncRNA LOC646329, (52)
2020 miR‑29b‑1, SMAD2/3
Zhan et al, LNC00858 CRC Repression Inhibits proliferation, promotes lnc00858, miR‑25‑3p, (53)
2020 apoptosis SMAD7
Li et al, CTBP1‑AS2 CRC Activation Promotes proliferation and LINCRNA CTBP1‑AS2, miR‑ (54)
2021 invasion, inhibits apoptosis 93‑5p, TGF‑β/SMAD2/3
Shen et al, TUG1 CRC Activation Promotes metastasis TGF‑β, lncRNA TUG1, (11)
2020 TWIST1
Wu et al, LNC00941 CRC Activation Promotes EMT lnc00941, SMAD4, TGF‑β/ (49)
2021 SMAD2/3
Luo et al, CASC9 CRC Activation Promotes proliferation, inhibits LINCRNA CASC9, TGF‑β2/ (55)
2019 apoptosis TERT, SMAD3
Huang et al, LINC01278 HCC Activation Promotes proliferation, LINC01278, miR‑1258, (58)
2020 migration, and invasion SMAD2/3
Wu et al, SBF2‑AS1 HCC Activation Promotes proliferation, LINCRNA SBF2‑AS1, (59,60)
2021; Li et al, migration, and invasion miR‑361‑5p, TGF‑β1,
2018 LINCRNA SBF2‑AS1 ,
miR‑140‑5p, TGFβR‑I
Li et al, SNAI3‑AS1 HCC Activation Promotes proliferation, LINCRNA SNAI3‑AS1, (61)
2019 migration, and invasion UPF1, SMAD7
Yang et al, NORAD HCC Activation Promotes proliferation, LINCRNA NORAD, (62)
2019 migration, and invasion miR‑2025p, SMAD2/3
Hu et al, UCA1 HCC Activation Promotes proliferation TGFβ, UCA1, HXK2 (63)
2018
Dong et al, MEG3 HCC Suppression Inhibits proliferation, MEG3, TGFβ1 (64)
2019 migration, and invasion
Chen et al, LNC00261 HCC Suppression Inhibits EMT and stem TGF‑β1, LINC00261, (65)
2022 cell‑like features SMAD3
Zhang et al, LNCRNA HCC Activation Promotes proliferation LNC34a, miR34a, (66)
2019 34a SMAD4
Wang et al, NNT‑AS1 HCC Activation Promotes CD4+ T cell LINCRNA‑NNT‑AS1, (67)
2020 inltration TGF‑β, TGFβR‑I, SMAD5
HU et al: TGF‑β SIGNAL LI NG PATHWAY‑RE LAT ED lncRNAs I N CANCER
8
Table I. Continued.
Interaction with
First author, Expression TGF‑β
year lncRNA Cancer type pattern signalling Cancer phenotype Molecular mechanism (Refs.)
Zhang et al, LNC00665 GC Activation Promotes proliferation, LINC00665, TGF‑β1, (71)
2021 invasion, and metastasis SMAD2, α‑SMA
Fu et al, LNC00978 GC Activation Promotes proliferation, LINC00978, TGFβ/ (72)
2018 invasion and metastasis, SMAD2
induces apoptosis
Xiong et al, POU3F3 GC Activation Promotes proliferation LINCRNA POU3F3, (74)
2015 TGF‐β/SMAD2/3
Huang et al, SGO1‑AS1 GC Suppression Inhibits EMT and LINCRNA SGO1‑AS1, (75)
2021 metastasis PTBP1, TGFβR‑I/II,
ZEB1
Sakai et al, ELIT‑1 GC Activation Promotes EMT progression LINCRNA ELIT‑1, (76)
2019 TGFβ/SMAD3
Su et al, MBNL1‑ GC Suppression Inhibits proliferation, LINCRNA MBNL1‑ (77)
2022 AS1 migration, and invasion AS1, miR‑424‑5p,
SMAD7
Zhang et al, CASC2 Breast Activation Promotes proliferation LINCRNA CASC2, (81)
2019 and metastasis TGFβ/SMAD2
Wu et al, CCAT2 Breast Activation Promotes proliferation LINCRNA CCAT2, (82)
2017 and metastasis TGFβ/SMAD2
Hou et al, ROR Breast Activation Promotes growth, migration, LINCRNA ROR, (83)
2018 and invasion TGFβ/SMAD2
Wang et al, ARHGAP5‑ Breast Suppression Inhibits migration LINCRNA ARHGAP5‑ (84)
2021 AS1 AS1, SMAD7
Ni et al, ADAMTS9‑ Breast Suppression Inhibits tumor growth, LINCRNA ADAMTS9‑ (85)
2021 AS2 promotes apoptosis and AS2, RPL22,
cell cycle arrest SMAD2
Mota et al, UCA1 Breast Activation Promotes aerobic MERLIN, SMAD2/ (86)
2018 glycolysis 3, UCA1
Bo et al, LNC00467 Breast Activation Promotes proliferation LINC00467, miR‑23b‑ (87)
2021 and metastasis 5p, TGF‑β2
Zhang et al, LNC00894‑ Breast Activation Promotes the development lnc00894‑002, miR‑200a/ (88)
2018 002 of tamoxifen resistance b‑3p, TGF‑β, ZEB1
Tang et al, DCST1‑ Breast Activation Promotes EMT and DCST1‑AS1, ANXA1, (89)
2020 AS1 chemoresistance TGF‑β/SMAD2
ONCOLOGY LETTERS 25: 107, 2023 9
Table I. Continued.
Interaction with
First author, Expression TGF‑β
year lncRNA Cancer type pattern signalling Cancer phenotype Molecular mechanism (Refs.)
Ren et al, HOTAIR Breast Activation Inhibits proliferation, HOTAIR, SMAD2/3/4 (90)
2018 migration, and invasion
Wang et al, ANCR Lung Suppression Inhibits migration and LINCRNA ANCR, (93)
2018 invasion TGF‑β1
Su et al, GASL1 Lung Suppression Inhibits tumor growth LINCRNA GASL1, (94)
2018 TGF‑β1
Lu et al, NKILA Lung Suppression Inhibits migration TGF‑β, LINCRNA (95)
2017 and invasion NKILA, NF‑κB
Xu et al, SMASR Lung Suppression Inhibits migration TGF‑β, SMAD2/3, (96)
2021 and invasion SMASR, TGFBR‑I
Wang et al, XIST Lung Suppression Inhibits proliferation LINCRNA XIST, miR‑ (97)
2018 and EMT 137, TGF‑β1
Ni et al, SOX2OT Lung Activation Promotes proliferation LINCRNA SOX2OT, (98)
2021 and metastasis miR‑194‑5p, RAC1,
TGF‑β
Shi et al, SNHG3 Lung Activation Promotes proliferation E2F1, LINCRNA (99)
2020 and migration SNHG3, TGF‑β
Zhu et al, LNC01232 Lung Activation Promotes proliferation FOXP3, LINC0123, (100)
2022 and migration IGF2BP2, TGFβR‑I
(mRNA)
Lu et al, TBILA Lung Activation Enhances the pro‑ TGFβ, TBILA, (101)
2018 survival pathway HGAL, RhoA
Jiang et al, HCP5 Lung Activation Promotes tumor growth TGF‑β/SMAD3, (102)
2019 and metastasis LINCRNA HCP5,
miR‑203, SNAI
Zhang et al, LINP1 Lung Suppression Inhibits proliferation, TGFβ1, LNCRNA (9)
2018 migration, and invasion LINP1
Chen et al, FOXD3‑ Thyroid Suppression Inhibits proliferation lncRNA FOXD3‑AS1, (103)
2020 AS1 and migration miR‑296‑5p, TGF‑β1/
SMADs
Zhao et al, ANRIL Thyroid Suppression Promoting invasion lncRNA ANRIL, TGF‑β/ (104)
2016 and metastasis SMADs, p15INK4B
Zhou et al, SPRY4‑ Thyroid Suppression Inhibits proliferation lncRNA SPRY4‑IT1, (105)
2018 IT1 and migration TGF‑β/SMAD
HU et al: TGF‑β SIGNAL LI NG PATHWAY‑RE LAT ED lncRNAs I N CANCER
10
Table I. Continued.
Interaction with
First author, Expression TGF‑β
year lncRNA Cancer type pattern signalling Cancer phenotype Molecular mechanism (Refs.)
Ju et al, NEF Cervical Suppression Inhibits migration LNCRNA NEF, (106)
2019 and invasion TGF‑β1
Cao et al, DANCR Cervical Suppression Inhibits migration LNCRNA DANCR, (107)
2019 and invasion miR‑665, TGFβR‑I,
ERK/SMAD
Feng et al, CTS Cervical Activation Promotes migration LNCRNA CTS, miR‑ (108)
2019 and invasion 505, ZEB2, TGF‑β/
SMAD
Mao et al, ANRIL Lymphoma Activation Promotes proliferation, lncRNA ANRIL, (109)
2021 inhibits apoptosis TGF‑β1
Li et al, UCA1 Glioma Activation Promotes EMT and UCA1, miR‑1, (110)
2018 stemness miR‑203a, slug
TGF‑β
Li et al, PVT1 Glioma Activation Promotes proliferation, P53, LINCRNA PVT1, (111)
2022 migration, invasion TGF‑β/SMAD
Ma et al, MIR210HG Endometrial Activation Promotes proliferation, LINCRNA MIR210HG, (112)
2021 migration, invasion, miR‑337‑3p/137,
and EMT HMGA2, TGF‑β/
SMAD3
Weng et al, SNHG16 Prostate Activation Promotes proliferation lncRNA SNHG16, (113)
2021 and migration miR‑373‑3p,
TGFβR‑II
Zhang et al, PVT1 Pancreatic Activation Promotes survival, adhesion, lncRNA PVT1, TGF‑β/ (114)
2018 migration and invasion SMAD
Papoutsoglou et al, MIR100HG Pancreatic Activation Promotes EMT and stemness TGFβ, MIR100HG (115)
2021
Zhou et al, LNC00462 Pancreatic Activation Promotes proliferation lnc00462, miR‑665, (116)
2018 and migration TGFβR‑I/TGFβR‑II,
SMAD2/3
Wu et al, PVT1 Ovarian Activation Promotes proliferation, LINCRNA PVT1, (117)
2021 inhibits apoptosis miR‑148a‑3p, AGO1,
TGF‑β
Huang et al, DANCR Ovarian Activation Promotes viability, migration, LINCRNA DANCR, (118)
2020 and invasion miR‑214, TGF‑β
ONCOLOGY LETTERS 25: 107, 2023 11
regulating EMT via the TGF‑β/SMAD pathway (114). In addi‑
tion, miR100HG controls the intensity of TGF‑β signalling via
the production of TGFβR‑I in tumours (115). Overexpression
of Lnc00462 increases expression levels of TGFβR‑I a nd
TGFβR‑II, thereby activating the SMAD2/3 pathway in
pancreatic cancer cells (116). miR‑665 is also a target of
lnc00462 (116). Taken together, these ndings indicate that
the lnc00462/miR‑665/TGFβR‑I/II regulatory network may
underlie the mechanism of pancreatic carcinogenesis.
In ovarian cancer, lncRNA PVT1 promotes tumour growth
and proliferation via the PVT1/miR‑148a‑3p/AGO1/TGF‑β
axis (117). In addition, DANCR is a sponge for miR‑214,
while KLF5 is a target of miR‑214 (118). Silencing DANCR
inhibits TGF‑β‑treated ovarian cancer cell viability, migration
and invasion via the miR‑214/KLF5 axis and induces apop
tosis (118).
In bladder cancer, lnc01451 directly targets LIN28 to
activate the TGF‑β/SMAD signalling pathway (119). In
terms of drug resistance, Zhuang et al (120) found that
gemcitabine‑induced aberrant TGF‑β1 regulation of the
LET/NF90/miR‑145 axis promotes urothelial bladder cancer
chemoresistance by enhancing cancer cell stemness.
In osteosarcoma (OS), high levels of lnc00174 form a
ceRNA network with miR‑378a‑3p/SSH2 and activate the
TGF‑β/SMAD pathway to promote OS cell proliferation (121).
The discovery of a large number of TGF‑β‑associated
lncRNAs, their extensive expression patterns in various types
of cancer (thyroid and cervical cancer, lymphoma, glioma and
endometrial, prostate, pancreatic, ovarian and bladder cancer)
and the biological properties that promote tumour cell prolif‑
eration, migration and invasion provides a novel basis for the
development of cancer diagnosis and therapy.
10. Conclusions and future perspectives
lncRNAs are differentially expressed in different tissue and
cells and are highly heterogeneous. They regulate gene expres‑
sion and intracellular homeostasis via multiple mechanisms,
including tumour cell proliferation, survival, migration and
genomic stability (122). The present review conrmed that
lncRNAs play an important role in tumour development,
similar to protein‑coding genes, and are associated with
multiple cellular signalling pathways. Although there are
multiple signalling pathways in tumours by which lncRNAs
may regulate cell proliferation, the TGF‑β signalling pathway
is widely distributed in tumours and serves a key role in the
development of different types of cancer (123). lncRNA tran‑
scription can activate or inhibit the TGF‑β signalling pathway
by interacting with other molecules in the cell, including
DNA, protein and RNA, to provide malignant transforma
tion signals. Thus, lncRNAs affect the pathology of different
cancer types (124,125). Table I lists lncRNAs associated with
the TGF‑β signalling pathway in cancer. In addition, these
lncRNAs may have different methods of targeting the TGF‑β
signalling pathway since they have high tissue and cell speci‑
city. These lncRNAs can also act in different cancer types
through the TGF‑β pathway. For example, lncRNA UCA1
promotes tumour cell proliferation and EMT in breast and
liver cancer and glioma (63,110). lncRNA PVT1 promotes
tumour cell proliferation in pancreatic and ovarian cancer and
Table I. Continued.
Interaction with
First author, Expression TGF‑β
year lncRNA Cancer type pattern signalling Cancer phenotype Molecular mechanism (Refs.)
Shi et al, LINC01451 Bladder Activation Promotes proliferation, LINC01451, LIN28, (119)
2021 invasion, and metastasis TGF‑β/SMAD
Zhuang et al, LET Bladder Activation Promotes chemoresistance TGFβ1, LINCRNA‑LET, (120)
2017 NF90, miR‑145
Zheng et al, LINC00174 Osteosarcoma Activation Promotes proliferation, LINC00174, miR‑378a‑ (121)
2021 invasion, and metastasis 3p, SSH2, TGF‑β/
SMAD
, downregulation; , upregulation; CRC, colorectal cancer; EMT, epithelial‑mesenchymal transition; GC, gastric cancer; HCC, heptocellular carcinoma; lncRNA, long non‑coding RNA; miR, microRNA;
TGF‑β, transforming growth factor β; TGFβR‑I/II, TGF‑β receptor type I/II.
HU et al: TGF‑β SIGNAL LI NG PATHWAY‑RE LAT ED lncRNAs I N CANCER
12
glioma (111,117). Although the same lncRNAs are involved
in the TGF‑β pathway in different cancer types, they act in
different ways, either directly targeting SMADs or forming a
ceRNA network with miRNAs, which makes clinical targeting
difcult (126). Overall, TGF‑β pathway‑associated lncRNAs
are differentially regulated in different types of cancer and
targeted therapy is a potential way to disrupt key signalling
pathways in tumour cells, such as the Wnt, Notch and TGF‑β
pathways, without compromising their essential functions in
normal tissue (49,126,127). The lncRNA network and TGF‑β
signalling pathway could reveal new cancer diagnosis and
treatment approaches.
Since the TGF‑β signalling pathway is related to tumour
development and metastasis, interfering with this cascade
via inhibitors may be a valuable strategy in tumour treatment
approaches. For example, SD‑208, an inhibitor of TGFβR‑I,
signicantly downregulates expression of miR‑135b, a key
tumour molecule, in SW‑48 colon cells and nude mice
implanted with tumours in situ (128). Han et al (129) found
that dexamethasone inhibits AKT and ERK phosphorylation
in colon cancer cells, leading to a decrease in cy61 expression,
which in turn blocks TGF‑β1‑induced migration. Similarly,
Koelink et al (130) found that 5‑aminosalicylic acid elimi
nates the TGF‑β1 cascade in HCT116 CRC cells and therefore
disrupts phosphorylation of downstream SMAD3. These
inhibitors or drugs act on an important molecular target in the
TGF‑β pathway, which affects the entire pathway. lncRNAs
only indirectly affect expression of certain related proteins in
the TGF‑β pathway and, to the best of our knowledge, little
is known about the potential involvement of lncRNAs in
direct regulation. For TGF‑β‑induced lncRNAs, inhibition of
TGF‑β expression may be a promising therapeutic approach.
Identifying these potential lncRNAs will provide a more
comprehensive understanding of regulation of the TGF‑β
pathway.
Acknowledgements
Not applicable.
Funding
The present study was supported by the Youth Program
of National Natural Science Foundation of China (grant
no. 81500169), the Hunan Provincial Groundbreaking
Platform Open Fund of University of South China (grant
no. 19K080) and the Student Research Learning and Innovative
Experimental Project of the University of South China (grant
nos. 20155760439 and X2019141).
Availability of data and materials
Not applicable.
Authors' contributions
ZH is responsible for writing the article. YL and ML revised the
manuscript for important intellectual content and constructed
gures. YZ and CW conceived the study. All authors have read and
approved the nal manuscript. Data authentication is not applicable.
Ethics approval and consent to participate
Not applicable.
Patient consent for publication
Not applicable.
Competing interests
The authors declare that they have no competing interests.
References
1. Dahariya S, Paddibhatla I, Kumar S, Raghuwanshi S, Pallepati A
and Gutti RK: Long non‑coding RNA: Classication, biogenesis
and functions in blood cells. Mol Immunol 112: 82‑92, 2019.
2. Zhang H, Chen Z, Wang X, Huang Z, He Z and Chen Y: Long
non‑coding RNA: A new player in cancer. J Hematol Oncol 6:
37, 201 3.
3. Bhat AA, Younes SN, Raza SS, Zarif L, Nisar S, Ahmed I, Mir R,
Kumar S, Sharawat SK, Hashem S, et al: Role of non‑coding
RNA networks in leukemia progression, metastasis and drug
resistance. Mol Cancer 19: 57, 2020.
4. Xu Y, Qiu M, Shen M, Dong S, Ye G, Shi X and Sun M: The
emerging regulatory roles of long non‑coding RNAs implicated
in cancer metabolism. Mol Ther 29: 2209‑2218, 2021.
5. Zhu D, Shi C, Jiang Y, Zhu K, Wang X and Feng W: Cisatracurium
inhibits the growth and induces apoptosis of ovarian cancer cells
by promoting lincRNA‑p21. Bioengineered 12: 1505‑1516, 2021.
6. Colak S and Ten Dijke P: Targeting TGF‑β signaling in cancer.
Trends Cancer 3: 56‑71, 2017.
7. Ikushima H and Miyazono K: TGF‑β signal transduction
spreading to a wider eld: A broad variety of mechanisms for
context‑dependent effects of TGF‑β. Cell Tissue Res 347: 37‑49,
2012.
8. Zhao M, Mishra L and Deng CX: The role of TGF‑β/SMAD4
signaling in cancer. Int J Biol Sci 14: 111‑123, 2018.
9. Zhang C, Hao Y, Wang Y, Xu J, Teng Y and Yang X:
TGF‑β/SMAD4‑regulated LncRNA‑LINP1 inhibits epithe
lial‑mesenchymal transition in lung cancer. Int J Biol Sci 14:
1715 ‑1723, 2 018.
10. Lu Z, Chen Z, Li Y, Wang J, Zhang Z, Che Y, Huang J, Sun S,
Mao S, Lei Y, et al: TGF‑β‑induced NKILA inhibits ESCC cell
migration and invasion through NF‑κB/MMP14 signaling. J Mol
Med (Berl) 96: 301‑313, 2018.
11. Shen X, Hu X, Mao J, Wu Y, Liu H, Shen J, Yu J and
Chen W: The long noncoding RNA TUG1 is required for
TGF‑β/TWIST1/EMT‑mediated metastasis in colorectal cancer
cells. Cell Death Dis 11: 65, 2020.
12. Tan YT, Lin JF, Li T, Li JJ, Xu RH and Ju HQ: LncRNA‑mediated
posttranslational modications and reprogramming of energy
metabolism in cancer. Cancer Commun (Lond) 41: 109‑120,
20 21.
13. Zhou B, Yang H, Yang C, Bao YL, Yang SM, Liu J and Xiao YF:
Translation of noncoding RNAs and cancer. Cancer Lett 497:
89 99, 2 021.
14. Peng WX, Koirala P and Mo YY: LncRNA‑mediated regulation
of cell signaling in cancer. Oncogene 36: 5661‑5667, 2017.
15. Brannan CI, Dees EC, Ingram RS and Tilghman SM: The
product of the H19 gene may function as an RNA. Mol Cell
Biol 10: 28‑36, 1990.
16. Kwok ZH and Tay Y: Long noncoding RNAs: Lincs between
human health and disease. Biochem Soc Trans 45: 805‑812, 2017.
17. Kong X, Duan Y, Sang Y, Li Y, Zhang H, Liang Y, Liu Y,
Zhang N and Yang Q: LncRNA‑CDC6 promotes breast cancer
progression and function as ceRNA to target CDC6 by sponging
microRNA‑215. J Cell Physiol 234: 9105‑9117, 2019.
18. Takeiwa T, Mitobe Y, Ikeda K, Hasegawa K, Horie K and
Inoue S: Long intergenic noncoding RNA promotes ovarian
cancer growth by modulating apoptosis‑related gene expression.
Int J Mol Sci 22: 11242, 2021.
19. Huang R, Nie W, Yao K and Chou J: Depletion of the lncRNA
RP11‑567G11.1 inhibits pancreatic cancer progression. Biomed
Pharmacother 112: 108685, 2019.
ONCOLOGY LETTERS 25: 107, 2023 13
20. Xu K, Cai Y, Zhang M, Zou H, Chang Z, Li D, Bai J, Xu J and
Li Y: Pan‑cancer characterization of expression and clinical
relevance of m6A‑related tissue‑elevated long non‑coding RNAs.
Mol Cancer 20: 31, 2021.
21. Lin J, Liao S, Liu Z, Li E, Wu X and Zeng W: LncRNA
FGD5‑AS1 accelerates cell proliferation in pancreatic cancer by
regulating miR‑520a‑3p/KIAA1522 axis. Cancer Biol Ther 22:
257‑266, 2021.
22. Sun H, Sun X, Zhang H, Yue A and Sun M: LncRNA‑PCAT1
controls the growth, metastasis and drug resistance of human
colon cancer cells. J BUON 25: 2180‑2185, 2020.
23. Shang C, Wang W, Liao Y, Chen Y, Liu T, Du Q, Huang J,
Liang Y, Liu J, Zhao Y, et al: LNMICC promotes nodal metas
tasis of cervical cancer by reprogramming fatty acid metabolism.
Cancer Re 78: 877‑890, 2018.
24. Bhan A, Soleimani M and Mandal SS: Long noncoding RNA and
cancer: A new paradigm. Cancer Res 77: 3965‑3981, 2017.
25. Ransohoff JD, Wei Y and Khavari PA: The functions and unique
features of long intergenic non‑coding RNA. Nat Rev Mol Cell
Biol 19: 143‑157, 2018.
26. Wei GH and Wang X: lncRNA MEG3 inhibit proliferation and
metastasis of gastric cancer via p53 signaling pathway. Eur Rev
Med Pharmacol Sci 21: 3850‑3856, 2017.
27. Hu W, Wang Z, Li Q, Wang J, Li L and Jiang G: Upregulation
of lincRNA‑p21 in thoracic aortic aneurysms is involved in the
regulation of proliferation and apoptosis of vascular smooth
muscle cells by activating TGF‑β1 signaling pathway. J Cell
Biochem 120: 4113‑4120, 2019.
28. Han M, Liao Z, Liu F, Chen X and Zhang B: Modulation of the
TGF‑β signaling pathway by long noncoding RNA in hepatocel‑
lular carcinoma. Biomark Res 8: 70, 2020.
29. Goyal B, Yadav SRM, Awasthee N, Gupta S, Kunnumakkara AB
and Gupta SC: Diagnostic, prognostic, and therapeutic signi
cance of long non‑coding RNA MALAT1 in cancer. Biochim
Biophys Acta Rev Cancer 1875: 188502, 2021.
30. Morikawa M, Derynck R and Miyazono K: TGF‑β and the TGF‑β
Family: Context‑dependent roles in cell and tissue physiology.
Cold Spring Harb Perspect Biol 8: a021873, 2016.
31. Heldin CH and Moustakas A: Signaling receptors for TGF‑β
family members. Cold Spring Harb Perspect Biol 8: a022053,
2016.
32. Syed V: TGF‑β signaling in cancer. J Cell Biochem 117:
1279‑1287, 2016.
33. Wrana JL, Attisano L, Wieser R, Ventura F and Massagué J:
Mechanism of activation of the TGF‑beta receptor. Nature 370:
341‑347, 1994.
34. Lan HY: Diverse roles of TGF‑β/Smads in renal brosis and
inammation. Int J Biol Sci 7: 1056‑1067, 2011.
35. Attisano L and Wrana JL: Smads as transcriptional co‑modula‑
tors. Curr Opin Cell Biol 12: 235‑243, 2000.
36. Miyazawa K and Miyazono K: Regulation of TGF‑β fa mi ly
signaling by inhibitory smads. Cold Spring Harb Perspect Biol 9:
a022095, 2017.
37. Kavsak P, Rasmussen RK, Causing CG, Bonni S, Zhu H,
Thomsen GH and Wrana JL: Smad7 binds to Smurf2 to form
an E3 ubiquitin ligase that targets the TGF beta receptor for
degradation. Mol Cell 6: 1365‑1375, 2000.
38. Ebisawa T, Fukuchi M, Murakami G, Chiba T, Tanaka K,
Imamura T and Miyazono K: Smurf1 interacts with transforming
growth factor‑beta type I receptor through Smad7 and induces
receptor degradation. J Biol Chem 276: 12477‑12480, 2001.
39. Ogunjimi AA, Briant DJ, Pece‑Barbara N, Le Roy C,
Di Guglielmo GM, Kavsak P, Rasmussen RK, Seet BT, Sicheri F
and Wrana JL: Regulation of Smurf2 ubiquitin ligase activity by
anchoring the E2 to the HECT domain. Mol Cell 19: 297‑308,
20 05.
40. Komuro A, Imamura T, Saitoh M, Yoshida Y, Yamori T,
Miyazono K and Miyazawa K: Negative regulation of trans
forming growth factor‑beta (TGF‑beta) signaling by WW
domain‑containing protein 1 (WWP1). Oncogene 23: 6914‑6923,
2004.
41. Zhang Y, Alexander PB and Wang XF: TGF‑β family signaling
in the control of cell proliferation and survival. Cold Spring Harb
Perspect Biol 9: a022145, 2017.
42. Peng D, Fu M, Wang M, Wei Y and Wei X: Targeting TGF‑β
signal transduction for brosis and cancer therapy. Molecular
Cancer 21: 104, 2022.
43. Moustakas A and Heldin CH: Signaling networks guiding
epithelial‑mesenchymal transitions during embryogenesis and
cancer progression. Cancer Sci 98: 1512‑1520, 2007.
44. Xie F, Ling L, van Dam H, Zhou F and Zhang L: TGF‑β signaling
in cancer metastasis. Acta Biochim Biophys Sin (Shanghai) 50:
121‑132, 2018 .
45. Ma S, Li X, Wang X, Cheng L, Li Z, Zhang C, Ye Z and Qian Q:
Current progress in CAR‑T cell therapy for solid tumors. Int
J Biol Sci 15: 2548‑2560, 2019.
46. Shen X and Zhao B: Efcacy of PD‑1 or PD‑L1 inhibitors and
PD‑L1 expression status in cancer: Meta‑analysis. BMJ 362:
k352 9, 2018.
47. Biller LH and Schrag D: Diagnosis and treatment of metastatic
colorectal cancer: A review. JAMA 325: 669‑685, 2021.
48. Harada S and Morlote D: Molecular pathology of colorectal
cancer. Adv Anat Pathol 27: 20‑26, 2020.
49. Wu N, Jiang M, Liu H, Chu Y, Wang D, Cao J, Wang Z, Xie X,
Han Y and Xu B: LINC00941 promotes CRC metastasis through
preventing SMAD4 protein degradation and activating the
TGF‑β/SMAD2/3 signaling pathway. Cell Death Differ 28:
219‑2 32, 2021.
50. Schwarzmueller L, Bril O, Vermeulen L and Léveillé N: Emerging
role and therapeutic potential of lncRNAs in colorectal cancer.
Cancers (Basel) 12: 3843, 2020.
51. Wang X, Lai Q, He J, Li Q, Ding J, Lan Z, Gu C, Yan Q, Fang Y,
Zhao X and Liu S: LncRNA SNHG6 promotes proliferation,
invasion and migration in colorectal cancer cells by activating
TGF‑beta/Smad signaling pathway via targeting UPF1 and
inducing EMT via regulation of ZEB1. Int J Med Sci 16: 51‑59,
2019.
52. Javanmard AR, Dokanehiifard S, Bohlooli M and Soltani BM:
LOC646329 long non‑coding RNA sponges miR‑29b‑1 and
regulates TGFβ signaling in colorectal cancer. J Cancer Res Clin
Oncol 146: 1205‑1215, 2020.
53. Zhan J, Tong J and Fu Q: Long non‑coding RNA LINC00858
promotes TP53‑wild‑type colorectal cancer progression by
regulating the microRNA‑25‑3p/SMAD7 axis. Oncol Rep 43:
1267‑1277, 2020.
54. Li Q, Yue W, Li M, Jiang Z, Hou Z, Liu W, Ma N, Gan W,
Li Y, Zhou T, et al: Downregulating Long Non‑coding RNAs
CTBP1‑AS2 inhibits colorectal cancer development by modu
lating the miR‑93‑5p/TGF‑β/SMAD2/3 pathway. Front Oncol 11:
626620, 2021.
55. Luo K, Geng J, Zhang Q, Xu Y, Zhou X, Huang Z, Shi KQ, Pan C
and Wu J: LncRNA CASC9 interacts with CPSF3 to regulate
TGF‑β signaling in colorectal cancer. J Exp Clin Cancer Res 38:
249, 2019.
56. Yang JD, Hainaut P, Gores GJ, Amadou A, Plymoth A and
Roberts LR: A global view of hepatocellular carcinoma: Trends,
risk, prevention and management. Nat Rev Gastroenterol
Hepatol 16: 589‑604, 2019.
57. Laube R, Sabih AH, Strasser SI, Lim L, Cigolini M and Liu K:
Palliative care in hepatocellular carcinoma. J Gastroenterol
Hepatol 36: 618‑628, 2021.
58. Huang WJ, Tian XP, Bi SX, Zhang SR, He TS, Song LY, Yun JP,
Zhou ZG, Yu RM and Li M: The β‑catenin/TCF‑4‑LINC0127
8‑miR‑1258‑Smad2/3 axis promotes hepatocellular carcinoma
metastasis. Oncogene 39: 4538‑4550, 2020.
59. Wu YH, Yu B, Chen WX, Ai X, Zhang W, Dong W and Shao YJ:
Downregulation of lncRNA SBF2‑AS1 inhibits hepatocel
lular carcinoma proliferation and migration by regulating the
miR‑361‑5p / TGF β1 signaling pathway. Aging (Albany NY) 13:
19260 ‑19271, 2021.
60. Li Y, Liu G, Li X, Dong H, Xiao W and Lu S: Long non‑coding
RNA SBF2‑AS1 promotes hepatocellular carcinoma progression
through regulation of miR‑140‑5p‑TGFBR1 pathway. Biochem
Biophys Res Commun 503: 2826‑2832, 2018.
61. Li Y, Guo D, Ren M, Zhao Y, Wang X, Chen Y, Liu Y, Lu G and
He S: Long non‑coding RNA SNAI3‑AS1 promotes the prolifera‑
tion and metastasis of hepatocellular carcinoma by regulating the
UPF1/Smad7 signalling pathway. J Cell Mol Med 23: 6271‑6282,
2019.
62. Yang X, Cai JB, Peng R, Wei CY, Lu JC, Gao C, Shen ZZ,
Zhang PF, Huang XY, Ke AW, et al: The long noncoding RNA
NORAD enhances the TGF‑β pathway to promote hepatocel
lular carcinoma progression by targeting miR‑202‑5p. J Cell
Physiol 234: 12051‑12060, 2019.
63. Hu ML, Wang XY and Chen WM: TGF‑β1 upregulates the
expression of lncRNA UCA1 and its downstream HXK2 to
promote the growth of hepatocellular carcinoma. Eur Rev Med
Pharmacol Sci 22: 4846‑4854, 2018.
HU et al: TGF‑β SIGNAL LI NG PATHWAY‑RE LAT ED lncRNAs I N CANCER
14
64. Dong H, Zhang Y, Xu Y, Ma R, Liu L, Luo C and Jiang W:
Downregulation of long non‑coding RNA MEG3 promotes
proliferation, migration, and invasion of human hepatocellular
carcinoma cells by upregulating TGF‑β1. Acta Biochim Biophys
Sin (Shanghai) 51: 645‑652, 2019.
65. Chen Z, Xiang L, Li L, Ou H, Fang Y, Xu Y, Liu Q, Hu Z,
Huang Y, Li X and Yang D: TGF‑β1 induced deficiency of
linc00261 promotes epithelial‑mesenchymal‑transition and
stemness of hepatocellular carcinoma via modulating SMAD3.
J Transl Med 20: 75, 2022.
66. Zhang L, Niu H, Ma J, Yuan BY, Chen YH, Zhuang Y,
Chen GW, Zeng ZC and Xiang ZL: The molecular mechanism of
LncRNA34a‑mediated regulation of bone metastasis in hepato
cellular carcinoma. Mol Cancer 18: 120, 2019.
67. Wang Y, Yang L, Dong X, Yang X, Zhang X, Liu Z, Zhao X and
Wen T: Overexpression of NNT‑AS1 activates TGF‑β signa ling
to decrease tumor CD4 lymphocyte inltration in hepatocellular
carcinoma. Biomed Res Int 2020: 8216541, 2020.
68. Tayob N, Kanwal F, Alsarraj A, Hernaez R and El‑Serag HB: The
performance of AFP, AFP‑3, DCP as biomarkers for detection of
hepatocellular carcinoma (HCC): A Phase 3 Biomarker Study in
the United States. Clin Gastroenterol Hepatol: Feb 3, 2022 doi:
10.1016/j.cgh.2022.01.047 (Epub ahead of print).
69. Smyth EC, Nilsson M, Grabsch HI, van Grieken NC and
Lordick F: Gastric cancer. Lancet 396: 635‑648, 2020.
70. Thrift AP and El‑Serag HB: Burden of gastric cancer. Clin
Gastroenterol Hepatol 18: 534‑542, 2020.
71. Zhang X and Wu J: LINC00665 promotes cell proliferation,
invasion, and metastasis by activating the TGF‑β pathway in
gastric cancer. Pathol Res Pract 224: 153492, 2021.
72. Fu M, Huang Z, Zang X, Pan L, Liang W, Chen J, Qian H,
Xu W, Jiang P and Zhang X: Long noncoding RNA LINC00978
promotes cancer growth and acts as a diagnostic biomarker in
gastric cancer. Cell Prolif 51: e12425, 2018.
73. Moreau JM, Velegraki M, Bolyard C, Rosenblum MD and Li Z:
Transforming growth factor‑β1 in regulatory T cell biology. Sci
Immunol 7: eabi4613, 2022.
74. Xiong G, Yang L, Chen Y and Fan Z: Linc‑POU3F3 promotes
cell proliferation in gastric cancer via increasing T‑reg distribu‑
tion. Am J Transl Res 7: 2262‑2269, 2015.
75. Huang D, Zhang K, Zheng W, Zhang R, Chen J, Du N, Xia Y,
Long Y, Gu Y, Xu J and Deng M: Long noncoding RNA
SGO1‑AS1 inactivates TGFβ signaling by facilitating TGFB1/2
mRNA decay and inhibits gastric carcinoma metastasis. J Exp
Clin Cancer Res 40: 342, 2021.
76. Sakai S, Ohhata T, Kitagawa K, Uchida C, Aoshima T, Niida H,
Suzuki T, Inoue Y, Miyazawa K and Kitagawa M: Long
Noncoding RNA ELIT‑1 Acts as a Smad3 cofactor to facilitate
TGFβ/Smad signaling and promote epithelial‑mesenchymal
transition. Cancer Res 79: 2821‑2838, 2019.
77. Su J, Chen D, Ruan Y, Tian Y, Lv K, Zhou X, Ying D and Lu Y:
LncRNA MBNL1‑AS1 represses gastric cancer progression via
the TGF‑β pathway by modulating miR‑424‑5p/Smad7 axis.
Bioengineered 13: 6978‑6995, 2022.
78. Li H, Wang M, Zhou H, Lu S and Zhang B: Long Noncoding
RNA EBLN3P promotes the progression of liver cancer via
alteration of microRNA‑144‑3p/DOCK4 signal. Cancer Manag
Res 12: 9339‑9349, 2020.
79. Fahad Ullah M: Breast cancer: Current perspectives on the
disease status. Adv Exp Med Biol 1152: 51‑64, 2019.
80. Januškevičienė I and Petrikaitė V: Heterogeneity of breast
cancer: The importance of interaction between different tumor
cell populations. Life Sci 239: 117009, 2019.
81. Zhang Y, Zhu M, Sun Y, Li W, Wang Y and Yu W: Upregulation
of lncRNA CASC2 suppresses cell proliferation and metastasis
of breast cancer via inactivation of the TGF‑β signaling pathway.
Oncol Res 27: 379‑387, 2019.
82. Wu ZJ, Li Y, Wu YZ, Wang Y, Nian WQ, Wang LL, Li LC,
Luo HL and Wang DL: Long non‑coding RNA CCAT2 promotes
the breast cancer growth and metastasis by regulating TGF‑β
signaling pathway. Eur Rev Med Pharmacol Sci 21: 706‑714,
2017.
83. Hou L, Tu J, Cheng F, Yang H, Yu F, Wang M, Liu J, Fan J and
Zhou G: Long noncoding RNA ROR promotes breast cancer by
regulating the TGF‑β pathway. Cancer Cell Inter national 18: 142,
2018.
84. Wang CL, Li JC, Zhou CX, Ma CN, Wang DF, Wo LL, He M,
Yin Q, He JR and Zhao Q: Long non‑co ding RNA ARHGAP5 ‑AS1
inhibits migration of breast cancer cell via stabilizing SMAD7
protein. Breast Cancer Res Treat 189: 607‑619, 2021.
85. Ni K, Huang Z, Zhu Y, Xue D, Jin Q, Zhang C and Gu C: The
lncRNA ADAMTS9‑AS2 regulates RPL22 to modulate TNBC
progression controlling the TGF‑β signaling pathway. Front
Oncol 11: 654472, 2021.
86. Mota MSV, Jackson WP, Bailey SK, Vayalil P, Landar A,
Rostas JW III, Mulekar MS, Samant RS and Shevde LA:
Deciency of tumor suppressor Merlin facilitates metabolic
adaptation by co‑operative engagement of SMAD‑Hippo
signaling in breast cancer. Carcinogenesis 39: 1165‑1175,
2018.
87. Bo H, Zhang W, Zhong X, Chen J, Liu Y, Cheong KL, Fan R and
Tang S: LINC00467, driven by copy number amplication and
DNA demethylation, is associated with oxidative lipid metabo‑
lism and immune inltration in breast cancer. Oxid Med Cell
Longev 2021: 4586319, 2021.
88. Zhang X, Wang M, Sun H, Zhu T and Wang X: Downregulation
of LINC00894‑002 contributes to tamoxifen resistance
by enhancing the TGF‑β signaling pathway. Biochemistry
(Mosc) 83: 603‑611, 2018.
89. Tang L, Chen Y, Chen H, Jiang P, Yan L, Mo D, Tang X and Yan F:
DCST1‑AS1 Promotes TGF‑β‑induced epithelial‑mesenchymal
transition and enhances chemoresistance in triple‑negative
breast cancer cells via ANXA1. Front Oncol 10: 280, 2020.
90. Ren Y, Jia HH, Xu YQ, Zhou X, Zhao XH, Wang YF, Song X,
Zhu ZY, Sun T, Dou Y, et al: Paracrine and epigenetic control
of CAF‑induced metastasis: The role of HOTAIR stimulated by
TGF‑ß1 secretion. Mol Cancer 17: 5, 2018.
91. Toumazis I, Bastani M, Han SS and Plevritis SK: Risk‑based
lung cancer screening: A systematic review. Lung Cancer 147:
154‑186, 2020.
92. Nooreldeen R and Bach H: Current and future development in
lung cancer diagnosis. Int J Mol Sci 22: 8661, 2021.
93. Wang S, Lan F and Xia Y: lncRA ANCR inhibits non‑small cell
lung cancer cell migration and invasion by inactivating TGF‑β
pathway. Med Sci Monit 24: 6002‑6009, 2018.
94. Su WZ and Yuan X: LncRNA GASL1 inhibits tumor growth of
non‑small cell lung cancer by inactivating TGF‑β pathway. Eur
Rev Med Pharmacol Sci 22: 7282‑7288, 2018.
95. Lu Z, Li Y, Wang J, Che Y, Sun S, Huang J, Chen Z and He J:
Long non‑coding RNA NKILA inhibits migration and invasion
of non‑small cell lung cancer via NF‑κB/Snail pathway. J Exp
Clin Cancer Res 36: 54, 2017.
96. Xu L, Liu W, Li T, Hu Y, Wang Y, Huang L, Wang Y, Shao S,
Liu X and Zhan Q: Long non‑coding RNA SMASR inhibits the
EMT by negatively regulating TGF‑β/Smad signaling pathway
in lung cancer. Oncogene 40: 3578‑3592, 2021.
97. 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 Biomarkers 220: 333‑342, 2018.
98. Ni J, Zhang X, Li J, Zheng Z, Zhang J, Zhao W and Liu L:
Tumour‑derived exosomal lncRNA‑SOX2OT promotes bone
metastasis of non‑small cell lung cancer by targeting the
miRNA‑194‑5p/RAC1 signalling axis in osteoclasts. Cell Death
Dis 12: 662, 2021.
99. Shi J, Li J, Yang S, Hu X, Chen J, Feng J, Shi T, He Y, Mei Z,
He W, et al: LncRNA SNHG3 is activated by E2F1 and promotes
proliferation and migration of non‑small‑cell lung cancer cells
through activating TGF‑β pathway and IL‑6/JAK2/STAT3
pathway. J Cell Physiol 235: 2891‑2900, 2020.
100. Zhu L, Liu Y, Tang H and Wang P: FOXP3 activated‑LI NC01232
accelerates the stemness of non‑small cell lung carcinoma by
activating TGF‑β signaling pathway and recruiting IGF2BP2 to
stabilize TGFBR1. Exp Cell Res 413: 113024, 2022.
101. Lu Z, Li Y, Che Y, Huang J, Sun S, Mao S, Lei Y, Li N, Sun N
and He J: The TGFβ‑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 432: 156‑168, 2018.
102. Jiang L, Wang R, Fang L, Ge X, Chen L, Zhou M, Zhou Y,
Xiong W, Hu Y, Tang X, et al: HCP5 is a SMAD3‑responsive
long non‑coding RNA that promotes lung adenocarcinoma
metastasis via miR‑203/SNAI axis. Theranostics 9: 2460‑2474,
2019.
103. Chen Y, Gao H and Li Y: Inhibition of LncRNA FOXD3‑AS1
suppresses the aggressive biological behaviors of thyroid cancer
via elevating miR‑296‑5p and inactivating TGF‑β1/Smads
signaling pathway. Mol Cell Endocrinol 500: 110634, 2020.
ONCOLOGY LETTERS 25: 107, 2023 15
104. Zhao JJ, Hao S, Wang LL, Hu CY, Zhang S, Guo LJ, Zhang G,
Gao B, Jiang Y, Tian WG and Luo DL: Long non‑coding RNA
ANRIL promotes the invasion and metastasis of thyroid cancer
cells through TGF‑β/Smad signaling pathway. Oncotarget 7:
57903‑57918, 2016.
105. Zhou H, Sun Z, Li S, Wang X and Zhou X: LncRNA SPRY4‑IT
was concerned with the poor prognosis and contributed to the
progression of thyroid cancer. Cancer Gene Ther 25: 39‑46,
2018.
106. Ju W, Luo X and Zhang N: LncRNA NEF inhibits migration and
invasion of HPV‑negative cervical squamous cell carcinoma by
inhibiting TGF‑β pathway. Biosci Rep 39: BSR20180878, 2019.
10 7. Cao L, Jin H, Zheng Y, Mao Y, Fu Z, Li X and Dong L:
DANCR‑mediated microRNA‑665 regulates proliferation and
metastasis of cervical cancer through the ERK/SMAD pathway.
Cancer Sci 110: 913‑925, 2019.
108. Feng S, Liu W, Bai X, Pan W, Jia Z, Zhang S, Zhu Y and Tan W:
LncRNA‑CTS promotes metastasis and epithelial‑to‑mesen
chymal transition through regulating miR‑505/ZEB2 axis in
cervical cancer. Cancer Lett 465: 105‑117, 2019.
109. Mao S, Jin J, Li Z and Yang W: Knockdown of long non‑coding
RNA ANRIL inhibits the proliferation and promotes the apop‑
tosis of Burkitt lymphoma cells through the TGF‑β1 signaling
pathway. Mol Med Rep 23: 146, 2021.
110. Li Z, Liu H, Zhong Q, Wu J and Tang Z: LncRNA UCA1 is
necessary for TGF‑β‑induced epithelial‑mesenchymal transi
tion and stemness via acting as a ceRNA for Slug in glioma
cells. FEBS Open Bio 8: 1855‑1865, 2018.
111. Li Z, Li M, Xia P, Wang L and Lu Z: Targeting long non‑coding
RNA PVT1/TGF‑β/Smad by p53 prevents glioma progression.
Cancer Biol Ther 23: 225‑233, 2022.
112. Ma J, Kong FF, Yang D, Yang H, Wang C, Cong R and Ma XX:
lncRNA MIR210HG promotes the progression of endometrial
cancer by sponging mi R‑337‑3p/137 via the HMGA2‑TGF‑β/Wnt
pathway. Mol Ther Nucleic Acids 24: 905‑922, 2021.
113. Weng W, Liu C, Li G, Ruan Q, Li H, Lin N and Chen G: Long
non‑coding RNA SNHG16 functions as a tumor activator
by sponging miR‑373‑3p to regulate the TGFβ‑R2 /SMAD
pathway in prostate cancer. Mol Med Rep 24: 843, 2021.
114. Zhang X, Feng W, Zhang J, Ge L, Zhang Y, Jiang X, Peng W,
Wang D, Gong A and Xu M: Long non‑coding RNA
PVT1 promotes epithelial‑mesenchymal transition via the
TGF‑β/Smad pathway in pancreatic cancer cells. Oncol Rep 40:
1093 ‑1102, 2018.
115. Papoutsoglou P, Rodrigues‑Junior DM, Morén A, Bergman A,
Pontén F, Coulouarn C, Caja L, Heldin CH and Moustakas A:
The noncoding MIR100HG RNA enhances the autocrine func‑
tion of transforming growth factor β signaling. Oncogene 40:
3748 ‑376 5, 2 021.
116. Zhou B, Guo W, Sun C, Zhang B and Zheng F: Linc00462
promotes pancreatic cancer invasiveness through the
miR‑665/TGFBR1‑TGFBR2/SMAD2/3 pathway. Cell Death
Dis 9: 706, 2018.
117. Wu Y, Gu W, Han X and Jin Z: LncRNA PVT1 promotes the
progression of ovarian cancer by activating TGF‑β pathway via
miR‑148a‑3p/AGO1 axis. J Cell Mol Med 25: 8229‑8243, 2021.
118. Huang P, Qi B, Yao H, Zhang L, Li Y and Li Q: Knockdown of
DANCR suppressed the biological behaviors of ovarian cancer
cells treated with transforming growth factor‑β (TGF‑β) by
sponging MiR‑214. Med Sci Monit 26: e922760, 2020.
119. Shi H, Xie J, Wang K, Li W, Yin L, Wang G, Wu Z, Ni J, Mao W,
Guo C and Peng B: LINC01451 drives epithelial‑mesenchymal
transition and progression in bladder cancer cells via
LIN28/TGF‑β/Smad pathway. Cell Signal 81: 109932, 2021.
120. Zhuang J, Shen L, Yang L, Huang X, Lu Q, Cui Y, Zheng X,
Zhao X, Zhang D, Huang R, et al: TGFβ1 promotes gemcitabine
resistance through regulating the LncRNA‑LET/NF90/miR‑145
signaling axis in bladder cancer. Theranostics 7: 3053‑3067,
2017.
121. Zheng C, Li R, Zheng S, Fang H, Xu M and Zhong L: LINC00174
facilitates cell proliferation, cell migration and tumor growth of
osteosarcoma regulating the TGF‑β/SMAD signaling pathway
and upregulating SSH2 expression. Front Mol Biosci 8: 697773,
20 21.
122. Huarte M: The emerging role of lncRNAs in cancer. Nat Med 21:
1253‑1261, 2015.
123. Tzavlaki K and Moustak as A: TGF‑β signaling. Biomolecules 10:
487, 2020.
124. Lai XN, Li J, Tang LB, Chen WT, Zhang L and Xiong LX:
MiRNAs and LncRNAs: Dual roles in TGF‑β signaling‑regu
lated metastasis in lung cancer. Int J Mol Sci 21: 1193, 2020.
125. Fan Y, Shen B, Tan M, Mu X, Qin Y, Zhang F and Liu Y:
TGF‑β‑induced upregulation of malat1 promotes bladder cancer
metastasis by associating with suz12. Clin Cancer Res 20:
1531‑1541, 2014.
126. Zhang M, Weng W, Zhang Q, Wu Y, Ni S, Tan C, Xu M,
Sun H, Liu C, Wei P and Du X: The lncRNA NEAT1 activates
Wnt/β‑catenin signaling and promotes colorectal cancer progres
sion via interacting with DDX5. J Hematol Oncol 11: 113, 2018.
12 7. Tao S, Chen Q, Lin C and Dong H: Linc00514 promotes breast
cancer metastasis and M2 polarization of tumor‑associated
macrophages via Jagged1‑mediated notch signaling pathway.
J Exp Clin Cancer Res 39: 191, 2020.
128. Akbari A, Ghahremani MH, Mobini GR, Abastabar M,
Akhtari J, Bolhassani M and Heidari M: Down‑regulation
of miR‑135b in colon adenocarcinoma induced by a TGF‑β
receptor I kinase inhibitor (SD‑208). Iran J Basic Med Sci 18:
856861, 2015.
129. Han S, Bui NT, Ho MT, Kim YM, Cho M and Shin DB:
Dexamethasone inhibits TGF‑β1‑induced cell migration by
regulating the ERK and AKT pathways in human colon cancer
cells via CYR61. Cancer Res Treat 48: 1141‑1153, 2016.
130. Koelink PJ, Hawinkels LJAC, Wiercinska E, Sier CF, Ten Dijke P,
Lamers CB, Hommes DW and Verspaget HW: 5‑Aminosalicylic
acid inhibits TGF‑beta1 signalling in colorectal cancer cells.
Cancer Lett 287: 82‑90, 2010.
This work is licensed u nder a Creative Commons
Attribution-NonCommercial-NoDerivat ives 4.0
International (CC BY-NC-ND 4.0) License.
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Transforming growth factor β (TGF-β) has long been identified with its intensive involvement in early embryonic development and organogenesis, immune supervision, tissue repair, and adult homeostasis. The role of TGF-β in fibrosis and cancer is complex and sometimes even contradictory, exhibiting either inhibitory or promoting effects depending on the stage of the disease. Under pathological conditions, overexpressed TGF-β causes epithelial-mesenchymal transition (EMT), extracellular matrix (ECM) deposition, cancer-associated fibroblast (CAF) formation, which leads to fibrotic disease, and cancer. Given the critical role of TGF-β and its downstream molecules in the progression of fibrosis and cancers, therapeutics targeting TGF-β signaling appears to be a promising strategy. However, due to potential systemic cytotoxicity, the development of TGF-β therapeutics has lagged. In this review, we summarized the biological process of TGF-β, with its dual role in fibrosis and tumorigenesis, and the clinical application of TGF-β-targeting therapies.
Article
Full-text available
Studies over the past decades have implicated lncRNAs in promoting the development, migration and invasion of gastric cancer (GC). However, the role and mechanism of lncRNA MBNL1-AS1 in GC promotion are poorly understood. In this research, qRT-PCR showed that MBNL1-AS1 was down-regulated in GC tissues and cells. Cell experiments and the animal study demonstrated that MBNL1-AS1 knockdown accelerated GC cell proliferation, migration, and invasion, thus restraining cell apoptosis. Meanwhile, overexpression of MBNL1-AS1 repressed GC cell promotion. Bioinformatics analysis confirmed that MBNL1-AS1 binds to miR-424-5p via negative modulation. Rescue experiments showed that decreased miR-424-5p level inhibited GC cell promotion by silencing MBNL1-AS1. Furthermore, Smad7 was identified as a target of miR-424-5p that could reverse the promotion of GC cell growth mediated by miR-424-5p. Western blot results proved that MBNL1-AS1 affected TGF-β/SMAD pathways by regulating the miR-424-5p/Smad7 axis. Collectively, MBNL1-AS1 restrained GC growth via the miR-424-5p/Smad7 axis and thus could be a promising target for GC therapy. These findings illustrate that lncRNA MBNL1-AS1, as a tumor suppressor gene, participates in GC progression by regulating miR-424-5p/Smad7 axis, thus activating TGF-β/EMT pathways. The evidence may provide a potential marker for GC patients.
Article
Full-text available
Glioma is a primary intracranial malignant tumor with poor prognosis, and its pathogenesis is unclear. This study discussed the impact of p53/lncRNA plasmacytoma variant translocation 1 (lncRNA PVT1)/transforming growth factor beta (TGF-β)/Smad axis on the biological characteristics of glioma. Glioma and normal tissues were collected, in which relative lncRNA PVT1 and p53 expression was assessed. Pearson’s analysis was adopted for the correlation analysis between lncRNA PVT1 and p53. Short interfering RNA (siRNA) against lncRNA PVT1 (siRNA-PVT1), siRNA-p53 or both was transfected into the glioma cells to evaluate effects of lncRNA PVT1 and p53 on cell proliferation, migration, invasion, and apoptosis. Mouse xenograft model of glioma was established to verify function of lncRNA PVT1 and p53 in vivo. Relationship among p53, lncRNA PVT1 and TGF-β/Smad was predicted and confirmed. Glioma tissues and cells showed downregulated p53 expression and increased lncRNA PVT1 expression. An adverse relationship was noted between p53 expression and lncRNA PVT1 expression. p53 was shown to be enriched in the lncRNA PVT1 promoter region and resulted in its suppression. p53 inhibited glioma cell proliferation, migration, and invasion, and induced apoptosis as well as arrested tumor growth by downregulating lncRNA PVT1. LncRNA PVT1was found to bind to TGF-β and activate TGF-β/Smad pathway, promoting progression of glioma. Consequently, p53 exerts anti-oncogenic function on glioma development by suppressing lncRNA PVT1 and subsequently inactivating TGF-β/Smad pathway.
Article
Full-text available
Emerging evidence suggests that long non-coding RNAs (lncRNAs) play important roles in the metastasis and recurrence of hepatocellular carcinoma (HCC). A kinds of lncRNAs were found to be involved in regulating epithelial–mesenchymal transition (EMT) or stem-like traits in human cancers, however, the molecular mechanism and signaling pathways targeting EMT and stemness remains largely unknown. Previously, we found that linc00261 was down-regulated in HCC and associated with multiple worse clinical pathological parameters and poor prognosis. Here, we show that linc00261 was down-regulated in TGF-β1 stimulated cells, and forced expression of linc00261 attenuated EMT and stem-like traits in HCC. Linc00261 also inhibited the tumor sphere forming in vitro and decreased the tumorigenicity in vivo. Furthermore, we revealed that linc00261 suppressed the expression and phosphorylation of SMAD3 (p-SMAD3), which could be core transcriptional modulator in TGF-β1 signaling mediated EMT and the acquisition of stemness traits. A negative correlation between linc00261 and p-SMAD3 was determined in HCC samples. Conclusion: Our study revealed that linc00261 suppressed EMT and stem-like traits in HCC cells by inhibiting TGF-β1/SMAD3 signaling.
Article
Full-text available
Breast cancer (BRCA) is a malignant tumor with a high incidence and poor prognosis in females. However, its pathogenesis remains unclear. In this study, based on bioinformatic analysis, we found that LINC00467 was highly expressed in BRCA and was associated with tumor metastasis and poor prognosis. The genomic and epigenetic analysis showed that LINC00467 may also be regulated by copy number amplification (CNA), chromatin openness, and DNA methylation. In vitro experiments showed that it could promote the proliferation, migration, and invasion of BRCA cells. Competitive endogenous RNA (ceRNA) regulatory network analysis and weighted gene coexpression network analysis (WGCNA) suggested that LINC00467 may play a role in signaling pathways of peroxisomal lipid metabolism, immunity, and others through microRNAs (miRNAs) targeting transforming growth factor beta 2 (TGFB2). In addition, copy number amplification and high expression of LINC00467 were associated with the low infiltration of CD8+ and CD4+ T cells. In conclusion, we found that LINC00467, driven by copy number amplification and DNA demethylation, may be a potential biomarker for the diagnosis and prognosis of BRCA and a tumor promoter acting as a potential therapeutic target for BRCA as well.
Article
Full-text available
Background Although thousands of long noncoding RNAs (lncRNAs) have been annotated, only a few lncRNAs have been characterized functionally. In this study, we aimed to identify novel lncRNAs involved in the progression of gastric carcinoma (GC) and explore their regulatory mechanisms and clinical significance in GC. Methods A lncRNA expression microarray was used to identify differential lncRNA expression profiles between paired GCs and adjacent normal mucosal tissues. Using the above method, the lncRNA SGO1-AS1 was selected for further study. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and in situ hybridization (ISH) were performed to detect SGO1-AS1 expression in GC tissues. Gain-of-function and loss-of-function analyses were performed to investigate the functions of SGO1-AS1 and its upstream and downstream regulatory mechanisms in vitro and in vivo. Results SGO1-AS1 was downregulated in gastric carcinoma tissues compared to adjacent normal tissues, and its downregulation was positively correlated with advanced clinical stage, metastasis status and poor patient prognosis. The functional experiments revealed that SGO1-AS1 inhibited GC cell invasion and metastasis in vitro and in vivo. Mechanistically, SGO1-AS1 facilitated TGFB1/2 mRNA decay by competitively binding the PTBP1 protein, resulting in reduced TGFβ production and, thus, preventing the epithelial-to-mesenchymal transition (EMT) and metastasis. In addition, in turn, TGFβ inhibited SGO1-AS1 transcription by inducing ZEB1. Thus, SGO1-AS1 and TGFβ form a double-negative feedback loop via ZEB1 to regulate the EMT and metastasis. Conclusions SGO1-AS1 functions as an endogenous inhibitor of the TGFβ pathway and suppresses gastric carcinoma metastasis, indicating a novel potential target for GC treatment.
Article
Full-text available
Patients with advanced ovarian cancer usually exhibit high mortality rates, thus more efficient therapeutic strategies are expected to be developed. Recent transcriptomic studies revealed that long intergenic noncoding RNAs (lincRNAs) can be a new class of molecular targets for cancer management, because lincRNAs likely exert tissue-specific activities compared with protein-coding genes or other noncoding RNAs. We here show that an unannotated lincRNA originated from chromosome 10q21 and designated as ovarian cancer long intergenic noncoding RNA 1 (OIN1), is often overexpressed in ovarian cancer tissues compared with normal ovaries as analyzed by RNA sequencing. OIN1 silencing by specific siRNAs significantly exerted proliferation inhibition and enhanced apoptosis in ovarian cancer cells. Notably, RNA sequencing showed that OIN1 expression was negatively correlated with the expression of apoptosis-related genes ras association domain family member 5 (RASSF5) and adenosine A1 receptor (ADORA1), which were upregulated by OIN1 knockdown in ovarian cancer cells. OIN1-specifc siRNA injection was effective to suppress in vivo tumor growth of ovarian cancer cells inoculated in immunodeficient mice. Taken together, OIN1 could function as a tumor-promoting lincRNA in ovarian cancer through modulating apoptosis and will be a potential molecular target for ovarian cancer management.
Article
Transforming growth factor–β1 (TGF-β1) is inextricably linked to regulatory T cell (T reg ) biology. However, precisely untangling the role for TGF-β1 in T reg differentiation and function is complicated by the pleiotropic and context-dependent activity of this cytokine and the multifaceted biology of T regs . Among CD4 ⁺ T cells, T regs are the major producers of latent TGF-β1 and are uniquely able to activate this cytokine via expression of cell surface docking receptor glycoprotein A repetitions predominant (GARP) and αv integrins. Although a preponderance of evidence indicates no essential roles for T reg -derived TGF-β1 in T reg immunosuppression, TGF-β1 signaling is crucial for T reg development in the thymus and periphery. Furthermore, active TGF-β1 instructs the differentiation of other T cell subsets, including T H 17 cells. Here, we will review TGF-β1 signaling in T reg development and function and discuss knowledge gaps, future research, and the TGF-β1/T reg axis in the context of cancer immunotherapy and fibrosis.
Article
Background: AFP, AFP L-3 and DCP in combination or in GALAD were tested for HCC surveillance in retrospective cohort and case-control studies. However, there is a paucity of prospective data and no phase 3 biomarker studies from North American populations. Methods: We conducted a prospective-specimen collection, retrospective-blinded-evaluation (PRoBE) cohort study in patients with cirrhosis enrolled in a 6-monthly surveillance with liver imaging and AFP. Blood samples were prospectively collected every 6 months and analyzed in a retrospective blinded fashion. True positive rate (TPR) and false positive rate (FPR) for any or early HCC were calculated within 6, 12 and 24 months of HCC diagnosis based on published thresholds for biomarkers individually, in combination and in GALAD and HES scores. We calculated the area under the receiver operating curve (AUROC) and estimated TPR based on an optimal threshold at a fixed FPR of 10%. Results: The analysis was conducted in a cohort of 534 patients; 50 developed HCC (68% early) and 484 controls with negative imaging. GALAD had the highest TPR (63.6, 73.8, 71.4% for all HCC, and 53.8, 63.3, 61.8 % for early HCC within 6, 12 and 24 months, respectively) but FPR of 21.5-22.9%. However, there were no differences in AUROC among GALAD, HES, AFP-3 or DCP. At a fixed 10% FPR, TPR for GALAD dropped (42.4, 45.2, 46.9%) and was not different from HES (36.4, 40.5, 40.8%) or AFP-3 alone (39.4, 45.2, 44.9%). Conclusions: In a prospective cohort phase 3 biomarker study, GALAD was associated with a considerable improvement in sensitivity for HCC detection but an increase in false positive results. GALAD performance was modest and not different from AFP-3 alone or HES.
Article
Non-small cell lung carcinoma (NSCLC) is one of the most common malignant tumors worldwide with high incidence and mortality. Long non-coding RNAs (lncRNAs) have been reported to affect human cancer progression. The present study aimed to investigate the regulatory role and mechanism of long intergenic non-protein coding RNA 1232 (LINC01232) in NSCLC cells. RT-qPCR results revealed that LINC01232 expression was high in NSCLC cells. Flow cytometry and sphere formation assays indicated that LINC01232 significantly promoted NSCLC cell stemness. Luciferase reporter assay and ChIP assay validated that forkhead box P3 (FOXP3) could bind to LINC01232 promoter and activate LINC01232 transcription. Further, LINC01232 was certified to activate TGF-β signaling pathway through regulating transforming growth factor beta receptor 1 (TGFBR1). After RIP and RNA pull down assays, insulin like growth factor 2 mRNA binding protein 2 (IGF2BP2) was proven as the RNA-binding protein (RBP) for LINC01232. LINC01232 promoted TGFBR1 mRNA stability via recruiting IGF2BP2. Subsequently, LINC01232 was verified to accelerate NSCLC cell stemness and induce macrophage M2 polarization via upregulating TGFBR1. Taken together, FOXP3 activated-LINC01232 accelerated NSCLC cell stemness by activating TGF-β signaling pathway and recruiting IGF2BP2 to stabilize TGFBR1, which might offer a rationale for lncRNA-based treatment to NSCLC.