ArticlePDF Available

Bmi1 drives hepatocarcinogenesis by repressing the TGFβ2/SMAD signalling axis

Authors:

Abstract and Figures

Bmi1 is overexpressed in one-third of hepatocellular carcinoma (HCC) patients and acts as an oncogene in hepatocarcinogenesis. However, the underlying mechanism is unclear. The role of TGFβ signalling in HCC is not well defined as well. Here, we report that TGFβ2 is a target of Bmi1 in HCC and has a tumour-suppressing role. In Bmi1-knockout mouse livers and HCC cell lines, TGFβ2/SMAD cascade proteins were upregulated. TGFβ2 expression was inversely correlated with Bmi1 expression in human and mouse HCC tissues. In vitro, Bmi1 knockdown activated TGFβ2/SMAD signalling and led to cell apoptosis via upregulation of p15 and p21. TGFβ2 inhibition rescued the inhibitory effect of Bmi1 knockdown on HCC cell survival, proliferation, and cell-cycle progression. In vivo, restoration of TGFβ2 expression blocked Bmi1/Ras-driven hepatocarcinogenesis in mice. Chromatin immunoprecipitation and luciferase reporter assays revealed that Bmi1 repressed TGFβ2 expression by binding to its promoter as a co-factor of polycomb repressor complex 1. Our findings elucidate the molecular mechanism underlying hepatic Bmi1-driven carcinogenesis and highlight the importance of TGFβ2 as a tumour suppressor in HCC development.
Content may be subject to copyright.
Oncogene
https://doi.org/10.1038/s41388-019-1043-8
ARTICLE
Bmi1 drives hepatocarcinogenesis by repressing the TGFβ2/SMAD
signalling axis
Bin Li1Yuyuan Chen1Fei Wang2Jun Guo1Wen Fu1Min Li3Qichang Zheng3Yong Liu1Lingling Fan4
Lei Li1Chuanrui Xu 1
Received: 13 January 2019 / Revised: 22 September 2019 / Accepted: 24 September 2019
© The Author(s), under exclusive licence to Springer Nature Limited 2019
Abstract
Bmi1 is overexpressed in one-third of hepatocellular carcinoma (HCC) patients and acts as an oncogene in
hepatocarcinogenesis. However, the underlying mechanism is unclear. The role of TGFβsignalling in HCC is not well
dened as well. Here, we report that TGFβ2 is a target of Bmi1 in HCC and has a tumour-suppressing role. In Bmi1-
knockout mouse livers and HCC cell lines, TGFβ2/SMAD cascade proteins were upregulated. TGFβ2 expression was
inversely correlated with Bmi1 expression in human and mouse HCC tissues. In vitro, Bmi1 knockdown activated TGFβ2/
SMAD signalling and led to cell apoptosis via upregulation of p15 and p21. TGFβ2 inhibition rescued the inhibitory effect
of Bmi1 knockdown on HCC cell survival, proliferation, and cell-cycle progression. In vivo, restoration of TGFβ2
expression blocked Bmi1/Ras-driven hepatocarcinogenesis in mice. Chromatin immunoprecipitation and luciferase reporter
assays revealed that Bmi1 repressed TGFβ2 expression by binding to its promoter as a co-factor of polycomb repressor
complex 1. Our ndings elucidate the molecular mechanism underlying hepatic Bmi1-driven carcinogenesis and highlight
the importance of TGFβ2 as a tumour suppressor in HCC development.
Introduction
Bmi1 is a member of the mammalian polycomb repressor
complex 1 (PRC1) and is involved in the regulation of
development, stem cell self-renewal, the cell cycle, and
senescence [14]. Bmi1 reportedly is overexpressed and
acts as an oncogene in multiple tumour types, including
breast cancer [5], colon carcinoma [6], non-small cell lung
cancer [7,8], glioblastoma [9], ovarian cancer [10], and
bladder cancer [11]. We and other groups have demon-
strated that Bmi1 is overexpressed and functions as an
oncogene in human hepatocellular carcinoma (HCC) [12
14]. Our previous studies demonstrated that Bmi1 repres-
sion by either virus-encapsulated shRNA or liposome-
delivered siRNA inhibits HCC growth in mice [15,16]. In
accordance herewith, Chiba et al. reported that Bmi1 is
overexpressed in human HCC cell lines, and knockdown
(KD) of Bmi1 diminishes the side populationin these
lines [17]. Together, these studies indicate that Bmi1 plays a
critical role in HCC and is a potential treatment target.
However, its molecular mechanism underlying HCC for-
mation and development remains unclear.
As a major member of the polycomb group protein
family, Bmi1 acts on its targets through repressive epige-
netic modication by ubiquitylating nucleosomal histone
H2A Lys119 [18]. Bmi1 regulates development and stem
cell self-renewal by repressing the INK4a/ARF locus,
which encodes p16INK4a and p14ARF (p19ARF in mice)
These authors contributed equally: Bin Li, Yuyuan Chen
*Chuanrui Xu
xcr@hust.edu.cn
1School of Pharmacy, Tongji Medical College, Huazhong
University of Science and Technology, Wuhan 430030, China
2Department of Biochemistry and Molecular Biology, School of
Basic Medicine, Huazhong University of Science and Technology,
Wuhan 430030, China
3Department of Hepatobiliary Surgery, Union Hospital, Tongji
Medical College, Huazhong University of Science and
Technology, Wuhan 430022, China
4Stem Cell Center, Union Hospital, Tongji Medical College,
Huazhong University of Science and Technology, Wuhan 430022,
China
Supplementary information The online version of this article (https://
doi.org/10.1038/s41388-019-1043-8) contains supplementary
material, which is available to authorized users.
1234567890();,:
1234567890();,:
[3,19,20]. In tumour development, Bmi1 functions in both
INK4a/ARF-dependent and -independent manners. Bmi1
represses INK4a/ARF in non-small cell lung cancer and
prostate cancer [21,22], but does not act on INK4a/ARF in
glioma and Ewing sarcoma [9,23]. In addition, Bmi1
reportedly inhibits either p16 or p19 [2,7]. Our previous
studies showed that Bmi1 drives HCC development inde-
pendent of INK4a/ARF [13,14]. Considering that Bmi1 is a
critical regulator in maintaining HCC growth, we specu-
lated that a hitherto uncovered signalling pathway mediates
hepatic carcinogenesis driven by Bmi1.
TGFβis a homodimer that exists in three isoforms in
mammalian cells: TGFβ1, TGFβ2, and TGFβ3[24,25]. All
TGFβligands transduce signal through the same receptor
signalling systems to activate downstream SMAD or BMP
signalling [25]. The three isoforms have non-redundant
roles in regulating cell growth, differentiation, matrix pro-
duction, and apoptosis [24]. In cancer, TGFβhas dual roles:
it inhibits tumour formation in the early stage and promotes
tumour metastasis in the advanced stage. Among the three
isoforms, TGFβ1 is the most widely investigated in many
cancer types [25]. However, there are few reports on the
role of TGFβ2 in cancer.
In this study, we screened potential targets of Bmi1 in
HCC cell lines and Bmi1-knockout (KO) mouse liver tis-
sues, and we found that TGFβ2 is a target of Bmi1 in HCC.
We conrmed the tumour-suppressor function of TGFβ2by
performing loss-of-function and gain-of-function experi-
ments in cell lines and mouse models. In addition, we
explored the mechanism underlying TGFβ2 repression by
Bmi1 in HCC cells. Our results uncover the mechanism by
which Bmi1 promotes hepatocarcinogenesis and illustrate
the signicant role of TGFβ/SMAD signalling in liver
cancer.
Results
TGFβ/SMAD signalling is regulated by Bmi1 in HCC
cells and in mice
To screen for targets of Bmi1 in HCC, we performed cDNA
microarray analyses of Bmi1-KO mouse liver tissues and
Bmi1-KD Hep3B and Huh7 cells. We chose Hep3B and
Huh7 because both cell lines exhibit an early-stage HCC
signature and thus are appropriate for the identication of
alterations in signalling during the onset stage of HCC [26].
We selected genes with a fold-change > 1.2 and a p-value <
0.05 in Bmi1-KO hepatocytes and KD HCC cells as com-
pared with control hepatocytes and HCC cells, respectively.
In Bmi1-KO hepatocytes, 927 genes were signicantly up-
or downregulated. In Bmi1-KD Huh7 and Hep3B cells, 493
genes were signicantly up- or downregulated. Gene
ontology and pathway enrichment analyses using the
Database for Annotation, Visualization and Integrated
Discovery system (DAVID) revealed that TGFβsignalling
was the only pathway enriched in both Bmi1-KO mouse
hepatocytes and Bmi1-KD Huh7 and Hep3B cells (Fig. 1a, b).
Focusing on genes involved in TGFβ/SMAD signalling, we
found that most genes in this pathway were upregulated in
both the Bmi1-KO hepatocytes and the Bmi1-KD HCC
cells (Fig. 1c, d, and Supplementary Table 1). Therefore,
and considering the fact that TGFβ/SMAD signalling acts as
abrakeon the cell cycle whereas Bmi1 promotes cell-
cycle progression in HCC, we speculated that TGFβ/SMAD
signalling might be repressed in Bmi1-induced HCC.
To conrm our speculation and determine which isoform
is the major target of Bmi1, we examined TGFβ1, TGFβ2,
and TGFβ3 levels in Bmi1-KD Hep3B and Huh7 cells.
qRT-PCR analysis showed that only TGFβ2 mRNA was
increased in Bmi1-KD Hep3B and Huh7 cells, consistent
with the microarray data (Fig. 2a). Western blotting
revealed that TGFβ2 protein, but not TGFβ1, was upregu-
lated in the Bmi1-KO cells (Fig. 2b). Increased TGFβ2
expression after Bmi1 KD was conrmed by immuno-
uorescence staining (Fig. 2c). Accordingly,
TGFβ2 secretion was increased in Bmi1-KD compared with
wild-type Hep3B and Huh7 cell-culture medium (Fig. 2d).
Collectively, these data suggested that Bmi1 regulates
TGFβ/SMAD signalling via repressing TGFβ2 in HCC
cells.
TGFβ2 is inversely correlated with Bmi1 in mouse
and human HCC tissues
We previously reported that Bmi1 is overexpressed in a
large fraction of human HCC and demonstrated its role as
HCC driver gene in a mouse model [13,27]. Given that
TGFβ2 is a target of Bmi1, we speculated that TGFβ2
would be repressed by Bmi1 in mouse and human HCC
tissues. To conrm this hypothesis, we evaluated TGFβ2
and Bmi1 expression in these tissues by immunohis-
tochemistry. In Bmi1/Ras-induced mouse HCC, Bmi1 was
highly expressed in tumour tissues and sporadically
expressed in adjacent tissues, whereas TGFβ2 was expres-
sed mainly in tumour-adjacent tissues, but seldom in tumour
tissues (Fig. 3a). Similarly, in Akt/Ras-induced mouse
HCC, in which Bmi1 is not the driver, but is required for
HCC formation [27], Bmi1 was more strongly expressed in
tumour tissues than in non-tumour tissues, whereas TGFβ2
was expressed at lower levels in tumour tissues than in non-
tumour tissues (Fig. 3b). The inverse correlation between
Bmi1 and TGFβ2 expression in both Bmi1/Ras and Akt/Ras
HCC tissues was conrmed by western blotting (Fig. 3c, d).
Next, we evaluated Bmi1 and TGFβ2 expression in
human HCC samples. In ten fresh human HCC tissues, both
B. Li et al.
western blotting and immunohistochemical staining
demonstrated that Bmi1 was highly expressed in tumour
versus adjacent tissues, whereas TGFβ2 was expressed at
lower levels in HCC than in adjacent tissues (Fig. 4a, b).
Likewise, p-Smad2 and p-Smad3 were expressed at lower
levels in tumour tissues than in adjacent tissues (Fig. 4b).
We analysed the correlation between Bmi1 and TGFβ2
expression using a HCC tissue microarray containing 75
pairs of HCC and adjacent tissues. Bmi1 was expressed in
41% (31/75) of HCC tissues, but in only 15% (11/75) of
adjacent tissues (Fig. 4c). TGFβ2 was expressed in 37%
(28/75) of HCC tissues and in 60% (45/75) of adjacent
tissues. These data conrmed that TGFβ2 and Bmi1
expression is inversely correlated in human HCC tissues,
corroborating a potential causal relation between Bmi1 and
TGFβ2 expression in HCC development.
Bmi1 blocks TGFβ2/SMAD signalling in HCC cells
In the canonical pathway, TGFβbinds to and activates its
receptors, subsequently activating downstream SMAD
proteins through phosphorylation and nuclear translocation
[25]. To conrm that TGFβ2/SMAD signalling is involved
in Bmi1-induced HCC, we examined SMAD protein levels
in Bmi1-KD HCC cells. Silencing of Bmi1 led to increased
TGFβ2 expression in both Hep3B and Huh7 cells (Fig. 5a).
Consequently, Smad2 and Smad3 phosphorylation was
increased. Smad4 showed evident nuclear translocation.
Immunostaining demonstrated increased Smad2 and Smad3
phosphorylation and nuclear translocation, and Smad4
nuclear localization (Fig. 5b). Co-immunoprecipitation
assays revealed that both Samd2 and Smad3 co-
precipitated with anti-Smad4 antibody in Bmi1-KD cells,
Fig. 1 Microarray analysis identies TGFβsignalling as a downstream
target of Bmi1 in HCC. aSignalling pathways with fold-change > 1.2
and p< 0.05 in Bmi1/mice (8-week-old female mice, whole-body
knockout) were enriched by DAVID analysis. bSignalling pathways
enriched in Bmi1-KD Hep3B and Huh7 cells as indicated by DAVID-
based analysis. cExpression of TGFβcascade genes in Bmi1-KO mice
analysed by microarray. dmRNA levels of TGFβcascade genes in
Bmi1-KD Hep3B and Huh7 cells. (red =upregulation, green =
downregulation)
Bmi1 drives hepatocarcinogenesis by repressing the TGFβ2/SMAD signalling axis
indicating that more Smad2 and Smad3 can bind Smad4
upon Bmi1 KD (Fig. 5c). Notably, Smad2/3 complex acti-
vates p21cip1 and p15INK4b in cooperation with C/EBPβand
Miz-1, respectively [25]. In line herewith, phosphorylation
and nuclear translocation of Smad2 and Smad3 led to
increased p21cip1 and p15INK4b expression in Bmi1-KD
Hep3B and Huh7 cells (Fig. 5a). Correspondingly, cyclin
D1 expression was reduced, suggesting that increased
p21cip1 and p15INK4b blocked cell-cycle progression. To
exclude possible off-target effects, we also knocked down
Fig. 2 Knockdown of Bmi1 leads to increased TGFβ2 in Hep3B and
Huh7 cells. Hep3B and Huh7 cells were treated with Bmi1 shRNA1
lentivirus for 3 days and then selected on puromycin for 2 days. Cells
were then collected and lysed for RNA or protein extraction, or used
for an immunouorescence assay. Medium was collected for the
detection of secreted TGFβ2. aRT-qPCR analysis of TGFβ1, TGFβ2,
and TGFβ3 in Bmi1-KD Hep3B and Huh7 cells. rRNA was used as a
control and data are expressed as the mean ± SD (n=4). **p< 0.01.
bWestern blot for Bmi1 and TGFβ2 expression in Bmi1-KD Hep3B
and Huh7 cells. cImmunouorescence of Bmi1 and TGFβ2 in Bmi1-
KD Hep3B and Huh7 cells. Nuclei were stained with DAPI. Scale bar
=50 μm. dLevels of secreted TGFβ2 detected by ELISA in Bmi1-KD
and WT cells. Data are expressed as the mean ± SD (n=3). **p< 0.01
B. Li et al.
Bmi1 using a Bmi1-siRNA pool of three siRNAs
(Bmi1 siRNA2) in Hep3B and Huh7 cells. As expected,
similar effects were observed in the two cell lines after
Bmi1 KD (Supplementary Fig. S1ac). In addition, we
analysed the expression of betaglycan (also known as TGFβ
receptor type III, TGFBR3) in Hep3B and Huh7 cells. Both
Hep3B and Huh7 cells express high levels of betaglycan in
comparison with HeLa or DU145 cells, suggesting that
TGFβ2 may activate SMAD signalling by binding beta-
glycan (Supplementary Fig. S2). Collectively, these data
indicated that Bmi1 repressed TGFβ2 and downstream
SMAD signalling in HCC cells.
TGFβ2/SMAD signalling suppresses
hepatocarcinogenesis by inhibiting cell proliferation
As Bmi1 does not act on INK4a/ARF in HCC cells [13,27],
we next investigated whether Bmi1 promotes HCC cell
proliferation via repression of TGFβ2 using gain- and loss-
of-function experiments. In Hep3B and Huh7 cells, siRNA-
mediated Bmi1 silencing led to cell death, whereas treat-
ment with the TGFβreceptor inhibitor LY2157299
increased the survival of Bmi1-siRNA-treated HCC cells,
indicating that Bmi1 promotes cell growth via repressing
TGFβsignalling in these cells (Fig. 6a). Simultaneous
siRNA-mediated TGFβ2 silencing restored the growth of
Huh7 and Hep3B cells inhibited by Bmi1 KD (Fig. 6b). KD
of both Bmi1 and TGFβ2 was conrmed by western blot-
ting (Fig. 6c). These data corroborated that TGFβ2is
involved in cell-growth inhibition. A BrdU incorporation
assay showed that proliferation was increased by TGFβ2
inhibition in Bmi1-KD HCC cells (Fig. 6d). FACS analysis
revealed that TGFβ2 inhibition suppressed Bmi1-KD-
induced apoptosis (Fig. 6e). Bmi1 is essential for main-
tenance of the tumour-initiating capability of HCC cells
[14]; we determined whether this effect is dependent on
TGFβ2. A colony-formation assay showed that colonies
were reduced in Bmi1-KD Hep3B and Huh7 cells, whereas
TGFβ2 suppression reversed this effect (Fig. 6f). Of note,
KD of TGFβ2 alone only slightly promoted survival,
growth, proliferation, and migration of Hep3B and Huh7
cells, possibly because TGFβ2 is repressed by Bmi1 in
these two cell lines. Again, we conrmed these results using
Bmi1-siRNA/shRNA (Bmi1 siRNA2/shRNA2) and/or
TGFβ2 siRNA/shRNA (TGFβ2 siRNA2/shRNA2) pools of
three siRNA/shRNA sequences to knock down Bmi1 and/or
TGFβ2 in Hep3B and Huh7 cells, respectively (Supple-
mentary Fig. S3af). TGFβ2 repression could rescue HCC
cell growth repressed by Bmi1 KD (Supplementary
Fig. S4). Inhibition of TGFβ2 with its inhibitor pirfenidone
rescued the growth Hep3B and Huh7 blocked by the Bmi1
inhibitor PTC-209 (Supplementary Fig. S5). Collectively,
Fig. 3 TGFβ2 protein levels
negatively correlate with Bmi1
expression in mouse HCC.
Expression of TGFβ2 and Bmi1
was determined in mouse
HCC tissues generated by
hydrodynamic injection of
Bmi1/Ras or Akt/Ras together
with transposase sleeping
beauty(SB) into FVB/N mice
(6-week-old female mice,
n=3 in each group).
aImmunohistochemical analysis
of Bmi1 and TGFβ2 expression
in Bmi1/Ras mouse HCC
tissues. bImmunohistochemical
analysis of Bmi1 and TGFβ2in
Akt/Ras mouse HCC tissues.
Red dotted lines outline HCC
nodules. Nindicates non-
tumour tissue and Tindicates
tumour tissues. c,dWestern blot
for Bmi1 and TGFβ2 expression
in Bmi1/Ras and Akt/Ras mouse
HCC tissues. Samples were
from three mice in each group.
Scale bar =50 μm
Bmi1 drives hepatocarcinogenesis by repressing the TGFβ2/SMAD signalling axis
these experiments demonstrated that TGFβ2 repressed HCC
cell proliferation and Bmi1 promoted HCC cell proliferation
via repressing TGFβ2.
As Bmi1 drives cell-cycle progression in many cancer
types, including HCC, we examined whether the stimula-
tory effect of Bmi1 on HCC cell-cycle progression depends
on TGFβ2 repression. Bmi1 KD by siRNA resulted in the
accumulation of Hep3B and Huh7 cells in G1 phase
(Supplementary Fig. S6a). Western blotting showed that p-
Smad2 and p-Samd3 as well as cyclins D and E were
reduced following Bmi1 KD (Supplementary Fig. S6b).
Accordingly, cell-cycle inhibitors p15, p21, and c-Myc
were increased. In contrast, inhibition of TGFβ2 by either
inhibitor or siRNA alleviated the G1 arrest in the Bmi1-KD
HCC cells (Supplementary Fig. S6a). p-Smad2 and p-
Smad3 as well as p15, p21, and c-Myc were decreased,
whereas cyclins D and E were increased (Supplementary
Fig. S6b).
TGFβ1 signals via the same downstream proteins and
contributes to the similar cellular mechanisms as TGFβ2.
TGFβ2 siRNA reportedly inhibits TGFβ1 expression to
some extent [28]. To rule out the possibility that TGFβ1
played a role in HCC cell survival and growth after both
Bmi1 and TGFβ2 KD, we evaluated TGFβ1 expression in
HCC cells treated with TGFβ2 siRNA and the effect of
TGFβ1 KD on Bmi1-KD HCC cells. In line with the
observation by Oh et al. [28], TGFβ2 siRNA slightly
reduced the level of TGFβ1 (Supplementary Fig. S7a). In
addition, cell growth in Hep3B and Huh7 cells after Bmi1
KD was promoted strongly by TGFβ2 KD, but not by
TGFβ1 KD (Supplementary Fig. S7b, c). These results
indicated that TGFβ2, not TGFβ1, mediates the inhibitory
role of Bmi1 KD in HCC cells. Together, these data indi-
cated that Bmi1 promotes cell-cycle progression via
repressing TGFβ2/SMAD signalling in HCC cells.
TGFβ2 is a direct target of Bmi1 in HCC cells
We next asked how Bmi1 represses TGFβ2 in HCC cells.
As a core protein of PRC1, Bmi1 generally acts on its target
genes as a transcriptional repressor [2932]. Therefore, we
investigated whether Bmi1 directly binds to the TGFβ2
promoter using luciferase reporter and chromatin immuno-
precipitation (ChIP) assays. Noma et al. reported that the
Fig. 4 TGFβ2 expression is
inversely correlated with Bmi1
in clinical HCC tissues. aBmi1
and TGFβ2 levels in ten human
HCC tissues determined by
western blotting. Prepresents
para-tumour tissue and T
represents tumour tissue. Note
that 50-kD TGFβ2 bands refer to
its full-length or latent form
(cleaved but still non-covalently
bound). bRepresentative images
of H&E staining and
immunohistochemical staining
of Bmi1 and TGFβ2 in ten
human HCC tissues. Scale bar
=20 μm. Nrepresents non-
tumour tissue and Trepresents
tumour tissue. Staining intensity
was calculated as integrated
optic density in the same area in
different sections using software
Image-Pro Plus and then
normalized to that in para-
tumour sections. #p< 0.05,
**p< 0.01. cRepresentative
images and statistics of
Bmi1 and TGFβ2 protein
levels determined by
immunohistochemical staining
using a human HCC tissue
microarray containing 75 paired
HCC and adjacent tissues
B. Li et al.
promoter sequence of TGFβ2 stretches from 1729 to +63,
with a core promoter sequence spanning from 508 to +63
that has transcriptional activity equivalent to that of the full
promoter [33]. To ensure luciferase expression, we cloned
both the full (2000 to +100, promoter 1) and the core
(508 to +63, promoter 2) TGFβ2 promoters into the
luciferase reporter pGL3 (Fig. 7a). Both promoter sequen-
ces effectively drove luciferase expression, consistent with
the observation of Noma et al. (Fig. 7b). Notably, KD of
Bmi1 in Huh7 cells signicantly increased luciferase
expression under both TGFβ2 promoters, whereas over-
expression of Bmi1 had the opposite effect (Fig. 7c). These
results suggested that Bmi1 binds to the TGFβ2 core
promoter region (promoter 2). To conrm this, we per-
formed a ChIP assay using antibodies against Bmi1 and
Ring1B. We used Ring1B because this is an another core
protein of PRC1 [3032]. We designed two primer pairs to
detect eluted TGFβ2 promoter DNA (Fig. 7d). As expected,
TGFβ2 promoter DNA was precipitated by antibodies
against both Bmi1 and Ring1B (Fig. 7e, f). Overexpression
of Bmi1 led to signicantly higher precipitation of TGFβ2
promoter DNA. These results conrmed that Bmi1 directly
binds to the TGFβ2 promoter together with Ring1B.
Together with the luciferase reporter assays, these experi-
ments indicated that Bmi1 protein binds to the TGFβ2
promoter via PRC1 and thus inhibits TGFβ2 transcription.
Fig. 5 Canonical TGFβ2/SMAD signalling is activated by silencing
Bmi1 in Hep3B and Huh7 cells. aWestern blot of TGFβ2/SMAD
pathway proteins in Hep3B and Huh7 cells after Bmi1 knockdown
with siRNA1. Proteins were extracted from Hep3B or Huh7 cells after
Bmi1 siRNA1 transfection for 48 h. Scramble siRNA (SC) was used as
a control. bImmunouorescence of p-Smad2, p-Smad3, and Smad4 in
Hep3B and Huh7 cells. p-Smad2, p-Smad3, and Smad4 were stained
red, and cell nuclei were counterstained with DAPI (blue). Scale
bar =20 μm. cWestern blot detection of Smad2 and Smad3 pre-
cipitated with Smad4 antibody. After 3 days of infection with
Bmi1 shRNA1 lentivirus and 2 days of puromycin selection, Hep3B
and Huh7 cells were collected and lysed. Cell lysates were incubated
with anti-Smad4 antibody for 2 h and then with protein A/G agarose at
4 °C overnight. Levels of Smad2, Smad3, and Smad4 were analyzed
by western blotting
Bmi1 drives hepatocarcinogenesis by repressing the TGFβ2/SMAD signalling axis
B. Li et al.
Ectopic expression of TGFβ2 inhibits Bmi1/Ras-
induced HCC in mice
The fact that Bmi1 together with Ras can induce HCC in
mice is direct evidence of the oncogenic role of Bmi1 in
hepatocarcinogenesis [13,27]. Thus, we next asked whether
repression of TGFβ2 is responsible for Bmi1-induced HCC
formation and progression. To address this question, we
examined whether restoration of TGFβ2 could block HCC
formation induced by Bmi1/Ras in mice. We cloned TGFβ2
into pT3-EF1a/Bmi1 downstream of Bmi1, with an inde-
pendent internal ribosome entry site (IRES) (Fig. 8a). We
injected FVB/N mice with the sleeping beauty transposase
together with Bmi1/Ras or Bmi1/TGFβ2/Ras and monitored
tumour formation (Fig. 8b). In the second week post
injection, western blotting showed that Bmi1 and Ras pro-
tein levels in Bmi1/TGFβ2/Ras mice were comparable to
those in Bmi1/Ras mice, whereas TGFβ2 was expressed in
Bmi1/TGFβ2/Ras-, but not in Bmi1/Ras-injected liver tis-
sues (Supplementary Fig. S8). After 20 weeks, Bmi1/Ras
injection resulted in multiple HCC nodules spread over the
liver surface, whereas Bmi1/TGFβ2/Ras-injected mice had
only sporadic HCC tumours on the liver surface (Fig. 8c).
Bmi1/TGFβ2/Ras-injected mice had similar body weights
as Bmi1/Ras mice, but their livers weighed less, indicating
reduced tumour burden in Bmi1/TGFβ2/Ras mice (Fig. 8d).
Quantitative analysis showed that each mouse in the Bmi1/
Ras group (n=8) had at least two tumours on the liver,
with an average tumour number of six and a largest tumour
volume of 1560 mm3(Fig. 8e). In contrast, only two mice in
the Bmi1/TGFβ2/Ras group (n=10) had tumours (four and
one, respectively), and the largest tumour volume was
17 mm3. In Bmi1/Ras-induced HCC, TGFβ2 and p-Smad2/
p-Smad3 were repressed, whereas cyclins D1 were robustly
expressed (Fig. 8f). In Bmi1/TGFβ2/Ras HCC, TGFβ2 and
p-Smad2/p-Smad3 levels were evidently higher and cyclin
D1 levels were lower than those in Bmi1/Ras tumours.
Slower tumour growth was conrmed by attenuated
Ki67 staining in Bmi1/TGFβ2/Ras HCC tissues. These
results suggested that the restoration of TGFβ2 blocked
hepatocarcinogenesis induced by Bmi1 and Ras. To conrm
that the effects of restoration of TGFβ2 were attributable to
Bmi1, we examined the effect of TGFβ2 silencing in Bmi1-
KO tumours using a xenograft tumour model. In sub-
cutaneous tumours established using Hep3B cells, shRNA-
mediated Bmi1 silencing signicantly inhibited tumour
growth, whereas KD of TGFβ2 alone had a slightly sti-
mulatory effect (Supplementary Fig. S9a). As expected, co-
infection with TGFβ2 shRNA lentivirus partially reversed
the inhibitory effect of Bmi1 repression on tumour growth.
Examination of tumour volumes and weights indicated that
TGFβ2 KD reversed the effect of Bmi1 KO on tumour
growth (Supplementary Fig. S9b, c). Western blotting
conrmed that both Bmi1 and TGFβ2 were repressed by
shRNA in those tumours (Supplementary Fig. S9d). These
results corroborated that increased TGFβ2 is responsible for
the inhibitory effect of Bmi1 KD on HCC growth and that
Bmi1 drives HCC by repressing TGFβ2.
Discussion
The current study demonstrated that elevated Bmi1
repressed TGFβ2 expression in HCC cells by directly
binding to its promoter, thus blocking its transcription.
Repression of TGFβ2 inhibited SMAD signalling and
resulted in cell-cycle progression and the propagation of
HCC cells. In mice, overexpression of TGFβ2 in the liver
signicantly suppressed hepatic carcinogenesis driven by
Bmi1. Therefore, our ndings provide robust evidence to
support that TGFβ2 is a direct target of Bmi1 in HCC and
that TGFβ/SMAD signalling functions as a brakeon
Bmi1-driven hepatic carcinogenesis (Supplementary Fig.
S10).
In this study, we focused on TGFβ2 and SMAD sig-
nalling for several reasons. First, we found that SMAD
signalling was elevated in both Bmi1-KO mouse hepato-
cytes and Bmi1-KD human HCC cell lines. Second, SMAD
signalling blocks cell-cycle progression, exactly opposite to
the role of Bmi1 in HCC. Third, Bmi1 negatively regulates
gene expression at the transcriptional level, whereas SMAD
proteins are activated mainly by phosphorylation. There-
fore, we speculated that Bmi1 represses the SMAD pathway
via transcriptional inhibition of TGFβs. Finally, only
TGFβ2 among the TGFβisoforms was repressed by Bmi1
(Fig. 2). Accordingly, the role of TGFβ2 in Bmi1-driven
HCC was examined in this study.
The rst signicance of this study is that we uncovered
an important target of Bmi1 in HCC. A number of studies
revealed that Bmi1 acts as an oncogene independently of its
ability to repress INK4a/ARF in some cancers, including
Fig. 6 Blocking TGFβ2 reverses the growth inhibition caused by
Bmi1 suppression. aCell viabilities of Hep3B and Huh7 cells after
transfection with Bmi1 siRNA1 and/or treatment with TGFβ2 inhibitor
LY2157299 for 48 h. bCell viabilities of Hep3B and Huh7 cells after
transfection with Bmi1 siRNA1 and/or TGFβ2 siRNA1 for 48 h.
cProtein levels of Bmi1 and TGFβ2 detected by western blotting in
Hep3B and Huh7 cells treated with Bmi1 and/or TGFβ2 siRNA1 for
48 h. dResults of BrdU assay to determine the proliferation of Hep3B
and Huh7 cells treated with Bmi1 siRNA1 and/or TGFβ2 siRNA1 for
48 h. eFACS analysis of apoptosis in Hep3B and Huh7 cells treated
with Bmi1 siRNA1 and/or TGFβ2 siRNA1 for 48 h. fColonies formed
by Hep3B and Huh7 cells infected with Bmi1 shRNA1 and/or
TGFβ2 shRNA1 lentivirus. Note that colonies formed by Bmi1-KD
and TGFβ2-KD cells were smaller than those formed by Bmi1 WT and
TGFβ2-KD cells, possibly because of the larger amount of virus used
for double knockdown in the former. Data are expressed as the mean ±
SD (n=4). * or #p< 0.05, **p< 0.01
Bmi1 drives hepatocarcinogenesis by repressing the TGFβ2/SMAD signalling axis
HCC [9,13,14,17,23,3437]. Therefore, there are other
targets involved in Bmi1-driven tumorigenesis that
remained to be identied. Our results from both Bmi1-KO
mice and Bmi1-KD HCC cell lines indicated that the levels
of TGFβ2 and downstream effectors were increased upon
Bmi1 suppression. Importantly, TGFβ2 repression restored
Fig. 7 Bmi1 inhibits TGFβ2
transcription in vitro by binding
to its promoter. aSchematic
illustration of the two luciferase
reporter constructs used.
bLuciferase activities in Huh7
cells transfected with luciferase
reporter constructs containing
entire or core promoter for 48 h.
cLuciferase activities in Huh7
cells transfected with luciferase
reporter plasmids and Bmi1-
siRNA or Bmi1-expression
plasmid for 48 h. dLocation of
the two primer pairs used for
ChIP on the TGFβ2 promoter.
e,fChIP assay results using
respectively Ring1B and Bmi1
antibodies to pull down TGFβ2
promoter DNA. Chromatin was
extracted from Huh7 cells
transfected with pT3-Bmi1 or
pT3. IgG and pT3 were used as
control antibody and empty
vector, respectively. Data are
expressed as mean ± SD (n=4).
*or#p< 0.05. ** or ##p< 0.01
B. Li et al.
HCC cell growth inhibited by Bmi1 silencing, whereas
repression of TGFβ2 alone did not affect HCC cell growth,
indicating that TGFβ2/SMAD signalling mediates the reg-
ulatory function of Bmi1 on HCC cell proliferation. Hence,
these data demonstrated that TGFβ2/SMAD proteins are
critical targets of Bmi1 in HCC.
Furthermore, our ndings explain how Bmi1 regulates
cell-cycle progression in HCC independent of INK4a/ARF.
Bmi1 drives hepatocarcinogenesis by repressing the TGFβ2/SMAD signalling axis
Bmi1 has a dual role in inducing tumorigenesis: it promotes
cell-cycle progression and inhibits apoptosis [4]. In the
canonical pathway, Bmi1 promotes cell-cycle progression
by repressing p16INK4A and blocks apoptosis by inhibiting
p14ARF [38]. However, how Bmi1 regulates cell-cycle
progression independent of INK4a/ARF in cancer remains
largely unknown. Our results showed that Bmi1 KD led to
cell-cycle arrest in G1, whereas repression of TGFβ2 could
relieve the blockade. Interestingly, TGFβs reportedly inhibit
cell proliferation by causing growth arrest in the G1 phase
[39]. TGFβ-induced G1 arrest has been attributed to the
inhibitory effect of TGFβon both the levels and activities of
G1 cyclins and various cyclin-dependent protein kinases
[40], as well as on cyclin-dependent kinase inhibitors,
including p21WAF1, p27KiPl, p16, and pl5INK4B [39,41,42].
The present study highlights the important role of TGFβ2in
controlling cell-cycle progression and helps understand how
Bmi1 regulates the cell cycle via TGFβ2 in HCC cells.
Based on these previous ndings and our observations, we
conclude that Bmi1 represses TGFβ2 and subsequent
SMAD proteins and hence, promotes cell-cycle progression
via multiple cell-cycle-regulatory pathways.
Interestingly, several recent reports revealed that Bmi1
acts on TGFβ/SMAD signalling in other cancer types.
Gargiulo et al. showed that Bmi1 controls glioblastoma
multiforme development by regulating TGFβ/BMP signal-
ling [43]. Kim et al. reported that Bmi1 suppresses senes-
cence and prolongs life span by inhibiting TGFβsignalling
in normal human oral keratinocytes [44]. In general, our
data are consistent with these studies in that Bmi1 drives
tumour formation by regulating TGFβsignalling. However,
we provided more robust evidence of the role of TGFβ2in
Bmi1-induced HCC. The HCC mouse model experiments
demonstrated that repression of TGFβ2 is essential for Bmi1
to promote cell-cycle progression and HCC cell prolifera-
tion. When TGFβ2 was restored, Bmi1 failed to induce
HCC formation. In addition, our study claried the reg-
ulatory action of Bmi1 on TGFβ2. Using ChIP and luci-
ferase assays, we demonstrated that Bmi1 directly binds to
the promoter of TGFβ2 to represses its transcription.
Our data showed that Bmi1 KD led to increased secre-
tion of TGFβ2, which may act in an autocrine and/or
paracrine manner. Of note, secreted TGFβs form homo-
dimers after cleavage and then bind to receptors either
directly or with the assistance of the accessory co-receptor
betaglycan [45,46]. Betaglycan is particular critical for
TGFβ2, which acts in both betaglycan-dependent and
-independent pathways. In the canonical pathway, TGFβ2
rst binds to betaglycan, then to its receptor Tβ-R2, which
recruits Tβ-R1. In the non-canonical pathway, TGFβ2 binds
to Tβ-R2/Tβ-R1 pair directly and activates SMAD signal-
ling [47,48]. One study reported that binding of TGFβ2to
Tβ-R2 requires co-expression of Tβ-RI or Tβ-RIII [49]. Our
results indicate that betaglycan is highly expressed in
Hep3B and Huh7 cells, suggesting that TGFβ2 may act in a
betaglycan-dependent manner in HCC cells. However, we
did not examine the impact of betaglycan deletion on
TGFβ2/SMAD signalling transduction. Whether TGFβ2/
SMAD signalling transduction depends on betaglycan in
HCC remains to be investigated in future.
Another important nding of our study is that the TGFβ/
SMAD axis functions as a tumour suppressor during HCC
development, which is contrary to the conclusion reached
by Dropmann et al. [50]. TGFβplays contradictory roles in
tumorigenesis: on the one hand, it suppresses tumour for-
mation; on the other hand, it promotes metastasis. In normal
and premalignant cells, TGFβenforces homoeostasis and
suppresses tumour formation through cell-autonomous
tumour-suppressive effects (cytostasis, differentiation, and
apoptosis) or effects from the micro-environment (sup-
pression of inammation and stroma-derived mitogens)
[25]. After tumour formation, cancer cells may lose TGFβ
tumour-suppressive responses and use TGFβto their
advantage to advance immune evasion, growth factor pro-
duction, transformation into an invasive phenotype, and
metastatic dissemination [25]. Dropmann et al. reported that
TGFβ2 was highly expressed in many HCC patients, and its
levels were reversely correlated with survival, demonstrat-
ing an oncogenic role of TGFβ2[50]. We observed that
TGFβ2 repression by Bmi1 promoted HCC formation,
whereas restoration of TGFβ2 expression blocked tumour
formation induced by Bmi1 and Ras. Conversely, KD of
TGFβ2 restored tumour growth inhibited by Bmi1 repres-
sion in HCC xenografts. Therefore, our study conrms that
TGFβ2 is a tumour suppressor in HCC, at least at the early
stages.
Fig. 8 TGFβ2 inhibits tumour formation induced by Bmi1 and Ras in
FVB/N mice. aSchematic illustration of the four constructs used for
hydrodynamic injections. bIllustration of the establishment of trans-
genic Bmi1/Ras and Bmi1/TGFβ2/Ras HCC mice via hydrodynamic
injection. In both groups, Bmi1, Bmi1-TGFβ2, and Ras plasmids are
injected at the same amount into each FVB/N mouse (female, 6 weeks
old, n=8 for the Bmi1/Ras group and n=10 for the Bmi1/TGFβ2/
Ras group). cGross morphological images of livers from Bmi1/Ras
and Bmi1/TGFβ2/Ras mice. dBody/liver weights of Bmi1/Ras and
Bmi1/TGFβ2/Ras mice. eTumour numbers and maximal tumour
volumes in Bmi1/Ras and Bmi1/TGFβ2/Ras mice. Data are expressed
as the mean ± SD (n=8 for the Bmi1/Ras group and n=10 for the
Bmi1/TGFβ2/Ras group). *p< 0.05, **p< 0.01. fHaematoxylin and
eosin and immunohistochemical staining of liver tissues in Bmi1/Ras
and Bmi1/TGFβ2/Ras mice. Statistical analysis was performed based
on the ratio of positively stained nuclei versus total nuclei or relative
staining intensity in the tumour tissues. Relative staining intensity was
calculated by determining the integrated optic density with Image-Pro
Plus in Bmi1/TGFβ2/Ras HCC tissues and then normalized to that in
Bmi1/Ras tissues. Data are the mean ± SD (n=3 mouse sections for
both Bmi1/Ras and Bmi1/TGFβ2/Ras groups). *p< 0.05, **p< 0.01.
Scale bar =20 μm. Nindicates non-tumour tissues and Tindicates
tumour tissues
B. Li et al.
Our study provides several useful hints for targeted
therapy against either Bmi1 or TGFβin HCC. First, our data
indicate that TGFβ2 functions as a tumour suppressor in
early-stage HCC, especially in Bmi1-driven HCC. Both
cell-culture and mouse studies supported its anti-tumour
role in HCC formation or growth. Therefore, monotherapy
targeting TGFβmay be not applicable to HCC patients in
early stages or with high Bmi1 levels, and might even
worsen rather than cure the cancer. Second, therapeutic
intervention blocking Bmi1 in HCC may enhance TGFβ2
expression and subsequent SMAD signalling activation. As
we have demonstrated, activated TGFβ2/SMAD signalling
blocks HCC cell growth. However, whether it would pro-
mote HCC cell metastasis later on remains unknown. Given
that TGFβ/SMAD also promotes metastasis, sequential
treatment with Bmi1 inhibitors followed by TGFβ2 inhibi-
tors would be worthy of exploring. Last, but not least, our
tissue microarray data showed that some HCC tissues
exhibit high TGFβ2 expression, in line with ndings in a
previous report [50]. However, the role of elevated TGFβ2
has not been clearly demonstrated in experimental or
mechanistic studies. Considering the dual roles of TGFβin
different contexts, whether therapies targeting TGFβ2 are
applicable in these patients requires more pre-clinical
studies.
Regretfully, we did not examine the long-term effects of
TGFβexpression in HCC progression and potential
metastasis, and thus failed to evaluate its role in advanced
HCC. We plan to evaluate the nal outcome of over-
expressing TGFβin HCC development and progression in a
follow-up study. In addition, we noticed that in some clin-
ical tissues, increased Bmi1 was not accompanied with
decreased TGFβ2. It is possible that TGFβ2 is also regu-
lated by factors other than Bmi1 considering that there are
plentiful known driver oncogenes and signalling molecules
in HCC. Conversely, Bmi1 may have other targets other
than TGFβ2 in HCC. To elucidate the roles and mechan-
isms of both Bmi1 and TGFβ2 in hepatocarcinogenesis
further, more clinical samples and mouse models will be
utilized in our future studies, and we also hope to uncover
novel functions of Bmi1 and TGFβsignalling in other liver
cancer types.
Materials and methods
Cell culture and lentiviral infection
Human Hep3B and Huh7 HCC cell lines were purchased
from China Center for Type Culture Collection at Wuhan
University, China. Both cell lines were authenticated using
STR proling and treated with plasmocin (25 μg/mL) for
2 weeks before used in this study. The cells were cultured in
DMEM supplemented with 10% foetal bovine serum at 37 °C.
Lentivirus expressing Bmi1 shRNA was transfected into
HCC cells to knock down Bmi1 expression. Three days
post infection, the cells were expanded and selected with
1μg/mL puromycin for 2 days, and then harvested for
protein and RNA extraction.
Constructs, siRNA, and transfection
The Bmi1-targeting shRNA1 construct Bmi1/pLKO.1
(TRCN0000020155, NM_005180.5-693s1c1) used to
knock down Bmi1 expression was obtained from Open-
Biosystems (Thermo Fisher, Waltham, MA, USA). Control
SC/pLKO.1 (with a scrambled sequence) plasmids were
obtained from Addgene. TGFβ1 shRNA (sc-270322-SH),
Bmi1 siRNA2 (sc-29814) and shRNA2 (sc-29814-v), and
TGFβ2 siRNA2 (sc-39802), shRNA1 (sc-39802-SH), and
shRNA2 (sc-39802-v) lentivirus pools were from Santa
Cruz Biotechnology (Santa Cruz, CA, USA). All shRNA
constructs were packaged into lentivirus and used for
transfection. The following siRNAs were used in this study:
Bmi1 siRNA1 sense: 5-CCAGACCACUACUGAAUA
UAA-3, anti-sense: 5-UUAUAUUCAGUAGUGGUCUG
GUU-3; TGFβ2 siRNA1 sense: 5-CACUCGAUAUGGA
CCAGUUTT-3, anti-sense: 5-AACUGGUCCAUAUC
GAGUG-3. SiRNA oligos were transfected into Huh7 and
Hep3B cells using Lipofectamine 2000 (Invitrogen, Carls-
bad, CA, USA), according to the manufacturers instruc-
tions. The hyperactive sleeping beauty construct (pCMV/
SB), Bmi1 construct pT3-EF1α-Bmi1-V5, N-Ras construct
pCaggs-NRasV12, and Akt construct pT3-myr-Akt-HA
were provided by Dr Xin Chen of the University of Cali-
fornia, San Francisco. The pT3-EF1αvector containing
duplicated inverted repeats for sleeping beauty-mediated
integration and the EF1αpromoter (pT3-EF1α) used for
hydrodynamic injection were previously described [51].
Construction of pT3-EF1α-Bmi1-V5, pCaggs-NRasV12,
and pT3-myrAkt-HA has been described elsewhere
[13,52]. Human TGFβ2 and an IRES sequences were
cloned into pT3-EF1α-Bmi1-V5 to generate pT3-EF1α-
Bmi1-Flag-IRES-TGFβ2-V5. All plasmids were puried
using the Endotoxin-free Maxi Prep Kit (Omega Bio-Tek,
Norcross, GA, USA) before injection into mice.
Cell viability, proliferation, growth, colony-
formation, and cell-cycle analyses
Hep3B or Huh7 cells were transfected with siRNA or
shRNA or treated with drugs for 48 h. ShRNA-transfected
cells were treated with puromycin (1 µg/mL) for 48 h. For
cell viability assays, cells were seeded into 96-well plates at
5000 cells/well and then transfected with siRNA or treated
with drugs. Then, cell viability was determined using an
Bmi1 drives hepatocarcinogenesis by repressing the TGFβ2/SMAD signalling axis
MTT detection kit (Beyotime, Beijing, China). To assay cell
growth, 5000 cells/well were seeded in 12-well plates and
cells were recounted at indicated time points. Cell pro-
liferation was assayed using BrdU labelling as described
previously [53]. For colony-formation assays, 5 × 103viable
transfected cells/well were seeded in 6-well plates and
maintained in complete medium for 1 week. Colonies were
xed with methanol and stained with 0.1% crystal violet in
20% methanol. Colonies were counted with Image J (NIH,
Bethesda, MD, USA). Triplicate wells were prepared for
each transfectant and all experiments were repeated twice.
The cell cycle was analysed by ow cytometry (Cyto-
micsTM FC 500; Beckman Coulter, Brea, CA, USA) after
propidium iodide staining, and the results were analysed
using FlowJo7.6 (FlowJo, Ashland, OR, USA).
Quantitative reverse-transcription (RT-q)PCR,
western blotting, and co-immunoprecipitation
Total RNA was extracted from cells or frozen liver tissues
using TRIzol (Invitrogen) and digested with DNase I. After
reverse transcription, SYBR green-based qPCR was carried
out to detect mRNA levels of genes of interest. The primer
pairs used are listed in Supplementary Table 2.
For western blotting, liver or HCC tissues/cells were
lysed in M-PER Mammalian Protein Extraction Buffer
(Thermo Fisher, Rockford, IL, USA) plus Proteinase Inhi-
bitor Cocktail (Roche, Indianapolis, IN, USA) and Halt
Phosphatase Inhibitor Cocktail (Thermo Fisher). Proteins
were quantied using a BCA protein assay kit (Thermo
Fisher).
For co-immunoprecipitation, cells were harvested and
incubated with IP-lysis buffer (Beyotime). Cell lysates were
incubated with rabbit anti-Smad4 monoclonal antibody
(1:50, Cell Signalling) at 4 °C for 2 h, followed by incu-
bation with 20 μL of protein A/G agarose (Santa Cruz
Biotechnology, CA, USA) at 4 °C overnight. Immunopre-
cipitates were washed four times with lysis buffer and
analysed by western blotting using anti-Smad2 and anti-
Smad3 monoclonal antibodies (1:1000, Cell Signalling).
Antibodies used for western blotting and co-
immunoprecipitation are listed in Supplementary Table 3.
Enzyme-linked immunosorbent assay (ELISA) for
TGFβ2
Hep3B and Huh7 cells were infected with Bmi1 shRNA
lentivirus for 48 h and selected on puromycin for 48 h.
Then, Bmi1 KD and control (infected with scramble
shRNA) cells were seeded into 6-well plates at 4 × 105
cells per well. After 24 h, the culture supernatant was
collected. TGFβ2 concentrations were measured using an
ELISA kit for human TGFβ2(CusabioBiotechCo.,
Houston, TX, USA) according to the manufacturers
instruction. In brief, puried human TGFβ2 standards and
samples were incubated in a high-binding 96-well
microtiter plate pre-coated with TGFβ2 antibody at 37 °C
for 2 h. Subsequently, liquid was removed and diluted
biotin-conjugated antibody was added to each well. After
1 h of incubation at 37 °C and washing, the plate was
incubated with horseradish peroxidase-avidin working
solution at 37 °C for 1 h. After nal washing, the plates
were incubated with 3,3,5,5-tetramethylbenzidine at
37 °C in the dark for 30 min. Finally, the optical density at
450 nm was determined with a Multilabel Reader Spec-
trophotometer (Victor X5, Perkin Elmer). The con-
centration of TGFβ2 in each well was calculated based on
the standard curve.
Histology, immunohistochemistry, and
immunouorescence
Mice were euthanized and the livers were removed and
rinsed in PBS. Samples collected from the livers were either
frozen in dry ice for RNA and protein extraction or xed
overnight in fresh, cold 4% paraformaldehyde. Fixed tissue
samples were embedded in parafn and sectioned to 5 μm
for haematoxylin and eosin (H&E) or immunohistochemical
staining. For immunouorescence staining, cells were xed
with 4% paraformaldehyde, permeabilized with 0.1% Triton
X-100, and pre-treated with 0.5% goat serum. The cells
were incubated with primary antibodies for 1.5 h and sub-
sequently with Alexa Flour 596-conjugated AfniPure goat
anti-rabbit IgG (Proteintech, Rosemont, IL, USA) at a 1:100
dilution for 45 min. Nuclei were counterstained with DAPI.
Antibodies used for immunohistochemistry and immuno-
uorescence are listed in Supplementary Table 3. Staining
intensity was quantied using Image-Pro Plus (Media
Cybernetics, Rockville, MD, USA, version 7.0) to calculate
the integrated optic density.
Primary and xenograft HCC mouse models
Wild-type FVB/N mice and BALB/c nude mice were
obtained from Huaukang Technology Corporation (Beijing,
China) and were fed standard rodent chow. All mice were
housed, fed, and treated in accordance with protocols
approved by the Animal Experiments Ethical Committee at
Huazhong University of Science and Technology. All the
mice used in this study were randomized to different groups
using an online randomization tool Randomizer (https://www.
randomizer.org/), but the animal experiments were not blin-
ded. Hydrodynamic transfection was performed as described
previously [51]. To generate Bmi1/Ras HCC in mice, plas-
mids (pT3-EF1α-Bmi1-V5:pCaggs-NRasV12:pCMV-SB =
25:25:4 μgorpT3-EF1α-Bmi1-FLAG-TGFβ2-V5:pCaggs-
B. Li et al.
NRasV12:pCMV-SB =25:25:4 μg per 20 g of mouse weight)
were diluted in 2 mL of saline, ltered through a 0.22-μm
lter, and injected into the tail vein of 6-week-old female
FVB/N mice (1820 g) in 57 s. The mice were monitored
weekly and sacriced at 20 weeks post injection for growth
evaluation and histological examination. To generate Akt/Ras
HCC in mice, plasmids (pT3-myr-Akt-HA:pCaggs-
NRasV12:pCMV-SB =25:25:4 μg per 20 g of mouse weight)
were injected into the tail vein of 6-week-old female FVB/N
mice (1820 g) in 57s.
For the xenograft tumour model, subcutaneous tumours
were established by inoculating 5 × 106Hep3B cells in the
front armpit of 6-week-old female BALB/c nude mice
(1820 g). Prior to inoculation, the Hep3B cells were
infected with Bmi1 and/or TGFβ2 shRNA lentivirus and
selected with puromycin (1 µg/mL). Mice inoculated with
Hep3B cells transfected with sc/pLKo.1 lentivirus served as
a control. Tumour sizes and mouse weights were measured
simultaneously. Tumour volume (V) was monitored by
measuring the length (L) and width (W) with a vernier
caliper and calculated as V=(L×W2) × 0.5. At the end of
the experiment, mice were sacriced and tumours were
collected and photographed.
Microarray analysis
Liver tissues of Bmi1/mice (whole-body knockout) were
kindly provided by Dr Xin Chen of the University of
California, San Francisco. Total RNA was extracted from
HCC cells and 8-week-old female wild type and Bmi1/
mouse liver tissues. Human (HuGene 1.0 ST; Affymetrix,
Santa Clara, CA, USA) and mouse (MoGene 1.0 ST;
Affymetrix) GeneChip arrays were used for gene expression
proling. Hybridization, washing, staining, scanning, and
data analysis were performed by Phalanx Biotech Group
(Palo Alto, CA, USA). Expression levels were analysed
using Microarray Analysis Suite 5.0 (Affymetrix). The
microarray data were deposited in the NCBI Gene
Expression Omnibus public database (http://www.ncbi.nlm.
nih.gov/geo/, accession number GSE97172).
Patients and tissue samples
Ten fresh HCC tissue samples were collected at Union
Hospital afliated to the Tongji Medical College of Huaz-
hong University of Science and Technology (Wuhan,
China) in 2016. The collection of HCC tissues was
approved by Medical Ethics Committees of Huazhong
University of Science and Technology and written informed
consent was obtained from all patients before surgery. The
HCC tissue microarray (Cat Number: LivH150CS03; Lot
Number.: XT15-037) was from Shanghai Outdo Biotech
Company (Shanghai, China).
Luciferase reporter and ChIP assays
The regions 2000 to +100 and 508 to +63 of the human
TGFβ2 promoter were PCR-amplied and cloned into the
pGL3 luciferase vector. To analyse TGFβ2 transcriptional
activity, Huh7 cells were transfected with the pGL3 plasmid
and an internal control reporter plasmid, pRL-TK (Promega,
Madison, WI, USA), along with Bmi1-siRNA or pT3-Bmi1
plasmid. Forty-eight hours after transfection, luciferase
activity was measured with the Dual-Glo luciferase assay
system (Promega) according to the manufacturers
instructions.
ChIP assays were essentially performed as previously
described [54,55]. ChIP DNA was analysed by qPCR with
SYBR Green (Bio-Rad Laboratories, Berkeley, California,
USA) in an ABI-7500 instrument (Thermo Fisher) using the
following primers: primer 1F: 5-TTGGGAGGCTGTGA
CTGA-3, primer 1R: 5-GCTGTGGGTAAGGGAGGA-3;
primer 2F: 5-TAATACAGGAGGGAAGCC-3, primer
2R: 5-TGCCAGCAGATAACATCA-3. Antibodies used
were as follows: rabbit anti-Ring1B (Abcam, Cambridge,
MA, USA) and normal rabbit IgG (Abcam).
Statistical analysis
Data are reported as the mean ± standard deviation (SD).
Means of two groups were compared using Studentsttest
(SPSS Software, Chicago, IL, USA). Means of multiple
groups were compared by one-way ANOVA and Dun-
netts posttests. Variances between multiple groups were
statistically compared. Values of p<0.05 and p<0.01
were considered signicant and highly signicant,
respectively. Sample sizes of animal experiments were
calculated using the resource equation:E=total number
of animalstotal number of groups, in which Eshould lie
between 10 and 20 [56].
Acknowledgements This study was supported by the National Science
Foundation of China (81572723 and 81872253) and the Innovation
Foundation of Higher Education of China (2016JCTD109).
Compliance with ethical standards
Conict of interest The authors declare that they have no conicts of
interest.
Publishers note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional afliations.
References
1. Jacobs JJ, Kieboom K, Marino S, DePinho RA, van Lohuizen M.
The oncogene and Polycomb-group gene bmi-1 regulates cell
proliferation and senescence through the ink4a locus. Nature.
1999a;397:1648.
Bmi1 drives hepatocarcinogenesis by repressing the TGFβ2/SMAD signalling axis
2. Molofsky AV, Pardal R, Iwashita T, Park IK, Clarke MF, Mor-
rison SJ. Bmi-1 dependence distinguishes neural stem cell self-
renewal from progenitor proliferation. Nature. 2003;425:9627.
3. Park IK, Qian D, Kiel M, Becker MW, Pihalja M, Weissman IL,
et al. Bmi-1 is required for maintenance of adult self-renewing
haematopoietic stem cells. Nature. 2003;423:3025.
4. Park IK, Morrison SJ, Clarke MF. Bmi1, stem cells, and senes-
cence regulation. J Clin Investig. 2004;113:1759.
5. Pietersen AM, Horlings HM, Hauptmann M, Langerod A,
Ajouaou A, Cornelissen-Steijger P, et al. EZH2 and BMI1
inversely correlate with prognosis and TP53 mutation in breast
cancer. Breast Cancer Res. 2008;10:R109.
6. Dhawan S, Tschen SI, Bhushan A. Bmi-1 regulates the Ink4a/Arf
locus to control pancreatic beta-cell proliferation. Genes Dev.
2009;23:90611.
7. Dovey JS, Zacharek SJ, Kim CF, Lees JA. Bmi1 is critical for
lung tumorigenesis and bronchioalveolar stem cell expansion.
Proc Natl Acad Sci USA. 2008;105:1185762.
8. Becker M, Korn C, Sienerth AR, Voswinckel R, Luetkenhaus K,
Ceteci F, et al. Polycomb group protein Bmi1 is required for
growth of RAFdriven non-small-cell lung cancer. PLoS ONE.
2009;4:e4230.
9. Bruggeman SW, Hulsman D, Tanger E, Buckle T, Blom M,
Zevenhoven J, et al. Bmi1 controls tumor development in an
Ink4a/Arf-independent manner in a mouse model for glioma.
Cancer Cell. 2007;12:32841.
10. Jacobs JJ, Scheijen B, Voncken JW, Kieboom K, Berns A, van
Lohuizen M. Bmi-1 collaborates with c-Myc in tumorigenesis by
inhibiting c-Myc-induced apoptosis via INK4a/ARF. Genes Dev.
1999b;13:267890.
11. Kalra N, Kumar V. c-Fos is a mediator of the c-myc-induced
apoptotic signaling in serum-deprived hepatoma cells via the p38
mitogen-activated protein kinase pathway. J Biol Chem.
2004;279:253139.
12. Levy L, Renard CA, Wei Y, Buendia MA. Genetic alterations and
oncogenic pathways in hepatocellular carcinoma. Ann N Y Acad
Sci. 2002;963:2136.
13. Xu CR, Lee S, Ho C, Bommi P, Huang SA, Cheung ST, et al.
Bmi1 functions as an oncogene independent of Ink4A/Arf
repression in hepatic carcinogenesis. Mol Cancer Res.
2009;7:193745.
14. Chiba T, Miyagi S, Saraya A, Aoki R, Seki A, Morita Y, et al. The
polycomb gene product BMI1 contributes to the maintenance of
tumor-initiating side population cells in hepatocellular carcinoma.
Cancer Res. 2008;68:77429.
15. Qi S, Li B, Yang T, Liu Y, Cao S, He X, et al. Validation of Bmi1
as a therapeutic target of hepatocellular carcinoma in mice. Int J
Mol Sci. 2014;15:2000421.
16. Yang T, Li B, Qi S, Liu Y, Gai Y, Ye P, et al. Co-delivery of
doxorubicin and Bmi1 siRNA by folate receptor targeted lipo-
somes exhibits enhanced anti-tumor effects in vitro and in vivo.
Theranostics. 2014;4:1096111.
17. Chiba T, Seki A, Aoki R, Ichikawa H, Negishi M, Miyagi S, et al.
Bmi1 promotes hepatic stem cell expansion and tumorigenicity in
both Ink4a/Arf-dependent and -independent manners in mice.
Hepatology. 2010;52:111123.
18. Goel HL, Chang C, Pursell B, Leav I, Lyle S, Xi HS, et al. VEGF/
neuropilin-2 regulation of Bmi-1 and consequent repression of
IGF-IR dene a novel mechanism of aggressive prostate cancer.
Cancer Discov. 2012;2:90621.
19. Molofsky AV, He S, Bydon M, Morrison SJ, Pardal R. Bmi-1
promotes neural stem cell self-renewal and neural development
but not mouse growth and survival by repressing the p16Ink4a
and p19Arf senescence pathways. Genes Dev. 2005;19:14327.
20. Sangiorgi E, Capecchi MR. Bmi1 is expressed in vivo in intestinal
stem cells. Nat Genet. 2008;40:91520.
21. Fan C, He L, Kapoor A, Gillis A, Rybak AP, Cutz JC, et al. Bmi1
promotes prostate tumorigenesis via inhibitingp16(INK4A)
and p14(ARF) expression. Biochim Biophys Acta. 2008;
1782:6428.
22. Vonlanthen S, Heighway J, Altermatt HJ, Gugger M, Kappeler A,
Borner MM, et al. The bmi-1 oncoprotein is differentially
expressed in non-small cell lung cancer and correlates with
INK4A-ARF locus expression. Br J Cancer. 2001;84:13726.
23. Douglas D, Hsu JH, Hung L, Cooper A, Abdueva D, van Door-
ninck J, et al. BMI-1 promotes ewing sarcoma tumorigenicity
independent of CDKN2A repression. Cancer Res. 2008;
68:650715.
24. Heldin CH, Miyazono K, ten Dijke P. TGF-beta signalling from
cell membrane to nucleus through SMAD proteins. Nature.
1997;390:46571.
25. Massague J. TGFbeta in Cancer. Cell. 2008;134:21530.
26. Coulouarn C, Factor VM, Thorgeirsson SS. Transforming growth
factor-beta gene expression signature in mouse hepatocytes pre-
dicts clinical outcome in human cancer. Hepatology. 2008;
47:205967.
27. Fan L, Xu C, Wang C, Tao J, Ho C, Jiang L, et al. Bmi1 is
required for hepatic progenitor cell expansion and liver tumor
development. PLoS ONE. 2012;7:e46472.
28. Oh S, Kim E, Kang D, Kim M, Kim JH, Song JJ. Transforming
growth factor-beta gene silencing using adenovirus expressing
TGF-beta1 or TGF-beta2 shRNA. Cancer Gene Ther.
2013;20:94100.
29. Kim JH, Yoon SY, Kim CN, Joo JH, Moon SK, Choe IS, et al.
The Bmi-1 oncoprotein is overexpressed in human colorectal
cancer and correlates with the reduced p16INK4a/p14ARF pro-
teins. Cancer Lett. 2004;203:21724.
30. Bentley ML, Corn JE, Dong KC, Phung Q, Cheung TK, Cochran
AG. Recognition of UbcH5c and the nucleosome by the Bmi1/
Ring1b ubiquitin ligase complex. EMBO J. 2011;30:328597.
31. McGinty RK, Henrici RC, Tan S. Crystal structure of the PRC1
ubiquitylation module bound to the nucleosome. Nature.
2014;514:5916.
32. Blackledge NP, Farcas AM, Kondo T, King HW, McGouran JF,
Hanssen LL, et al. Variant PRC1 complex-dependent H2A ubi-
quitylation drives PRC2 recruitment and polycomb domain for-
mation. Cell. 2014;157:144559.
33. Noma T, Glick AB, Geiser AG, OReilly MA, Miller J, Roberts
AB, et al. Molecular cloning and structure of the human trans-
forming growth factor-beta 2 gene promoter. Growth Factors.
1991;4:24755.
34. Datta S, Hoenerhoff MJ, Bommi P, Sainger R, Guo WJ, Dimri M,
et al. Bmi-1 cooperates with H-Ras to transform human mammary
epithelial cells via dysregulation of multiple growth-regulatory
pathways. Cancer Res. 2007;67:1028695.
35. Hoenerhoff MJ, Chu I, Barkan D, Liu ZY, Datta S, Dimri GP,
et al. BMI1 cooperates with H-RAS to induce an aggressive breast
cancer phenotype with brain metastases. Oncogene.
2009;28:302232.
36. Jagani Z, Wiederschain D, Loo A, He D, Mosher R, Fordjour P,
et al. The Polycomb group protein Bmi-1 is essential for the
growth of multiple myeloma cells. Cancer Res. 2010;70:552838.
37. Silva J, Garcia JM, Pena C, Garcia V, Dominguez G, Suarez D,
et al. Implication of polycomb members Bmi-1, Mel-18, and Hpc-
2 in the regulation of p16INK4a, p14ARF, h-TERT, and c-Myc
expression in primary breast carcinomas. Clin Cancer Res.
2006;12:692936.
38. Kreso A, van Galen P, Pedley NM, Lima-Fernandes E, Frelin C,
Davis T, et al. Self-renewal as a therapeutic target in human
colorectal cancer. Nat Med. 2014;20:2936.
39. Datto MB, Li Y, Panus JF, Howe DJ, Xiong Y, Wang XF.
Transforming growth factor beta induces the cyclin-dependent
B. Li et al.
kinase inhibitor p21 through a p53-independent mechanism. Proc
Natl Acad Sci USA. 1995;92:55459.
40. Reynisdottir I, Polyak K, Iavarone A, Massague J. Kip/Cip and
Ink4 Cdk inhibitors cooperate to induce cell cycle arrest in
response to TGF-beta. Genes Dev. 1995;9:183145.
41. Li CY, Suardet L, Little JB. Potential role of WAF1/Cip1/p21 as a
mediator of TGF-beta cytoinhibitory effect. J Biol Chem.
1995;270:49714.
42. Sandhu C, Garbe J, Bhattacharya N, Daksis J, Pan CH, Yaswen P,
et al. Transforming growth factor beta stabilizes p15INK4B pro-
tein, increases p15INK4B-cdk4 complexes, and inhibits cyclin
D1-cdk4 association in human mammary epithelial cells. Mol Cell
Biol. 1997;17:245867.
43. Gargiulo G, Cesaroni M, Serresi M, de Vries N, Hulsman D,
Bruggeman SW, et al. In vivo RNAi screen for BMI1 targets
identies TGF-beta/BMP-ER stress pathways as key regulators of
neural- and malignant glioma-stem cell homeostasis. Cancer Cell.
2013;23:66076.
44. Kim RH, Lieberman MB, Lee R, Shin KH, Mehrazarin S, Oh JE,
et al. Bmi-1 extends the life span of normal human oral kerati-
nocytes by inhibiting the TGF-beta signaling. Exp Cell Res.
2010;316:26008.
45. Batlle E, Massague J. Transforming growth factor-beta signaling
in immunity and cancer. Immunity. 2019;50:92440.
46. Mitchell EJ, Lee K, OConnor-McCourt MD. Characterization of
transforming growth factor-beta (TGF-beta) receptors on BeWo
choriocarcinoma cells including the identication of a novel 38-
kDa TGF-beta binding glycoprotein. Mol Biol Cell.
1992;3:1295307.
47. del Re E, Babitt JL, Pirani A, Schneyer AL, Lin HY. In the
absence of type III receptor, the transforming growth factor
(TGF)-beta type II-B receptor requires the type I receptor to bind
TGF-beta2. J Biol Chem. 2004;279:2276572.
48. Rotzer D, Roth M, Lutz M, Lindemann D, Sebald W, Knaus P.
Type III TGF-beta receptor-independent signalling of TGF-beta2
via TbetaRII-B, an alternatively spliced TGF-beta type II receptor.
EMBO J. 2001;20:48090.
49. Rodriguez C, Chen F, Weinberg RA, Lodish HF. Cooperative
binding of transforming growth factor (TGF)-beta 2 to the types I
and II TGF-beta receptors. J Biol Chem. 1995;270:1591922.
50. Dropmann A, Dediulia T, Breitkopf-Heinlein K, Korhonen H,
Janicot M, Weber SN, et al. TGF-beta1 and TGF-beta2 abundance
in liver diseases of mice and men. Oncotarget. 2016;7:19499518.
51. Tward AD, Jones KD, Yant S, Cheung ST, Fan ST, Chen X, et al.
Distinct pathways of genomic progression to benign and malig-
nant tumors of the liver. Proc Natl Acad Sci USA.
2007;104:147716.
52. HoC,WangC,MattuS,DestefanisG,LaduS,DeloguS,etal.AKT
(v-akt murine thymoma viral oncogene homolog 1) and N-Ras
(neuroblastoma ras viral oncogene homolog) coactivation in the
mouse liver promotes rapid carcinogenesis by way of mTOR
(mammalian target of rapamycin complex 1), FOXM1 (forkhead box
M1)/SKP2, and c-Myc pathways. Hepatology. 2012;55:83345.
53. Wojtowicz JM, Kee N. BrdU assay for neurogenesis in rodents.
Nat Protoc. 2006;1:1399405.
54. Jiang Y, Yan B, Lai W, Shi Y, Xiao D, Jia J, et al. Repression of
Hox genes by LMP1 in nasopharyngeal carcinoma and modula-
tion of glycolytic pathway genes by HoxC8. Oncogene.
2015;34:607991.
55. Shi Y, Tao Y, Jiang Y, Xu Y, Yan B, Chen X, et al. Nuclear
epidermal growth factor receptor interacts with transcriptional
intermediary factor 2 to activate cyclin D1 gene expression trig-
gered by the oncoprotein latent membrane protein 1. Carcino-
genesis. 2012;33:146878.
56. Charan J, Kantharia ND. How to calculate sample size in animal
studies? J Pharmacol Pharmacother. 2013;4:3036.
Bmi1 drives hepatocarcinogenesis by repressing the TGFβ2/SMAD signalling axis
... All cells were cultured in a 5% CO 2 humidification incubator at 37 ℃. PTC-209, a specific BMI1 inhibitor [8,31], was Conclusions: Our results reveal a novel molecular mechanism of OS development promoted by BMI1 and provides a new potential target for OS treatment. ...
... Polycomb group (PcG) proteins consist of polycomb repressive complex 1 (PRC1) and polycomb repressive complex 2 (PRC2) [7]. The proto-oncogene B lymphoma Mo-MLV insertion region 1 (BMI1), a key component of PRC1, is a transcriptional repressor that has been shown to be involved in tumorigenesis, cell cycle regulation and stem cell maintenance [8][9][10][11][12]. BMI1 acts as an oncogene by forming complexes with other members of the PcG to inhibit expression of tumor suppressor genes [13]. ...
... BMI1 acts as an oncogene by forming complexes with other members of the PcG to inhibit expression of tumor suppressor genes [13]. Previous reports indicated that BMI1 is associated with the occurrence, progression, and prognosis of multiple tumor types, including bladder cancer [12,14], non-small cell lung cancer [15], colon carcinoma [16], colitis-associated cancer [17], breast cancer [18], glioblastoma [19], and hepatocellular carcinoma [8]. BMI1 has also been studied in OS tissues and cell lines. ...
Article
Full-text available
Background Osteosarcoma (OS) is the most common malignant tumor of bone, and the clinical efficacy of current treatments and associated survival rates need to be further improved by employing novel therapeutic strategies. Although various studies have shown that BMI1 protein is universally upregulated in OS cells and tissues, its specific role and underlying mechanism have not yet been fully explored. Methods Expression of BMI1 protein in OS cells was detected by western blot. The effect of BMI1 on proliferation and migration of OS cells (143B and U-2OS cell lines) was investigated in vitro using CCK-8, colony formation and transwell assays, and in vivo using subcutaneous tumorigenesis and lung metastasis assays in xenograft nude mice. Expression of epithelial–mesenchymal transition (EMT)-associated proteins was detected by immunofluorescence imaging. Bioinformatic analysis was performed using ENCODE databases to predict downstream targets of BMI1. SIK1 mRNA expression in osteosarcoma cells was detected by quantitative real-time reverse transcription PCR (qPCR). Chromatin immunoprecipitation-qPCR (ChIP-qPCR) was used to investigate expression of BMI1-associated, RING1B-associated, H2AK119ub-associated and H3K4me3-associated DNA at the putative binding region of BMI1 on the SIK1 promoter in OS cells. Results Using both in vitro and in vivo experimental approaches, we found that BMI1 promotes OS cell proliferation and metastasis. The tumor suppressor SIK1 was identified as the direct target gene of BMI1 in OS cells. In vitro experiments demonstrated that SIK1 could inhibit proliferation and migration of OS cells. Inhibition of SIK1 largely rescued the altered phenotypes of BMI1-deficient OS cells. Mechanistically, we demonstrated that BMI1 directly binds to the promoter region of SIK1 in a complex with RING1B to promote monoubiquitination of histone H2A at lysine 119 (H2AK119ub) and inhibit H3K4 trimethylation (H3K4me3), resulting in inhibition of SIK1 transcription. We therefore suggest that BMI1 promotes OS cell proliferation and metastasis by inhibiting SIK1. Conclusions Our results reveal a novel molecular mechanism of OS development promoted by BMI1 and provides a new potential target for OS treatment.
... TGF-β/SMAD signalling is critical in HCC development (Li et al., 2020). In principle, the TGF-β superfamily comprises nearly 30 kinds of structurally and functionally related proteins, such as BMPs, GDFs, GDNFs, and activins (Massague & Wotton, 2000). ...
... TGFβ exerts its biological activity through either SMAD-dependent or SMAD-independent pathways (Zhang, 2017). TGF-β binds to and activates its receptors and then activates downstream SMAD proteins through phosphorylation and nuclear translocation (Li et al., 2020). To understand the relationship between SQLE and TGF-β/SMAD signalling in HCC, we examined the receptors for TGF-β (TGF-β1, TGF-β2, and TGF-β3) mRNA levels in Huh7 and SMMC7721 cells after transfection with siRNAs against SQLE. ...
Article
Full-text available
Background and Purpose Squalene epoxidase (SQLE) is a key enzyme involved in cholesterol biosynthesis, but growing evidence also reveals that SQLE is abnormally expressed in some types of malignant tumours, even though the underlying mechanism remains poorly understood. Experimental Approach Bioinformatics analysis and RNA sequencing were applied to detect differentially expressed genes in clinical hepatocellular carcinoma (HCC). MTT, colony formation, AnnexinV‐FITC/PI, EdU, wound healing, transwell, western blot, qRT‐PCR, IHC, F‐actin, RNA‐sequencing, dual‐luciferase reporters, and H&E staining were used to investigate the pharmacological effects and possible mechanisms of SQLE. Key Results SQLE expression was specifically elevated in HCC, correlating with poor clinical outcomes. SQLE significantly promoted HCC growth, epithelial–mesenchymal transition, and metastasis both in vitro and in vivo. RNA sequencing and functional experiments revealed that the protumourigenic effect of SQLE on HCC was closely related to the activation of TGF‐β/SMAD signalling, but the stimulatory effect of SQLE on TGF‐β/SMAD signalling and HCC development is critically dependent on STRAP. SQLE expression is well correlated with STRAP in HCC, and further, to amplify TGF‐β/SMAD signalling, SQLE even transcriptionally increased STRAP gene expression mediated by AP‐2α. Finally, as a chemical inhibitor of SQLE, NB‐598 markedly inhibited HCC cell growth and tumour development. Conclusions and Implications Taken together, SQLE serves as a novel oncogene in HCC development by activating TGF‐β/SMAD signalling. Targeting SQLE could be useful in drug development and therapy for HCC.
... Furthermore, overexpression of BMI-1 has also been found in patients with myelodysplastic syndromes, chronic myelogenous leukemia, acute myeloid leukemia, and lymphoma [1,35,37,38]. BMI1 is overexpressed in one-third of patients with LIHC and is considered a key target for LIHC treatment [29]. Small molecule inhibitors targeting BMI1 have shown great potential in the treatment of other tumors. ...
Article
Full-text available
Background Numerous studies have shown that Schistosoma japonicum infection correlates with an increased risk of liver hepatocellular carcinoma (LIHC). However, data regarding the role of this infection in LIHC oncogenesis are scarce. This study aimed to investigate the potential mechanisms of hepatocarcinogenesis associated with Schistosoma japonicum infection. Methods By examining chronic liver disease as a mediator, we identified the genes contributing to Schistosoma japonicum infection and LIHC. We selected 15 key differentially expressed genes (DEGs) using weighted gene co-expression network analysis (WGCNA) and random survival forest models. Consensus clustering revealed two subgroups with distinct prognoses. Least Absolute Shrinkage and Selection Operator (LASSO) and Cox regression identified six prognostic DEGs, forming an Schistosoma japonicum infection-associated signature for strong prognosis prediction. This signature, which is an independent LIHC risk factor, was significantly correlated with clinical variables. Four DEGs, including BMI1, were selected based on their protein expression levels in cancerous and normal tissues. We confirmed BMI1's role in LIHC using Schistosoma japonicum-infected mouse models and molecular experiments. Results We identified a series of DEGs that mediate schistosomiasis, the parasitic disease caused by Schistosoma japonicum infection, and hepatocarcinogenesis, and constructed a suitable prognostic model. We analyzed the mechanisms by which these DEGs regulate disease and present the differences in prognosis between the different genotypes. Finally, we verified our findings using molecular biology experiments. Conclusion Bioinformatics and molecular biology analyses confirmed a relationship between schistosomiasis and liver hepatocellular cancer. Furthermore, we validated the role of a potential oncoprotein factor that may be associated with infection and carcinogenesis. These findings enhance our understanding of Schistosoma japonicum infection's role in LIHC carcinogenesis.
... In the context of HCC, BMI1 exhibits upregulated expression. It influences HCC development through diverse mechanisms, including its impact on the INK4a/ARF locus, involvement in the NF-κB signaling pathway, and modulation of the PTEN/PI3K/AKT signaling pathway (27)(28)(29)(30)(31). Moreover, BMI1 expression has been found to be closely associated with both HCC prognosis and recurrence (32). ...
Article
Full-text available
Background Hepatocellular carcinoma (HCC) is a malignancy with a bleak prognosis. Although emerging research increasingly supports the involvement of chromatin regulators (CRs) in cancer development, CRs in HCC patients have not received proportionate attention. This study aimed to investigate the role and prognostic significance of CRs in HCC patients, providing new insights for clinical diagnosis and treatment strategies. Methods We analyzed 424 samples in The Cancer Genome Atlas-Liver hepatocellular carcinoma (TCGA-LIHC) data to identify key CR genes associated with HCC prognosis by differential expression and univariate Cox regression analyses. LASSO-multivariate Cox regression method was used for construction of a prognostic signature and development of a CR-related prognosis model. The prognosis capacity of the model was evaluated via Kaplan-Meier method. Relationship between the model and tumor microenvironment (TME) was evaluated. Additionally, clinical variables and the model were incorporated to create a nomogram. The role of the prognostic gene MRG-binding protein (MRGBP) in HCC was elucidated by immunohistochemistry and semiquantitative analysis. Results A risk score model, comprising B-lymphoma Mo-MLV insertion region 1 (BMI1), chromobox 2 (CBX2), and MRGBP, was constructed. The area under the curve (AUC) of the CR-based signature is 0.698 (P<0.05), exhibiting robust predictive power. Functional and pathway analyses illuminated the biological relevance of these genes. Immune microenvironment analysis suggested potential implications for immunotherapy. Drug sensitivity analysis identified agents for targeted treatment. Clinical samples show that MRGBP is highly expressed in HCC tissues. Conclusions This CR-based signature shows promise as a valuable prognostic tool for HCC patients. It demonstrates predictive capabilities, independence from other clinical factors, and potential clinical applicability. In addition, we need more experiments to validate our findings. These findings offer insights into HCC prognosis and treatment, with implications for personalized medicine and improved patient outcomes.
... It is well established that aberrant expression of core PcG subunits contributes to tumor initiation and progression. In HCC, overexpression of EZH2 represses miR-622 through H3K27me3 deposition and results in CXCR4 upregulation and unfavorable prognosis, while BMI1 enhances TGFβ2/SMAD pathway and facilitates tumor cell proliferation and cell cycle progression [29,30]. In recent years, accumulating evidence suggested that other non-core accessory proteins alternatively constituting the PcG complex can also facilitate the pro-tumor process. ...
Article
Full-text available
Background Hepatocellular carcinoma (HCC) is an extensive heterogeneous disease where epigenetic factors contribute to its pathogenesis. Polycomb group (PcG) proteins are a group of subunits constituting various macro-molecular machines to regulate the epigenetic landscape, which contributes to cancer phenotype and has the potential to develop a molecular classification of HCC. Results Here, based on multi-omics data analysis of DNA methylation, mRNA expression, and copy number of PcG-related genes, we established an epigenetic classification system of HCC, which divides the HCC patients into two subgroups with significantly different outcomes. Comparing these two epigenetic subgroups, we identified different metabolic features, which were related to epigenetic regulation of polycomb-repressive complex 1/2 (PRC1/2). Furthermore, we experimentally proved that inhibition of PcG complexes enhanced the lipid metabolism and reduced the capacity of HCC cells against glucose shortage. In addition, we validated the low chemotherapy sensitivity of HCC in Group A and found inhibition of PRC1/2 promoted HCC cells’ sensitivity to oxaliplatin in vitro and in vivo. Finally, we found that aberrant upregulation of CBX2 in Group A and upregulation of CBX2 were associated with poor prognosis in HCC patients. Furthermore, we found that manipulation of CBX2 affected the levels of H3K27me3 and H2AK119ub. Contributions Our study provided a novel molecular classification system based on PcG-related genes data and experimentally validated the biological features of HCC in two subgroups. Our founding supported the polycomb complex targeting strategy to inhibit HCC progression where CBX2 could be a feasible therapeutic target.
... It is well-established that aberrant expression of core PcG subunits contributes to tumor initiation and progression. In HCC, overexpression of EZH2 represses miR-622 through H3K27me3 deposition and results in CXCR4 upregulation and unfavorable prognosis, while BMI1 enhances TGFβ2/SMAD pathway and facilitates tumor cell proliferation and cell cycle progression [25,26]. In recent years, accumulating evidence suggested that other non-core accessory proteins alternatively constituting the PcG complex can also facilitate the pro-tumor process. ...
Preprint
Full-text available
Background: Hepatocellular carcinoma (HCC) is an extensive heterogeneous disease where epigenetic factors contribute to its pathogenesis. Polycomb group (PcG) proteins are a group of subunits constituting various macro molecular machines to regulate the epigenetic landscape, which contribute to cancer phenotype and have potential to develop molecular classification of HCC. Results: Here, based on multi-omics data analysis of DNA methylation, mRNA expression and copy number of PcG-related genes, we established an epigenetic classification system of HCC, which divides the HCC patients into two subgroups with a significantly different outcome. Comparing these two epigenetic subgroups, we identified different metabolic features, which were related to epigenetic regulation of Polycomb Repressive Complex 1/2 (PRC1/2). Furthermore, we experimentally proved that inhibition of PcG complexes enhanced lipid metabolism and reduced the capacity of HCC cells against glucose shortage. In addition, we validated the low chemotherapy sensitivity of HCC in Group A, and found inhibition of PRC1/2 promoted HCC cells sensitivity to oxaliplatin in vitro and in vivo. Finally, we found that aberrant upregulation CBX2 in Group A and upregulation of CBX2 was associated with poor prognosis in HCC patients. Furthermore, we found manipulation of CBX2 affected the levels of H3K27me3 and H2AK119ub. Conclusions: Our study provided a novel molecular classification system based on PcG-related genes data, and experimentally validated the biological features of HCC in two subgroups. Our founding supported the polycomb complex targeting strategy to inhibit HCC progression where CBX2 could be a feasible therapeutic target.
Article
Full-text available
Purpose of Review Adult stem cells play a pivotal role in the regeneration and revival of tissues that have been affected by aging or injury. Emerging studies have provided strong evidence to support the idea that cells with properties resembling stem cells play a crucial role in the development and progression of various types of human cancers (Wang and Dick, Trends Cell Biol 15:494–501, 2005; Reya et al., Nature 414:105–11, 2001). This review will specifically focus on the interplay between cancer stem cells and various forms of cancer. Recent Findings The notion of cancer stem cells (CSCs) was initially explored in depth within the context of acute myelogenous leukemia (AML) and has since garnered significant interest in the field of cancer research (Nguyen et al., Nat Rev Cancer 12:133–43, 2012). Cancer stem cells share many similarities with stem cells in various aspects. CSCs are a subset of cells found within tumors that possess the ability to self-renew, differentiate, and induce tumor formation when introduced into an animal model. A specific group of cell surface markers, such as CD133, CD44, CD166, EpCAM, CD34, CD90, and CD24, are frequently employed in the identification and enrichment of CSCs (Zhou et al., Biochem Pharmacol 209:115441, 2023; Levin et al., Gastroenterology 139:2072–2082.e5, 2010; Shaikh et al., Cancer Biomarkers 16:301–7, 2016; Gao et al., Stem Cell Rev Rep 2023). Summary There is substantial evidence indicating that CSCs exhibit resistance to various standard treatments as well as playing an essential part in tumor recurrence along with the initiation of cancer metastasis. CSCs are also engaged in intercellular communication with various constituents within the tumor microenvironment (TME), thereby promoting the advancement of the tumor. The recent progress in the field of CSCs has generated hope regarding the potential for enhanced efficacy in future cancer treatments.
Article
Protein phosphatase 2A (PP2A) is an essential tumor suppressor, with its activity often hindered in cancer cells by endogenous PP2A inhibitory proteins like SE translocation (SET). SET/PP2A axis plays a pivotal role in the colony-formation ability of cancer cells and the stabilization of c-Myc and E2F1 proteins implicated in this process. However, in osteosarcoma cell line HOS, SET knock-down (KD) suppresses the colony-formation ability without affecting c-Myc and E2F1. This study aimed to unravel the molecular mechanism through which SET enhances the colony-formation ability of HOS cells and determine if it is generalized to other cancer cells. Transcriptome analysis unveiled that SET KD suppressed mTORC1 signaling. SET KD inhibited Akt phosphorylation, an upstream kinase for mTORC1. PP2A inhibitor blocked SET KD-mediated decrease in phosphorylation of Akt and a mTORC1 substrate p70S6K. A constitutively active Akt restored decreased colony-formation ability by SET KD, indicating the SET/PP2A/Akt/mTORC1 axis. Additionally, enrichment analysis highlighted that Bmi-1, a polycomb group protein, is affected by SET KD. SET KD decreased Bmi-1 protein by Akt inhibition but not by mTORC1 inhibition, and exogenous Bmi-1 expression rescued the reduced colony formation by SET KD. Four out of eight cancer cell lines exhibited decreased Bmi-1 by SET KD. Further analysis of these cell lines revealed that Myc activity plays a role in SET KD-mediated Bmi-1 degradation. These findings provide new insights into the molecular mechanism of SET-regulated colony-formation ability, which involved Akt-mediated activation of mTORC1/p70S6K and Bmi-1 signaling.
Article
Full-text available
Breast cancer is among the most common malignant cancers in women. B-cell-specific Moloney murine leukemia virus integration site 1 (BMI-1) is a transcriptional repressor that has been shown to be involved in tumorigenesis, the cell cycle, and stem cell maintenance. In our study, increased expression of BMI-1 was found in both human triple negative breast cancer and luminal A-type breast cancer tissues compared with adjacent tissues. We also found that knockdown of BMI-1 significantly suppressed cell proliferation and migration in vitro and vivo. Further mechanistic research demonstrated that BMI-1 directly bound to the promoter region of CDKN2D/BRCA1 and inhibited its transcription in MCF-7/MDA-MB-231. More importantly, we discovered that knockdown of CDKN2D/BRCA1 could promote cell proliferation and migration after repression by PTC-209. Our results reveal that BMI-1 transcriptionally suppressed BRCA1 in TNBC cell lines, whereas in luminal A cell lines, CDKN2D was the target gene. This provides a reference for the precise treatment of different types of breast cancer in clinical practice.
Article
Full-text available
Background and aims: Patients with persistent positive hepatitis B surface antigen (HBsAg), even with a low HBV-DNA load, have a higher risk of hepatocellular carcinoma (HCC) than those without HBV infection. Given that tumor stemness has a critical role in the occurrence and maintenance of neoplasms, this study aimed to explore whether HBsAg affects biological function and stemness of HCC by regulating microRNA, and to explore underlying mechanisms. Methods: We screened out miR-203a, the most significant down-regulated microRNA in the microarray analysis of HBsAg-positive samples and focused on that miRNA in the ensuing study. In vitro and in vivo functional experiments were performed to assess its regulatory function. The effect of miR-203a on stemness and the possible correlation with BMI1 were analyzed in this study. Results: MiR-203a was significantly down-regulated in HBsAg-positive HCC with the sharpest decrease shown in microarray analysis. The negative correlation between miR-203a and HBsAg expression was confirmed by quantitative real-time PCR after stimulation or overexpression/knockdown of HBsAg in cells. We demonstrated the function of miR-203a in inhibiting HCC cell proliferation, migration, clonogenic capacity, and tumor development in vivo. Furthermore, the overexpression of miR-203a remarkably increases the sensitivity of tumor cells to 5-FU treatment and decreases the proportion of HCC cells with stem markers. In concordance with our study, the survival analysis of both The Cancer Genome Atlas database and samples in our center indicated a worse prognosis in patients with low level of miR-203a. We also found that BMI1, a gene maintains the self-renewal capacity of stem cells, showed a significant negative correlation with miR-203a in HCC specimen (p<0.001). Similarly, opposite BMI1 changes after overexpression/knockdown of miR-203a were also confirmed in vitro. Dual luciferase reporting assay suggested that miR-203a may regulate BMI1 expression by direct binding. Conclusions: HBsAg may promote the development of HCC and tumor stemness by inhibiting miR-203a, resulting in poor prognosis. miR-203a may serve as a crucial treatment target in HBsAg-positive HCC. More explicit mechanistic studies and animal experiments need to be conducted as a next step.
Article
Full-text available
TGF-β1 is a major player in chronic liver diseases promoting fibrogenesis and tumorigenesis through various mechanisms. The expression and function of TGF-β2 have not been investigated thoroughly in liver disease to date. In this paper, we provide evidence that TGF-β2 expression correlates with fibrogenesis and liver cancer development. Using quantitative realtime PCR and ELISA, we show that TGF-β2 mRNA expression and secretion increased in murine HSCs and hepatocytes over time in culture and were found in the human-derived HSC cell line LX-2. TGF-β2 stimulation of the LX-2 cells led to upregulation of the TGF-β receptors 1, 2, and 3, whereas TGF-β1 treatment did not alter or decrease their expression. In liver regeneration and fibrosis upon CCl4 challenge, the transient increase of TGF-β2 expression was accompanied by TGF-β1 and collagen expression. In bile duct ligation-induced fibrosis, TGF-β2 upregulation correlated with fibrotic markers and was more prominent than TGF-β1 expression. Accordingly, MDR2-KO mice showed significant TGF-β2 upregulation within 3 to 15 months but minor TGF-β1 expression changes. In 5 of 8 hepatocellular carcinoma (HCC)/hepatoblastoma cell lines, relatively high TGF-β2 expression and secretion were observed, with some cell lines even secreting more TGF-β2 than TGF-β1. TGF-β2 was also upregulated in tumors of TGFα/cMyc and DEN-treated mice. The analysis of publically available microarray data of 13 human HCC collectives revealed considerable upregulation of TGF-β2 as compared to normal liver. Our study demonstrates upregulation of TGF-β2 in liver disease and suggests TGF-β2 as a promising therapeutic target for tackling fibrosis and HCC.
Article
Full-text available
Bmi1 is a member of the polycomb group family of proteins, and it drives the carcinogenesis of various cancers and governs the self-renewal of multiple types of stem cells. Our previous studies have revealed that Bmi1 acts as an oncogene in hepatic carcinogenesis in an INK4a/ARF locus independent manner. However, whether Bmi1 can be used as a potential target for hepatocellular carcinoma treatment has not been fully confirmed yet. Here, we show that perturbation of Bmi1 expression by using short hairpin RNA can inhibit the tumorigenicity and tumor growth of hepatocellular carcinoma cells both in vitro and in vivo. Importantly, Bmi1 knockdown can block the tumor growth, both in the initiating stages and the fast growing stages. Cellular biology analysis revealed that Bmi1 knockdown induces cell cycle arrest and apoptosis. Our findings verify Bmi1 as a qualified treatment target for hepatocellular carcinoma (HCC) and support Bmi1 targeting treatment with chemotherapeutic agents.
Article
Full-text available
The Polycomb group of epigenetic enzymes represses expression of developmentally regulated genes in many eukaryotes. This group includes the Polycomb repressive complex 1 (PRC1), which ubiquitylates nucleosomal histone H2A Lys 119 using its E3 ubiquitin ligase subunits, Ring1B and Bmi1, together with an E2 ubiquitin-conjugating enzyme, UbcH5c. However, the molecular mechanism of nucleosome substrate recognition by PRC1 or other chromatin enzymes is unclear. Here we present the crystal structure of the human Ring1B-Bmi1-UbcH5c E3-E2 complex (the PRC1 ubiquitylation module) bound to its nucleosome core particle substrate. The structure shows how a chromatin enzyme achieves substrate specificity by interacting with several nucleosome surfaces spatially distinct from the site of catalysis. Our structure further reveals an unexpected role for the ubiquitin E2 enzyme in substrate recognition, and provides insight into how the related histone H2A E3 ligase, BRCA1, interacts with and ubiquitylates the nucleosome.
Article
Full-text available
Bmi1 gene overexpression is found in various human tumors and has been shown as a potential target for gene treatment. However, siRNA-based treatments targeting Bmi1 gene have been restricted to limited delivery, low bioavailability and hence relatively reduced efficacy. To overcome these barriers, we developed a folate receptor targeted co-delivery system folate-doxorubicin/Bmi1 siRNA liposome (FA-DOX/siRNA-L). The FA-DOX/siRNA-L was prepared through electrostatic interaction between folate doxorubicin liposome (FA-DOX-L) and Bmi1 siRNA. In vitro and in vivo studies showed that FA-DOX/siRNA-L inhibited tumor growth by combinatory role of Bmi1 siRNA and doxorubicin (DOX). Co-delivery of Bmi1 siRNA and DOX by FA-DOX/siRNA-L showed significantly higher efficacy than sole delivery of either DOX or Bmi1 siRNA. Real-time PCR and western blot analysis showed that FA-DOX/siRNA-L silenced the expression of Bmi1 gene. In addition, higher accumulation of the siRNA and DOX in tumor cells indicated that folate ligand displayed tumor targeting effect. These results suggest that Bmi1 is an effective therapeutic target for siRNA based cancer treatment that can be further improved by co-delivery of DOX through targeted liposomes.
Article
Full-text available
Chromatin modifying activities inherent to polycomb repressive complexes PRC1 and PRC2 play an essential role in gene regulation, cellular differentiation, and development. However, the mechanisms by which these complexes recognize their target sites and function together to form repressive chromatin domains remain poorly understood. Recruitment of PRC1 to target sites has been proposed to occur through a hierarchical process, dependent on prior nucleation of PRC2 and placement of H3K27me3. Here, using a de novo targeting assay in mouse embryonic stem cells we unexpectedly discover that PRC1-dependent H2AK119ub1 leads to recruitment of PRC2 and H3K27me3 to effectively initiate a polycomb domain. This activity is restricted to variant PRC1 complexes, and genetic ablation experiments reveal that targeting of the variant PCGF1/PRC1 complex by KDM2B to CpG islands is required for normal polycomb domain formation and mouse development. These observations provide a surprising PRC1-dependent logic for PRC2 occupancy at target sites in vivo.
Article
Full-text available
Tumor recurrence following treatment remains a major clinical challenge. Evidence from xenograft models and human trials indicates selective enrichment of cancer-initiating cells (CICs) in tumors that survive therapy. Together with recent reports showing that CIC gene signatures influence patient survival, these studies predict that targeting self-renewal, the key ‘stemness’ property unique to CICs, may represent a new paradigm in cancer therapy. Here we demonstrate that tumor formation and, more specifically, human colorectal CIC function are dependent on the canonical self-renewal regulator BMI-1. Downregulation of BMI-1 inhibits the ability of colorectal CICs to self-renew, resulting in the abrogation of their tumorigenic potential. Treatment of primary colorectal cancer xenografts with a small-molecule BMI-1 inhibitor resulted in colorectal CIC loss with long-term and irreversible impairment of tumor growth. Targeting the BMI-1–related self-renewal machinery provides the basis for a new therapeutic approach in the treatment of colorectal cancer.
Article
Full-text available
Calculation of sample size is one of the important component of design of any research including animal studies. If a researcher select less number of animals it may lead to missing of any significant difference even if it exist in population and if more number of animals selected then it may lead to unnecessary wastage of resources and may lead to ethical issues. In this article, on the basis of review of literature done by us we suggested few methods of sample size calculations for animal studies.
Article
Transforming growth factor (TGF)-β is a crucial enforcer of immune homeostasis and tolerance, inhibiting the expansion and function of many components of the immune system. Perturbations in TGF-β signaling underlie inflammatory diseases and promote tumor emergence. TGF-β is also central to immune suppression within the tumor microenvironment, and recent studies have revealed roles in tumor immune evasion and poor responses to cancer immunotherapy. Here, we present an overview of the complex biology of the TGF-β family and its context-dependent nature. Then, focusing on cancer, we discuss the roles of TGF-β signaling in distinct immune cell types and how this knowledge is being leveraged to unleash the immune system against the tumor.
Article
Epstein-Barr virus (EBV) causes human lymphoid malignancies, and the EBV product latent membrane protein 1 (LMP1) has been identified as an oncogene in epithelial carcinomas such as nasopharyngeal carcinoma (NPC). EBV can epigenetically reprogram lymphocyte-specific processes and induce cell immortalization. However, the interplay between LMP1 and the NPC host cell remains largely unknown. Here, we report that LMP1 is important to establish the Hox gene expression signature in NPC cell lines and tumor biopsies. LMP1 induces repression of several Hox genes in part via stalling of RNA polymerase II (RNA Pol II). Pol II stalling can be overcome by irradiation involving the epigenetic regulator TET3. Furthermore, we report that HoxC8, one of the genes silenced by LMP1, has a role in tumor growth. Ectopic expression of HoxC8 inhibits NPC cell growth in vitro and in vivo, modulates glycolysis and regulates the expression of tricarboxylic acid (TCA) cycle-related genes. We propose that viral latency products may repress via stalling key mediators that in turn modulate glycolysis.Oncogene advance online publication, 9 March 2015; doi:10.1038/onc.2015.53.