Journal Pre-proof Forkhead Box M1 transcription factor drives liver inflammation linking to hepatocarcinogenesis in mice Tomohide Kurahashi, Yuichi Yoshida, Satoshi Ogura, Mayumi Egawa, Kunimaro Furuta, Hayato Hikita, Takahiro Kodama, Ryotaro Sakamori, Shinichi Kiso, Yoshihiro Kamada, I-Ching Wang, Hidetoshi Eguchi, Eiichi Morii, Yuichiro Doki, Masaki Mori, Vladimir V. Kalinichenko, Tomohide Tatsumi, Tetsuo Takehara
PII: DOI: Reference:
S2352-345X(19)30144-4 https://doi.org/10.1016/j.jcmgh.2019.10.008 JCMGH 537
To appear in: Cellular and Molecular Gastroenterology and Hepatology Accepted Date: 15 October 2019 Please cite this article as: Kurahashi T, Yoshida Y, Ogura S, Egawa M, Furuta K, Hikita H, Kodama T, Sakamori R, Kiso S, Kamada Y, Wang I-C, Eguchi H, Morii E, Doki Y, Mori M, Kalinichenko VV, Tatsumi T, Takehara T, Forkhead Box M1 transcription factor drives liver inflammation linking to hepatocarcinogenesis in mice, Cellular and Molecular Gastroenterology and Hepatology (2019), doi: https://doi.org/10.1016/j.jcmgh.2019.10.008. This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain. © 2019 The Authors. Published by Elsevier Inc. on behalf of the AGA Institute.
FoxM1
Forkhead Box M1 transcription factor drives liver inflammation linking to hepatocarcinogenesis in mice CCL2
Hepatocytes
Macrophage Recruitment
Normal Liver
Liver Inflammation
Chronic hepatitis
Liver Fibrosis
Cirrhosis
Liver Cancer
HCC
1
Forkhead Box M1 transcription factor drives liver inflammation linking to
2
hepatocarcinogenesis in mice
3
Tomohide Kurahashi1#, Yuichi Yoshida1#*, Satoshi Ogura1, Mayumi Egawa1, Kunimaro Furuta1,
4
Hayato Hikita1, Takahiro Kodama1, Ryotaro Sakamori1, Shinichi Kiso1, Yoshihiro Kamada1, 2,
5
I-Ching Wang3,4, Hidetoshi Eguchi5, Eiichi Morii6, Yuichiro Doki5, Masaki Mori5, Vladimir V
6
Kalinichenko3, Tomohide Tatsumi1, Tetsuo Takehara1*
7
8
9
10
11
12
1
Department of Gastroenterology and Hepatology, Graduate School of Medicine, Osaka University,
Suita, Osaka, Japan 2
Department of Molecular Biochemistry and Clinical Investigation, Graduate School of Medicine,
Osaka University, Suita, Osaka, Japan 3
Center for Lung Regenerative Medicine, Divisions of Pulmonary Biology and Developmental
13
Biology, Cincinnati Children’s Hospital Medical Center, College of Medicine of the University of
14
Cincinnati, Cincinnati, Ohio, USA
15
16
17
18
19
4
Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan.
5
Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University,
Suita, Osaka, Japan 6
Department of Pathology, Graduate School of Medicine, Osaka University, Suita, Osaka, Japan
#
Tomohide Kurahashi and Yuichi Yoshida contributed equally to this work. 1
20
*Co-corresponding authors:
21
Tetsuo Takehara, MD, PhD, Department of Gastroenterology and Hepatology, Osaka University
22
Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan. e-mail:
23
[email protected]; tel: (81) 6-6879-3621, fax: (81) 6-6879-3629.
24
Yuichi Yoshida, MD, PhD, Department of Gastroenterology and Hepatology, Osaka University
25
Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan. e-mail:
26
[email protected]; tel: (81) 6-6879-3621, fax: (81) 6-6879-3629.
27
Short Title
28
Hepatocyte FoxM1 promotes liver inflammation.
29
Abbreviations
30
ALT, alanine aminotransferase; AS, antisense; CCL2, chemokine (C-C motif) ligand 2; CCl4, carbon
31
tetrachloride; DOX, doxycycline; FoxM1, Forkhead Box M1 transcription factor; GAPDH,
32
glyceraldehyde-3-phosphate dehydrogenase; HCC, hepatocellular carcinoma; HFHC,
33
high-fat/high-cholesterol; RT-PCR, reverse transcription polymerase chain reaction; S, sense; Tet,
34
tetracycline; TG, transgenic; WT, wild-type.
35
Acknowledgments
36
The authors thank Dr. Pradip Raychaudhuri (University of Illinois at Chicago) for providing
37
CMV-T7-FoxM1 and 6×CDX2-Luc plasmids.
38
Author Contributions 2
39
Study concept and design: T. Kurahashi, Y. Yoshida, S. Ogura, and T. Takehara. Acquisition of data:
40
T. Kurahashi, S. Ogura, M. Egawa, K. Furuta. Analysis and interpretation of data: T. Kurahashi, Y.
41
Yoshida, S. Ogura, M. Egawa, K. Furuta, H. Hikita, T. Kodama, R. Sakamori, S. Kiso, Y. Kamada, I.
42
C. Wang, H. Eguchi, E Morii, Y. Doki, M. Mori, V. V. Kalinichenko, T. Tatsumi, and T. Takehara.
43
Writing, review, or revision of the manuscript: T. Kurahashi, Y. Yoshida, and T. Takehara.
44
Conflicts of interest
45
The authors disclose no conflicts.
46
Funding
47
This work was supported in part by a Grant-in-Aid for Scientific Research from the Japan Society for
48
the Promotion of Science.
49
Word count (abstract): 244
50
Word count (excluding title page, methods, tables/figures, or references): 3207
51
Number of Tables and Figures 1 table and 11 figures
52
3
53
SYNOPSIS
54
Hepatocyte-specific FoxM1 transgenic (TG) mice develop spontaneous liver inflammation,
55
fibrosis and carcinogenesis. Spontaneous liver inflammation in TG mice was associated with
56
increased expression of chemokine CCL2. FoxM1-specific inhibitor effectively reduces liver
57
inflammation and CCL2 expression in models of liver injury.
58
4
59
ABSTRACT
60
BACKGROUND & AIMS: Liver inflammation has been recognized as a hallmark of
61
hepatocarcinogenesis. Although Forkhead Box M1 (FoxM1) is a well-defined oncogenic
62
transcription factor that is overexpressed in hepatocellular carcinoma (HCC), its role in liver
63
inflammation has never been explored.
64
METHODS: We generated hepatocyte-specific FoxM1 conditional transgenic (TG) mice using the
65
Cre-loxP and Tetracycline (Tet)-on systems to induce FoxM1 expression in a hepatocyte-specific and
66
time-dependent manner.
67
RESULTS: After treatment of Tet-derivatives doxycycline (DOX) to induce FoxM1, TG mice
68
exhibited spontaneous development of hepatocyte death with elevated serum ALT levels and hepatic
69
infiltration of macrophages. The removal of DOX in TG mice completely removed this effect,
70
suggesting that spontaneous inflammation in TG mice occurs in a hepatocyte FoxM1-dependent
71
manner. In addition, liver inflammation in TG mice was associated with increased levels of hepatic
72
and serum chemokine (C-C motif) ligand 2 (CCL2). In vitro transcriptional analysis confirmed that
73
CCL2 is a direct target of FoxM1 in murine hepatocytes. After receiving FoxM1 induction since
74
birth, all TG mice exhibited spontaneous HCC with liver fibrosis at 12 months of age. Hepatic
75
expression of FoxM1 was significantly increased in liver injury models. Finally, pharmacological
76
inhibition of FoxM1 reduced liver inflammation in models of liver injury.
77
CONCLUSIONS: Hepatocyte FoxM1 acts as a crucial regulator to orchestrate liver inflammation 5
78
linking to hepatocarcinogenesis. Thus, hepatocyte FoxM1 may be a potential target not only for the
79
treatment of liver injury but also for the prevention toward HCC.
80
Keywords: FoxM1; Liver inflammation; CCL2
81
6
82
Introduction
83
Hepatocellular carcinoma (HCC) is one of the most common types of malignant neoplasms
84
among primary liver cancers and a major cause of cancer-related deaths worldwide.1 It is well known
85
that HCC occurs in patients with chronic liver diseases, including hepatitis virus infections, alcoholic
86
or nonalcoholic fatty liver diseases, and other liver diseases.2 Irrespective of the etiology, chronic
87
liver inflammation promotes subsequent hepatic wound-healing responses, eventually leading to the
88
development of liver fibrosis and HCC.3, 4 Thus, the molecular mechanisms driving liver
89
inflammation to HCC need to be better understood to identify a therapeutic strategy to prevent HCC
90
in patients with chronic liver diseases.
91
The Forkhead box M1 (FoxM1) transcription factor is a member of the Fox family of
92
proteins that share a highly conserved structure in the winged helix DNA-binding domain.5-7 Several
93
studies have shown that FoxM1 acts as an important regulator for cell proliferation and is frequently
94
expressed at a higher level than normal in a variety of human cancers.7-12 Previous animal studies
95
have shown that FoxM1 is critical for cancer cell proliferation of HCC.13, 14 We previously reported
96
that a high FoxM1 expression defines poor prognosis in patients with HCC after surgical resection15
97
and that FoxM1 is regulated via the mevalonate pathway of lipid metabolism in hepatoma cell
98
lines.16 Consistent with our observations, FoxM1 was shown to be overexpressed in tumor tissues of
99
hepatitis B virus (HBV)-related HCC compared with the surrounded non-tumor tissues.17 In addition,
100
FoxM1 expression has been also shown to be increased in the livers of chronic hepatitis or liver 7
101
cirrhosis patients with HBV infection compared with the normal liver samples,17 suggesting that
102
FoxM1 may be involved in the initial step of hepatocarcinogenesis during the development of liver
103
injury.
104
In adult human organs, FoxM1 is abundantly expressed in testis, thymus, intestine, and
105
colon.18 By contrast, no FoxM1 expression is present in adult murine livers under normal
106
conditions;18 however, it is induced in the livers during liver regeneration after partial hepatectomy19
107
and during toxin-induced liver injury.20 These results suggest that FoxM1 may be involved in not
108
only hepatocyte proliferation but also liver inflammation. Consistent with these results, a previous
109
study demonstrated that FoxM1 is required for allergen-mediated goblet cell proliferation and
110
pulmonary inflammation through the induction of inflammatory cytokines or chemokines in a murine
111
model of chronic lung disorder.21 However, to date, the role of hepatocyte FoxM1 in liver
112
inflammation during the development of liver injury has remained unclear.
113
In this study, we hypothesized that hepatocyte FoxM1 affects the inflammatory
114
microenvironment of the liver. To clarify this issue, we developed a new murine model in which
115
ectopic overexpression of FoxM1 in hepatocytes is controlled in a time-dependent manner.
116
8
117
Results
118
Overexpression of FoxM1 in Murine Livers Causes Spontaneous Liver Injury and Fibrosis
119
To test whether increased FoxM1 expression is sufficient to drive liver injury, we
120
established hepatocyte-specific FoxM1 conditional TG (TetO7-FoxM1 tg/tg/
121
Rosa26-LSL-rtTA/Alb-Cretg/-) mice using the Tet-on and Cre-loxP systems (Figure 1A). To induce
122
FoxM1 expression, TG and WT (TetO7-FoxM1 tg/tg /Rosa26-LSL-rtTA/Alb-Cre-/-) mice were treated
123
with DOX. Western blot and immunohistochemical analyses using antibody against FoxM1
124
confirmed that ectopic overexpression of FoxM1 was induced in a liver- and DOX-specific manner
125
(Figure 1B-D). At 13 weeks of age, TG mice showed spontaneous elevations of serum ALT levels
126
and hepatocyte death, confirmed by an increased number of TUNEL-positive hepatocytes (Figure
127
1E-G). Furthermore, TG mice showed spontaneous liver fibrosis, confirmed by collagen-specific
128
picrosirius red staining or immunostaining for α-smooth muscle actin, a marker of activated hepatic
129
stellate cells (Figure 1H and I). This spontaneous liver fibrosis in TG mice was further assessed by
130
the increased expression of fibrosis-related genes in the livers (Figure 1J). Taken together,
131
overexpression of FoxM1 in the livers induced spontaneous liver injury and fibrosis.
132
Short-Term Overexpression of FoxM1 Induces Reversible Liver Inflammation With Macrophage
133
Recruitment
9
134
To investigate whether FoxM1 itself has a direct effect on spontaneous liver injury in TG
135
mice, we introduced transient overexpression of FoxM1 at 8 weeks of age for 3 days and repressed
136
its expression by removing DOX (Figure 2A and B). After 3 days of DOX treatment, TG mice
137
exhibited elevated serum ALT levels and hepatocyte death, confirmed by TUNEL staining; this
138
spontaneous liver injury was completely reversible by removing DOX (Figure 2C-E). Furthermore,
139
the increased liver injury in TG mice was associated with hepatic infiltration of inflammatory cells,
140
consisting of F4/80 positive macrophages; this spontaneous liver inflammation was also completely
141
reversed by removing DOX (Figure 2F). In addition, the fluorescence-activated cell sorting analysis
142
confirmed that the number of CD11b+F4/80+ macrophages was increased in the livers of TG mice in
143
a DOX-dependent manner (Figure 2G). These data indicate that short-term overexpression of FoxM1
144
is sufficient to induce liver injury and inflammation with macrophage recruitment.
145
To investigate the molecular mechanisms underlying the spontaneous recruitment of
146
hepatic macrophages in TG mice, we examined the gene expression of macrophage-related markers
147
in the livers of TG mice. TG mice showed a significant increase in the hepatic gene expression of
148
macrophage-related markers, including F4/80, Cd68, Nos2, Tnfα, Ccl2, Ccr2, Ccl5, Arg1, and Cd206
149
(Figure 2H). Among these markers, Ccl2 showed a 145-fold higher hepatic expression in TG mice
150
than in WT mice (Figure 2H). Consistently, 3-day DOX treatment resulted in increased serum CCL2
151
levels in TG mice, and this increase was completely reversed by removing DOX (Figure 2I).
10
152
To rule out the possibility that CCL2 induction results from increased liver injury in TG
153
mice, we overexpressed FoxM1 for 1 day with DOX (Figure 2J). There was no apparent increase in
154
serum ALT levels and no difference in serum ALT levels in WT and TG mice after 1 day of DOX
155
treatment (Figure 2K). However, there was a significant increase in hepatic gene expression of Ccl2
156
compared with WT mice at this earlier, 1 day time point (Figure 2L), suggesting that the increased
157
expression of hepatic Ccl2 gene in TG mice might occur prior to induction of liver injury.
158
Next, we titrated the levels of DOX in the drinking water and developed TG mice with
159
lower FoxM1 expression using low dose of DOX (DOX low dose: 0.01 mg/mL) (Figure 2M). Even
160
DOX low dose induced the moderate expression of FoxM1 (27.4%; Figure 2N) and an increase in
161
TUNEL-positive hepatocytes (Figure 2O) and serum ALT levels (Figure 2P) as was observed with
162
the higher dose of DOX (DOX on: 0.2 mg/mL). Furthermore, DOX low dose increased Ccl2 gene
163
expression in livers of TG mice as observed with DOX on (Figure 2Q).
164
CCL2 as a Direct Target of FoxM1 in Hepatocytes
165
To investigate whether hepatocyte FoxM1 regulates the expression of CCL2, we next
166
performed in vitro experiments using murine hepatocyte cell lines. si-RNA-mediated knock down of
167
FoxM1 resulted in a significant decrease in the gene expression of Ccl2 and protein expression of
168
CCL2 in murine hepatocytes cell lines BNL-CL2 (Figure 3A and B) and AML12 (Figure 3C and D).
11
169
We then performed in vitro transcriptional analysis to investigate whether CCL2 is a direct
170
target of FoxM1. One potential FoxM1 binding site was identified in the −2468/+67 bp promoter
171
region of the murine Ccl2 gene at −1343/−1338 bp (Figure 3E). Co-transfection of FoxM1
172
expression vector resulted in a significant increased activity of the −1401/+67 bp Ccl2 luciferase
173
reporter, and the deletion of the FoxM1 binding site in the Ccl2 promoter region −1136/+67 bp
174
abolished the capacity of FoxM1 to stimulate this activity, indicating that −1343/−1338 bp in the
175
Ccl2 promoter region functions a FoxM1 binding site (Figure 3E). To further confirm the binding of
176
FoxM1 to the promoter region of the Ccl2 gene, the chromatin immunoprecipitation assay was
177
performed in murine hepatocyte BNL-CL2 cells using two antibodies against FoxM1. This assay
178
showed the specific binding of FoxM1 protein to the Ccl2 promoter DNA (Figure 3F). Collectively,
179
the data indicate that CCL2 is a direct target of FoxM1 in hepatocytes and suggest the involvement
180
of hepatic CCL2 induction in the spontaneous macrophage recruitment of TG livers.
181
Hepatocytes Are the Major Source of CCL2 Production in TG Mice
182
To investigate which cell population produces CCL2 in TG mice, we examined the gene
183
expression of Ccl2 and the protein expression of CCL2 in hepatocytes and non-parenchymal cells
184
(NPCs) isolated from livers of WT and TG mice after 3 days of DOX treatment. Hepatocytes of TG
185
mice showed a significant increase in Ccl2 gene expression compared to those of WT mice, whereas
186
that in NPCs was comparable between the two groups (Figure 4A). Western blot analysis showed the
12
187
increased expression of CCL2 protein in hepatocytes of TG mice compared with those of WT mice
188
(Figure 4B). These data suggest that the major source of CCL2 is hepatocytes rather than NPCs in
189
TG mice.
190
FoxM1 Induction Has a Modest Effect on Cell Death in Cultured Hepatocytes in vitro
191
We investigated the effect of FoxM1 induction on hepatocyte cell viability and death in
192
vitro using primary cultured hepatocytes isolated from WT and TG mice. WT and TG hepatocytes
193
were cultured in vitro and were treated with 100 ng/ml of DOX for 24 hours. Western blot analysis
194
confirmed that FoxM1 protein was induced in TG hepatocytes treated with DOX (Figure 5A).
195
Consistent with in vivo data, in vitro treatment with DOX increased in Ccl2 gene expression and
196
CCL2 protein expression in the supernatant of TG hepatocytes but not in the supernatants of WT
197
hepatocytes (Figure 5B-E). The WST assay showed that DOX treatment resulted in a modest
198
reduction (0.95-fold) in cell viability of TG hepatocytes (Figure 5F and G) and the caspase 3/7
199
activity assay showed a modest increase (2.03-fold) in cell death of TG hepatocyte (Figure 5H and I).
200
Among cell cycle-related genes, gene expression of Ccnb1 and Skp2, known FoxM1-target genes,
201
increased in TG hepatocytes treated with DOX (Figure 5J-O). These in vitro data suggest that FoxM1
202
induction has a modest effect on hepatocyte death.
203
Hepatocyte-Specific CCL2 Inhibition Reduces Liver Injury in TG Mice
13
204
To investigate whether hepatocyte-derived CCL2 plays a role on the development of
205
spontaneous liver injury in TG mice, we inhibited CCL2 expression using hepatocyte-specific
206
N-acetylgalactosamine (GalNAc)-siRNA system.22 Subcutaneous administration of
207
GalNAc-conjugated siRNA against murine Ccl2 (GalNAc-siCcl2) reduced gene expression of
208
hepatic Ccl2 and serum levels of CCL2 compared with administration of GalNAc-siControl (control)
209
in TG mice after 3 days of DOX treatment, confirming that hepatocytes are the major source of
210
CCL2 production in TG mice (Figure 6A). Subcutaneous administration of GalNAc-siCcl2 also
211
reduced liver injury (Figure 6B) as shown by a reduced serum ALT levels and a reduced number of
212
TUNEL-positive hepatocytes compared with the control in TG mice after 3 days of DOX treatment
213
(Figure 6C-E). Furthermore, subcutaneous administration of GalNAc-siCcl2 reduced infiltration of
214
F4/80 positive macrophages compared with control in TG mice after 3 days of DOX treatment
215
(Figure 6F). Although TG mice did not show apparent liver fibrosis assessed by picrosirius red
216
staining after 3 days of DOX treatment (Figure 6G and H), subcutaneous administration of
217
GalNAc-siCcl2 resulted in a significant reduction of gene expression of hepatic Col1α2 compared
218
with control in TG mice after 3 days of DOX treatment (Figure 6I). Collectively, these data suggest
219
that hepatocyte-derived CCL2 contributes to liver injury in TG mice.
220
Spontaneous Liver Injury in TG Mice Occurs Through Hepatic Macrophage Recruitment Induced
221
by FoxM1 Expression
14
222
To investigate the consequence of hepatic macrophage recruitment in liver injury of TG
223
mice, we depleted the macrophages of these mice. Immunohistochemical or real-time RT-PCR
224
analysis showed that the administration of clodronate liposomes resulted in the successful depletion
225
of F4/80 positive macrophages in the livers of WT and TG mice (Figure 7A and B).
226
Macrophage-depleted TG mice exhibited lower serum ALT levels and TUNEL-positive hepatocytes
227
than PBS liposomes-treated TG mice (Figure 7C-E). These data suggest that spontaneous hepatocyte
228
death in TG mice is associated with hepatic macrophage recruitment induced by FoxM1 expression.
229
FoxM1-Dependent Liver Inflammation Leads to Hepatocarcinogenesis
230
In patients with chronic liver diseases, continuous liver inflammation and fibrosis is known
231
to lead to tumorigenesis. Therefore, we investigated whether long-term overexpression of FoxM1
232
affects hepatocarcinogenesis. At 48 weeks of age, although all TG mice treated with DOX after birth
233
were found to develop liver tumors, WT mice did not develop any tumors (Figure 8A-C).
234
Histological analysis showed that liver tumors in TG mice were hepatocellular carcinoma (HCC),
235
because tumor cells were large with hyperchromatic nuclei in compact growth pattern and did not
236
form any tubular structure (Figure 8B). In addition, liver tumors in TG mice showed increased
237
expression of Arginase-1 and Glypican-3, known makers of HCC, 23, 24 confirming that these tumors
238
are HCC (Figure 8B). The Ki-67 immunohistochemistry confirmed that liver tumors in TG mice
239
showed higher cell proliferation than the non-tumor regions of TG mice or the livers of WT mice
15
240
(Figure 8D, E). Furthermore, picrosirius red staining and increased hepatic expression of
241
fibrosis-related genes showed that the non-tumor regions of TG mice developed liver fibrosis (Figure
242
8F-H). Our data indicate that overexpression of FoxM1 in hepatocytes results in liver injury and
243
subsequent liver inflammation and fibrosis, leading to hepatocarcinogenesis.
244
Hepatic FoxM1 Is Induced in Humans and Mice With Chronic Liver Injury
245
To confirm the significance of FoxM1 in liver injury, we examined the expression of
246
FoxM1 in the liver tissues of patients with chronic liver disease, such as chronic hepatitis B, chronic
247
hepatitis C, nonalcoholic steatohepatitis, and liver cirrhosis. As we reported previously,15, 16 an
248
immunohistochemical analysis confirmed increased FoxM1expression in HCC tissues (Figure 9A).
249
This analysis also showed higher FoxM1 expression in liver tissues of chronic liver diseases
250
compared with normal liver tissues (Figure 9A). To confirm an increase in hepatic FoxM1 expression
251
during the development of chronic liver injury, we next used two murine models of chronic liver
252
injury induced by a high-fat/high-cholesterol (HFHC) diet or chronic administration of CCl4. The
253
gene expression of Foxm1 was increased as serum ALT levels were elevated in both models (Figure
254
9B). Immunohistochemical analysis also confirmed a higher hepatic expression of FoxM1 in chronic
255
liver disease tissues than in normal liver tissues (Figure 9C). Collectively, these data suggest a role of
256
FoxM1 in liver injury.
257
FoxM1 Inhibitor Reduces Liver Inflammation in a Liver Injury Model 16
258
We finally investigated whether the pharmacological inhibition of FoxM1 reduces liver
259
inflammation during the development of liver injury. For this purpose, we used a murine model of
260
liver injury fed a HFHC diet, which induced hepatic FoxM1 expression (Figure 9B and C). Mice fed
261
a HFHC diet were treated with a known FoxM1 inhibitor, thiostrepton.25, 26 The thiostrepton-treated
262
mice showed significantly lower serum ALT levels and fewer TUNEL-positive hepatocytes than the
263
control mice (Figure 10A-D). The thiostrepton-treated mice also showed significantly fewer F4/80
264
positive macrophages in the livers than the control mice (Figure 10E). Furthermore, thiostrepton
265
treatment resulted in a decrease of hepatic Ccl2 expression or serum CCL2 levels in mice with
266
HFHC diet (Figure 10F, G). On the other hand, thiostrepton treatment did not affect hepatocyte
267
proliferation in HFHC diet-fed mice (Figure 10H and I). The anti-inflammatory effect of thiostrepton
268
was also confirmed in TG mice, showing that thiostrepton treatment effectively reduced spontaneous
269
liver inflammation without significant anti-proliferation effect on hepatocytes (Figure 11A-I). These
270
findings suggest that FoxM1 inhibition may represent a therapeutic approach against liver injury.
17
271
272
DISCUSSION
Targeting liver inflammation is crucial for the development of new treatments against
273
chronic liver diseases.27, 28 Here, we identified hepatocyte FoxM1 as a key driver to trigger chronic
274
liver inflammation. Similar to the clinical course of patients with chronic liver disease,
275
hepatocyte-specific FoxM1 transgenic mice developed spontaneous liver inflammation and fibrosis,
276
leading to hepatocarcinogenesis. This FoxM1-dependent liver inflammation with hepatic
277
macrophage recruitment was associated with hepatocyte-derived chemokine CCL2 that was
278
identified as a novel direct target of FoxM1. Finally, we showed that inhibiting FoxM1 with its
279
specific inhibitor effectively reduced liver inflammation in a model of liver injury induced by HFHC
280
diet. Thus, our current study raised a possibility that targeting FoxM1 could be a novel therapeutic
281
approach for liver injury toward HCC.
282
FoxM1 has a cell-autonomous role in forcing the cell proliferation of a variety of cancers,
283
including HCC.29 Several lines of evidence demonstrated that FoxM1 is overexpressed in cancer
284
tissues compared with surrounding non-cancer tissues.7-12 However, little is known whether chronic
285
tissue damage might affect FoxM1 expression in the damaged tissues. According to previous reports,
286
FoxM1 is more expressed in the damaged lung tissues of patients with idiopathic pulmonary fibrosis
287
than those of normal patients30 and that FoxM1 expression is also increased in human gastritis tissues
288
with Helicobacter pylori infection compared with normal gastric tissues.31 In this study, we showed
289
that FoxM1 expression was induced in the damaged livers of murine models and was implicated in 18
290
human specimens of chronic liver diseases. These data indicate that FoxM1 might be involved in the
291
initial process of liver damage and inflammation prior to HCC development.
292
Chronic inflammation is a key characteristic of chronic liver disease.27, 28 The interaction
293
between hepatocytes and non-parenchymal cells via a variety of mediators such as cytokines or
294
chemokines32 has been shown to be crucial for driving liver inflammation; however, the signaling
295
pathways in hepatocytes are not yet fully understood. In this study, we showed that increased FoxM1
296
in murine livers resulted in enhanced liver inflammation. Among inflammatory mediators that were
297
increased in TG livers, CCL2 was found to have a putative FoxM1 binding site in its promoter region,
298
and in vitro experiments confirmed that FoxM1 is able to regulate CCL2 directly in hepatocytes.
299
CCL2 is a well-defined chemokine that recruits CCR2 positive monocytes/macrophages to trigger
300
liver inflammation and promote hepatocarcinogenesis.33, 34 Consistently, spontaneous liver
301
inflammation in TG mice was associated with an increased macrophage recruitment in the injured
302
livers, suggesting a novel cell-nonautonomous role of hepatocyte FoxM1 to trigger chronic liver
303
inflammation via direct regulation of inflammatory mediators and recruitment of inflammatory cells.
304
Therapeutic strategies to target FoxM1 in liver diseases, especially in HCC, has been
305
investigated with in vivo and in vitro experiments so far.14, 16 It has been shown that the inhibition of
306
FoxM1 activity effectively reduces liver tumors in a murine model of HCC.14 We recently
307
demonstrated that statins, well known cholesterol-lowering drugs, also reduce FoxM1 expression and
308
thereby induce cell death in human HCC cells.16 It has been reported that hepatocyte-specific FoxM1 19
309
knockout mice exhibit impaired liver regeneration after partial hepatectomy19 and
310
macrophage-specific FoxM1 knockout mice show delayed wound-healing response and worsened
311
liver function in a liver injury model induced by CCl4,35 implying that FoxM1 inhibition may worsen
312
liver damage and inflammation. In this study, however, we showed that thiostrepton, a
313
well-established FoxM1 inhibitor,25, 26 effectively improved liver damage and reduced liver
314
inflammation with macrophage recruitment and CCL2 expression in a murine liver injury model
315
induced by HFHC diet. Consistent with our data, a recent publication demonstrated that a FoxM1
316
inhibitor effectively reduces lung inflammation in response to house dust mite allergens in mice.36
317
These findings suggest that FoxM1 inhibitors target the cell-nonautonomous function of FoxM1
318
rather than its cell-autonomous function in vivo. Further studies will be needed to elucidate this issue.
319
In conclusion, we have demonstrated that hepatocyte FoxM1 drives inflammatory response
320
linking liver fibrosis and carcinogenesis via induction of inflammatory mediators. Because HCC
321
arises in a background of liver inflammation and fibrosis, targeting hepatocyte FoxM1 would be a
322
potential application not only for the treatment of liver injury but also for early intervention or
323
prevention toward HCC.
324
20
325
Materials and Methods All authors had access to the study data and had reviewed and approved the final
326
327
manuscript.
328
Mice
329
Albumin promoter-driven Cre recombinase (Alb-Cre) transgenic C57BL/6 mice (The
330
Jackson Laboratory, Bar Harbor, ME, USA) were crossed with TetO-GFP-FoxM1-∆N mice
331
(TetO7-FoxM1)
332
(rtTA)
333
Rosa26-LSL-rtTA/Alb-Cretg/-) and wild-type (WT, TetO7-FoxM1
334
mice were treated with low dose (DOX low dose: 0.01 mg/dL) or regular dose (DOX on: 0.2 mg/dL)
335
doxycycline hyclate (catalog D9891: Sigma-Aldrich, Saint Louis, MO, USA) dissolved in 5%
336
sucrose (catalog 30403-55, Nacalai Tesque, Kyoto, Japan) and supplied as drinking water to induce
337
hepatocyte-specific FoxM1 expression. The Alb-Cre transgene was detected using the following
338
sense (S) and antisense (AS) primers: 5′-GCGGCATGGTGCAAGTTGAAT-3′ (S) and
339
5′-CGTTCACCGGCATCAACGTTT-3′ (AS). PCR analysis was performed to detect for the
340
TetO-GFP-FoxM1-∆N
341
5′-CGACAAGCAGAAGAACGGCATC-3′ (S) and 5′-AGTAGGGAAAGTGGTCCTCAATCC-3′
342
(AS). The primers for detection of the ROSA26-LSL-rtTA transgene were as follows:
343
5′-GAGTTCTCTGCTGCCTCCTG-3′ (S) and 5′-AAGACCGCGAAGAGTTTGTC-3′ (AS). The
mice
37, 38
and Rosa26-loxP-STOP-loxP-reverse tetracycline transcriptional activator
(Rosa26-LSL-rtTA)
(Figure
transgene
1A).
with
21
Transgenic
the
(TG,
TetO7-FoxM1
tg/tg
/
tg/tg
/Rosa26-LSL-rtTA/Alb-Cre-/-)
following
primers:
344
wild-type
ROSA26
allele
was
detected
using
the
following
primers:
345
5′-GAGTTCTCTGCTGCCTCCTG-3′ (S) and 5′-CGAGGCGGATACAAGCAATA-3′ (AS). All
346
mice were maintained under specific pathogen-free conditions in automated watered and ventilated
347
cages on a 12-h light/dark cycle. All animal experiments of this study were performed with humane
348
care under approval from the Animal Care and Use Committee of Osaka University Medical School.
349
Real-Time Reverse Transcription Polymerase Chain Reaction
350
Total RNA was prepared using the QIAshredder and the RNeasy Mini kit (catalog 79656
351
and 74106, Qiagen, Hilden, Germany), and quantitative real-time reverse transcription polymerase
352
chain reaction (RT-PCR) was performed using the ReverTra Ace qPCR RT Master Mix (catalog
353
FSQ-201, Toyobo, Osaka, Japan) according to the manufacturer’s protocol. Quantitative real-time
354
RT-PCR reaction was performed with a THUNDERBIRD SYBR qPCR Mix (catalog
355
QPS-201,Toyobo) on a Quant Studio 6 Flex Real-Time PCR system (Thermo Fisher Scientific,
356
Waltham, MA, USA). The following primers used in this study were purchased from Qiagen: murine
357
Foxm1 (QT00120498), Gapdh (QT00199388), ColIα1 (QT00162204), ColIα2 (QT01055572), F4/80
358
(QT00099617),
359
(QT00167832), Ccl5 (QT01747165), Arg1 (QT00134288), Ccna2 (QT00102151), Ccnb1
360
(QT01757007) and Skp2 (QT00118573).
361
Sigma-Genosys:
362
5′-CCAAAGGTAACTGTCCTGGC-3′
Cd68
(QT00254051),
Nos2
(QT00100275),
Tnfα
(QT00104006),
Ccl2
The following primers were synthesized
5′-TCTGGGCTCACTATGCTGCA-3′ (AS) 22
for
(S) murine
by and
Ccr2;
363
5′-CAAAAACTGACTGGGCTTCC-3′ (S) and 5′-GCCCTTGATTCCAAAGAGTG-3′ (AS) for
364
Cd206. The expression values of the murine genes were normalized to the glyceraldehyde
365
3-phosphate dehydrogenase (Gapdh) level.
366
Serum Analysis
367
Blood samples were harvested from the inferior vena cava and centrifuged at 700 g for 10
368
minutes. Serum alanine aminotransferase (ALT) levels were measured using a colorimetric assay kit
369
(catalog 431-30901, Wako, Kyoto, Japan) according to the manufacturer’s protocol.
370
Immunohistochemical Analysis
371
Liver tissues were fixed overnight with 10% formaldehyde neutral buffer solution (catalog
372
37152-51, Nacalai Tesque), embedded in paraffin, and sectioned (4-µm thick). Tissue sections were
373
subjected to Mayer’s hematoxylin and eosin and immunohistochemical staining. We used the
374
following antibodies: rabbit anti-mouse FoxM1 (A-11, 1:500, catalog sc-271746, Santa Cruz
375
Biotechnology, Santa Cruz, CA, USA), rat anti-mouse F4/80 (1:100, catalog MCA497GA, Serotec,
376
Oxford, UK), rabbit anti-mouse α-smooth muscle actin (1:200, catalog ab5694, Abcam, Cambridge,
377
MA, USA), rabbit anti-mouse Ki-67(D3B5) (1:400, catalog 12202, Cell Signaling Technology,
378
Danvers, MA, USA) , goat anti-mouse arginase-1 (M-20,1:100, catalog sc-18355, Santa Cruz
379
Biotechnology), or mouse glypican-3 (F-3, 1:100, catalog sc-390587, Santa Cruz Biotechnology).
380
Overexpression of FoxM1 in hepatocytes of TG mice were assessed by immunohistochemical
381
staining with FoxM1 (D12D5) (1:200, catalog 5436, Cell Signaling Technology). Liver fibrosis was 23
382
quantified by measuring the picrosirius red (catalog 24901-500, Polysciences Inc., Warrington, PA,
383
USA) stained area using ImageJ (version 1.80, US National Institute of Health, Bethesda, MD, USA).
384
FoxM1 expression was quantified by measuring the area stained with anti-FoxM1 antibodies using
385
ImageJ software (version 1.80). Cells with nuclear DNA fragmentation were detected using a
386
transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) staining kit (catalog
387
S7100; Millipore, Molsheim, France) according to the manufacturer’s protocol. The numbers of
388
TUNEL or Ki-67 positive hepatocytes per view field were counted at 100× magnification.
389
Western Blot Analysis
390
Murine liver samples were homogenized in RIPA buffer containing 50 mM Tris (pH 7.5),
391
0.15 M NaCl, 0.1% SDS, 0.1% sodium deoxycholate, 1 mM EDTA, 1% Nonidet P-40, phosphatase
392
inhibitor cocktail (catalog 0574-61, Nacalai Tesque), and protease inhibitor cocktail (catalog
393
25955-11, Nacalai Tesque). For Western blot analysis, the primary antibodies anti-human FoxM1
394
(D12D5) (1:1000, catalog 5436, Cell Signaling Technology) and GAPDH (1:1000, catalog 5174,
395
Cell Signaling Technology) were used. The donkey anti-rabbit immunoglobulin G–horseradish
396
peroxidase (1:3000, catalog sc-2357, Santa Cruz Biotechnology) was used as a secondary antibody.
397
Preparation of Mononuclear Cells
398
The livers of WT or TG mice were passed through a 70-µm nylon mesh to create single
399
cell suspensions. The cell suspension was collected and parenchymal cells were isolated from
400
mononuclear cells by centrifugation at 50g for 5 min. The mononuclear cells were washed in PBS 24
401
and suspended in 40% Percol (catalog 17089101, GE Healthcare Biosciences AB, Uppsala, Sweden).
402
The suspended cells were gently overlaid onto 70% Percol (GE Healthcare Biosciences AB) and
403
centrifuged at 750g for 20 min. The mononuclear cells were collected from the interface, washed
404
twice in PBS, and used for flow cytometry.
405
Flow Cytometric Analysis
406
Prepared liver mononuclear cells were suspended in a solution of PBS, 0.3% w/v bovine
407
serum albumin, and 0.1% w/v sodium azide. To avoid nonspecific bindings of antibodies to Fc
408
receptors, the cells were pretreated with anti-CD16/32 antibody (clone 2.4G2, catalog 553141, BD
409
Biosciences, Franklin Lakes, NJ, USA) for 15 minutes. Then, the cells were stained for 20 min at
410
4°C with the following antibodies: allophyocyanin-conjugated anti-mouse CD11b (clone M1/70,
411
catalog 553312, BD Biosciences) and phycoerythrin-conjugated anti-mouse F4/80 (clone T45-2342,
412
catalog 565410, BD Biosciences) for macrophages. To label dead cells, 7-amino-actinomycin D
413
(7-AAD, catalog 559925, BD Biosciences) were used. These samples were subjected to flow
414
cytometric analysis using a fluorescence-activated cell sorting Canto II system (BD Biosciences).
415
Data were analyzed using FlowJo software (TreeStar, Ashland, OR, USA). The number of
416
CD11b+F4/80+ macrophages in a cell subset was determined using the following calculation: total
417
liver mononuclear cell number × corresponding cell subset proportion to the total cells.
418
Enzyme-Linked Immunosorbent Assay
419
The levels of serum CCL2 and secreted CCL2 protein from cell lines were measured using 25
420
MCP-1/CCL2 mouse uncoated ELISA kit (catalog 88-7391-86, Thermo Fisher Scientific) in
421
accordance with the manufacturer’s instructions.
422
Cell Culture
423
Murine primary hepatocytes and NPCs were isolated from WT and TG mice by two-step
424
collagenase-pronase perfusion as previously described.39 Briefly, the murine liver was perfused with
425
Liver Perfusion Medium (catalog 17701038; Thermo Fisher Scientific) for 5 minutes to remove
426
blood. For digestion of murine liver, the tissue was perfused with 53% pronase solution and 0.27%
427
collagenase solution for 1 and 5 minutes. All of the solutions were warmed to 37°C before use and
428
perfused at a flow rate of 4ml/min. The perfused murine liver was transferred into Dulbecco’s
429
modified Eagle’s medium containing 10% fetal calf serum and minced to release the hepatocytes.
430
The suspension was filtered through a cell strainer with a pore size of 70µm (catalog 352350,
431
Corning, NY, USA) and was washed three times with centrifugation at 50g for 1 minutes at 4°C.
432
After collecting the supernatant, the cell pellet was resuspended in William’s medium E with 10%
433
fetal calf serum. In a select experiment, the NPCs in the supernatant were pelleted at 400g for 5
434
minutes and subjected to Western blot and real-time RT-PCR analysis. Hepatocytes (25000 cells/cm2)
435
were seeded on type I collagen-coated microplate (catalog 4820-010, Iwaki Glass, Shizuoka, Japan)
436
in William’s medium E with 10% fetal calf serum, 2 mM L-glutamine, 10-7 M insulin, 10-7 M
437
dexamethasone, 100U/ml penicillin and 100U/ml streptomycin. Isolated hepatocytes with >90%
438
viability, determined by trypan blue exclusion, were cultured overnight. To induce FoxM1 protein 26
439
expression, primary hepatocytes isolated from WT and TG mice were treated with DOX for 24h.
440
After treating with DOX for 24 hours, WST assay and caspase-3/7 activity were measured in primary
441
hepatocytes of WT and TG mice using Cell count Reagent SF (catalog 07553-44, Nacalai Tesque)
442
and a luminescent substrate assay (Caspase-Glo assay, catalog G8093, Promega, Madison, WI, USA)
443
according to the manufacturer’s instructions. The murine non-transformed hepatocyte cell lines,
444
BNL-CL2 and AML12 were obtained from the American Type Culture Collection (Manassas, VA,
445
USA). For the small RNA interference assay, cells were transfected with either 20 nM murine Foxm1
446
siRNA (catalog MSS204338, Thermo Fisher Scientific) or control siRNA (catalog 12935112,
447
Thermo Fisher Scientific) using Lipofectamine RNAiMAX (catalog 13778150, Invitrogen
448
Corporation, Carlsbad, CA, USA) according to the manufacturer’s instructions.
449
Dual Luciferase Assay
450
The promoter sequences for murine Ccl2 genes were obtained from the National Center for
451
Biotechnology Information database. Murine Ccl2 promoter fragments were amplified by PCR of
452
murine
453
5′-TCCGGCCCATGAGAGAACTGCTT-3′
454
5′-ACTATGCCTGGCTCCTGGTA-3′ (S2, -1401/-1381), 5′-GAAGACTCCGCTCAGCCAC-3′ (S3,
455
-1136/-1117) and 5′-TGGCTTCAGTGAGAGTTGGCTGGT-3′ (AS, +67/+44). The Ccl2 promoter
456
nucleotide sequence was confirmed by DNA sequencing and cloned into a pGL3 basic luciferase
457
vector (Promega, Madison, WI, USA) to generate a reporter plasmid. We transfected BNL-CL2 with
genomic
DNA
using
following
sense
(S)
and (S1,
27
antisense
(AS)
primers:
-2468/-2445),
458
either cytomegalovirus promoter (CMV)-T7-tagged FoxM1 plasmid (CMV-T7-FoxM1) or
459
CMV-empty expression plasmid, as well as with luciferase reporters driven by the murine Ccl2
460
promoter lesions. After seeding cells and culturing them overnight, the plasmid was transfected with
461
FuGENE HD transfection reagent (catalog E2311; Promega) according to the manufacturer’s
462
instructions. We used the 6×CDX2-Luc plasmid, a known FoxM1 reporter, as a positive control for
463
CMV-T7-FoxM1 transcriptional activity. The CMV-Renilla luciferase plasmid was used as internal
464
controls to normalize transfection efficiency. A dual luciferase assay (Promega) was performed 48 h
465
after transfection, as described previously.38
466
Chromatin Immunoprecipitation Assay
467
BNL-CL2 cells were prepared using the SimpleChIP Plus Enzymatic Chromatin IP kit
468
(catalog 9005; Cell Signaling Technology). Nuclear extracts from BNL-CL2 cells were cross-linked
469
by the addition of formaldehyde, sonicated, and used for the immunoprecipitation with FoxM1 rabbit
470
polyclonal antibodies (K-19 and C-20) (catalog sc-500, sc-502; Santa Cruz Biotechnology). Reverse
471
cross-linked chromatin immunoprecipitation DNA samples were subjected to real-time PCR using
472
the
473
5′-TCAGGTCCAGGGAAGCATTCTG-3′ (S) and 5′-TGTGTTTATTCCATGGCAAGTGGTC-3′
474
(AS).
475
Synthesis of the triantennary N-acetylgalactosamine (GalNAc)-siRNA
476
oligonucleotides
specific
to
promoter
regions
of
the
murine
Ccl2
gene:
GalNAc-siRNA conjugates were synthesized by Nihon Gene Research Laboratories Inc., 28
477
as described previously.22 The target sequences for GalNAc-siCcl2 and GalNAc-siControl (control)
478
were 5'-TGAATGAGTAGCAGCAGGTGAGTGG-3' and 5'-GGTCTGATCACTGCTCGGT-3'.40
479
The designed triantennary GalNAc-siCcl2 and control are shown in Table 1. TG mice were injected
480
subcutaneously with 3mg/kg of GalNAc-siCcl2 or control 24 hours before DOX treatment, as
481
described previously.41
482
Depletion of Macrophages
483
Clodronate liposomes were prepared as previously described.42 To deplete macrophages,
484
the mice were injected intravenously with either 50 mg/kg of body weight clodronate liposomes or
485
PBS liposomes from 3 days before the experiment. Macrophage depletion in the livers was verified
486
using F4/80 immunostaining three days after intravenous injection.
487
Induction of Chronic Liver Injury
488
To induce chronic liver injury, C57BL/6 mice were subjected to a high-fat,
489
high-cholesterol (HFHC) diet (catalog D09100308, Research Diet, New Brunswick, NJ, USA) for
490
eight weeks or an intraperitoneal administration of carbon tetrachloride (CCl4; 0.5 ml/kg body wt)
491
two times a week for four weeks. The mice were killed at 48 h after final injection.
492
Inhibition of FoxM1 Activity
493
To inhibit FoxM1 activity, we used thiostrepton (catalog T8902, Sigma-Aldrich), a FoxM1
494
inhibitor. 25, 26 In a chronic liver injury model, the mice were injected intraperitoneally with either 30
495
mg/kg bw thiostrepton dissolved in 10% dimethyl sulfoxide (DMSO/PBS) or vehicle as a control for 29
43
496
a total of 7 injections over 2 weeks, as described previously.
TG mice were injected
497
intraperitoneally with thiostrepton or vehicle for two times from 3 days before DOX treatment.
498
Clinical Samples
499
Tumor and non-tumor liver tissues were obtained from patients who underwent surgical
500
liver resection from primary liver tumors. This study was performed according to the ethical
501
guidelines of the Declaration of Helsinki. Approval to use resected samples was obtained from the
502
Institutional Review Board Committee at Osaka University Hospital (No. 13556), and written
503
informed consent was obtained from all patients.
504
Statistical Analysis
505
Statistical analysis was performed using JMP Pro 13.0 software (SAS Institute, Cary, NC,
506
USA). Individual values (symbols) and means (bar) ± standard deviations (SD; error bars) from at
507
least 3 independent experiments are shown. The Student's t test was used to analyze the significant
508
difference between the two groups and one-way analysis of variance (one-way ANOVA) test was
509
performed to analyze the difference among groups. P values less than 0.05 were considered to be
510
statistically significant.
511
30
512
REFERENCES
513
1.
El-Serag HB. Hepatocellular carcinoma. N Engl J Med 2011;365(12):1118-27.
514
2.
El-Serag HB. Epidemiology of viral hepatitis and hepatocellular carcinoma. Gastroenterology 2012;142(6):1264-73 e1.
515
516
3.
research 2011;21(1):159-68.
517
518
4.
Taniguchi K, Karin M. NF-kappaB, inflammation, immunity and cancer: coming of age. Nature reviews Immunology 2018;18(5):309-24.
519
520
He G, Karin M. NF-kappaB and STAT3 - key players in liver inflammation and cancer. Cell
5.
Costa RH, Kalinichenko VV, Holterman AX, Wang X. Transcription factors in liver
521
development, differentiation, and regeneration. Hepatology (Baltimore, Md)
522
2003;38(6):1331-47.
523
6.
Costa RH. FoxM1 dances with mitosis. Nature cell biology 2005;7(2):108-10.
524
7.
Kalin TV, Ustiyan V, Kalinichenko VV. Multiple faces of FoxM1 transcription factor: lessons from transgenic mouse models. Cell cycle (Georgetown, Tex) 2011;10(3):396-405.
525
526
8.
Pilarsky C, Wenzig M, Specht T, Saeger HD, Grutzmann R. Identification and validation of
527
commonly overexpressed genes in solid tumors by comparison of microarray data. Neoplasia
528
2004;6(6):744-50.
529
530
9.
Kim IM, Ackerson T, Ramakrishna S, Tretiakova M, Wang IC, Kalin TV, Major ML, Gusarova GA, Yoder HM, Costa RH, Kalinichenko VV. The Forkhead Box m1 transcription 31
531
factor stimulates the proliferation of tumor cells during development of lung cancer. Cancer
532
research 2006;66(4):2153-61.
533
10.
Kalin TV, Wang IC, Ackerson TJ, Major ML, Detrisac CJ, Kalinichenko VV, Lyubimov A,
534
Costa RH. Increased levels of the FoxM1 transcription factor accelerate development and
535
progression of prostate carcinomas in both TRAMP and LADY transgenic mice. Cancer
536
research 2006;66(3):1712-20.
537
11.
Yoshida Y, Wang IC, Yoder HM, Davidson NO, Costa RH. The forkhead box M1
538
transcription factor contributes to the development and growth of mouse colorectal cancer.
539
Gastroenterology 2007;132(4):1420-31.
540
12.
Wang Z, Park HJ, Carr JR, Chen YJ, Zheng Y, Li J, Tyner AL, Costa RH, Bagchi S,
541
Raychaudhuri P. FoxM1 in tumorigenicity of the neuroblastoma cells and renewal of the
542
neural progenitors. Cancer research 2011;71(12):4292-302.
543
13.
Kalinichenko VV, Major ML, Wang X, Petrovic V, Kuechle J, Yoder HM, Dennewitz MB,
544
Shin B, Datta A, Raychaudhuri P, Costa RH. Foxm1b transcription factor is essential for
545
development of hepatocellular carcinomas and is negatively regulated by the p19ARF tumor
546
suppressor. Genes Dev 2004;18(7):830-50.
547
14.
Gusarova GA, Wang IC, Major ML, Kalinichenko VV, Ackerson T, Petrovic V, Costa RH. A
548
cell-penetrating ARF peptide inhibitor of FoxM1 in mouse hepatocellular carcinoma
549
treatment. The Journal of clinical investigation 2007;117(1):99-111. 32
550
15.
Egawa M, Yoshida Y, Ogura S, Kurahashi T, Kizu T, Furuta K, Kamada Y, Chatani N,
551
Hamano M, Kiso S, Hikita H, Tatsumi T, Eguchi H, Nagano H, Doki Y, Mori M, Takehara T.
552
Increased expression of Forkhead box M1 transcription factor is associated with
553
clinicopathological features and confers a poor prognosis in human hepatocellular carcinoma.
554
Hepatology research : the official journal of the Japan Society of Hepatology
555
2017;47(11):1196-205.
556
16.
Ogura S, Yoshida Y, Kurahashi T, Egawa M, Furuta K, Kiso S, Kamada Y, Hikita H, Eguchi
557
H, Ogita H, Doki Y, Mori M, Tatsumi T, Takehara T. Targeting the mevalonate pathway is a
558
novel therapeutic approach to inhibit oncogenic FoxM1 transcription factor in human
559
hepatocellular carcinoma. Oncotarget 2018;9(30):21022-35.
560
17.
Xia L, Huang W, Tian D, Zhu H, Zhang Y, Hu H, Fan D, Nie Y, Wu K. Upregulated FoxM1
561
expression induced by hepatitis B virus X protein promotes tumor metastasis and indicates
562
poor prognosis in hepatitis B virus-related hepatocellular carcinoma. Journal of hepatology
563
2012;57(3):600-12.
564
18.
Ye H, Kelly TF, Samadani U, Lim L, Rubio S, Overdier DG, Roebuck KA, Costa RH.
565
Hepatocyte nuclear factor 3/fork head homolog 11 is expressed in proliferating epithelial and
566
mesenchymal cells of embryonic and adult tissues. Mol Cell Biol 1997;17(3):1626-41.
567
568
19.
Wang X, Kiyokawa H, Dennewitz MB, Costa RH. The Forkhead Box m1b transcription factor is essential for hepatocyte DNA replication and mitosis during mouse liver 33
569
regeneration. Proceedings of the National Academy of Sciences of the United States of
570
America 2002;99(26):16881-6.
571
20.
Wang X, Hung NJ, Costa RH. Earlier expression of the transcription factor HFH-11B
572
diminishes induction of p21(CIP1/WAF1) levels and accelerates mouse hepatocyte entry into
573
S-phase following carbon tetrachloride liver injury. Hepatology (Baltimore, Md)
574
2001;33(6):1404-14.
575
21.
Ren X, Shah TA, Ustiyan V, Zhang Y, Shinn J, Chen G, Whitsett JA, Kalin TV, Kalinichenko
576
VV. FOXM1 promotes allergen-induced goblet cell metaplasia and pulmonary inflammation.
577
Mol Cell Biol 2013;33(2):371-86.
578
22.
Nair JK, Willoughby JL, Chan A, Charisse K, Alam MR, Wang Q, Hoekstra M, Kandasamy P,
579
Kel'in AV, Milstein S, Taneja N, O'Shea J, Shaikh S, Zhang L, van der Sluis RJ, Jung ME,
580
Akinc A, Hutabarat R, Kuchimanchi S, Fitzgerald K, Zimmermann T, van Berkel TJ, Maier
581
MA, Rajeev KG, Manoharan M. Multivalent N-acetylgalactosamine-conjugated siRNA
582
localizes in hepatocytes and elicits robust RNAi-mediated gene silencing. Journal of the
583
American Chemical Society 2014;136(49):16958-61.
584
23.
Capurro M, Wanless IR, Sherman M, Deboer G, Shi W, Miyoshi E, Filmus J. Glypican-3: a
585
novel serum and histochemical marker for hepatocellular carcinoma. Gastroenterology
586
2003;125(1):89-97.
587
24.
Timek DT, Shi J, Liu H, Lin F. Arginase-1, HepPar-1, and Glypican-3 are the most effective 34
588
panel of markers in distinguishing hepatocellular carcinoma from metastatic tumor on
589
fine-needle aspiration specimens. American journal of clinical pathology
590
2012;138(2):203-10.
591
25.
human cancer cells. PloS one 2009;4(5):e5592.
592
593
26.
27.
28.
29.
Raychaudhuri P, Park HJ. FoxM1: a master regulator of tumor metastasis. Cancer research 2011;71(13):4329-33.
600
601
Luedde T, Schwabe RF. NF-kappaB in the liver--linking injury, fibrosis and hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2011;8(2):108-18.
598
599
Elsharkawy AM, Mann DA. Nuclear factor-kappaB and the hepatic inflammation-fibrosis-cancer axis. Hepatology (Baltimore, Md) 2007;46(2):590-7.
596
597
Hegde NS, Sanders DA, Rodriguez R, Balasubramanian S. The transcription factor FOXM1 is a cellular target of the natural product thiostrepton. Nature chemistry 2011;3(9):725-31.
594
595
Bhat UG, Halasi M, Gartel AL. Thiazole antibiotics target FoxM1 and induce apoptosis in
30.
Penke LR, Speth JM, Dommeti VL, White ES, Bergin IL, Peters-Golden M. FOXM1 is a
602
critical driver of lung fibroblast activation and fibrogenesis. The Journal of clinical
603
investigation 2018;128(6):2389-405.
604
31.
Feng Y, Wang L, Zeng J, Shen L, Liang X, Yu H, Liu S, Liu Z, Sun Y, Li W, Chen C, Jia J.
605
FoxM1 is overexpressed in Helicobacter pylori-induced gastric carcinogenesis and is
606
negatively regulated by miR-370. Molecular cancer research : MCR 2013;11(8):834-44. 35
607
32.
2014;147(3):577-94.e1.
608
609
Marra F, Tacke F. Roles for chemokines in liver disease. Gastroenterology
33.
Li X, Yao W, Yuan Y, Chen P, Li B, Li J, Chu R, Song H, Xie D, Jiang X, Wang H. Targeting
610
of tumour-infiltrating macrophages via CCL2/CCR2 signalling as a therapeutic strategy
611
against hepatocellular carcinoma. Gut 2017;66(1):157-67.
612
34.
Teng KY, Han J, Zhang X, Hsu SH, He S, Wani NA, Barajas JM, Snyder LA, Frankel WL,
613
Caligiuri MA, Jacob ST, Yu J, Ghoshal K. Blocking the CCL2-CCR2 Axis Using
614
CCL2-Neutralizing Antibody Is an Effective Therapy for Hepatocellular Cancer in a Mouse
615
Model. Molecular cancer therapeutics 2017;16(2):312-22.
616
35.
Ren X, Zhang Y, Snyder J, Cross ER, Shah TA, Kalin TV, Kalinichenko VV. Forkhead box
617
M1 transcription factor is required for macrophage recruitment during liver repair. Mol Cell
618
Biol 2010;30(22):5381-93.
619
36.
Sun L, Ren X, Wang IC, Pradhan A, Zhang Y, Flood HM, Han B, Whitsett JA, Kalin TV,
620
Kalinichenko VV. The FOXM1 inhibitor RCM-1 suppresses goblet cell metaplasia and
621
prevents IL-13 and STAT6 signaling in allergen-exposed mice. Science signaling
622
2017;10(475).
623
37.
Wang IC, Zhang Y, Snyder J, Sutherland MJ, Burhans MS, Shannon JM, Park HJ, Whitsett
624
JA, Kalinichenko VV. Increased expression of FoxM1 transcription factor in respiratory
625
epithelium inhibits lung sacculation and causes Clara cell hyperplasia. Dev Biol 36
2010;347(2):301-14.
626
627
38.
Balli D, Ustiyan V, Zhang Y, Wang IC, Masino AJ, Ren X, Whitsett JA, Kalinichenko VV,
628
Kalin TV. Foxm1 transcription factor is required for lung fibrosis and
629
epithelial-to-mesenchymal transition. EMBO J 2013;32(2):231-44.
630
39.
Furuta K, Yoshida Y, Ogura S, Kurahashi T, Kizu T, Maeda S, Egawa M, Chatani N, Nishida
631
K, Nakaoka Y, Kiso S, Kamada Y, Takehara T. Gab1 adaptor protein acts as a gatekeeper to
632
balance hepatocyte death and proliferation during acetaminophen-induced liver injury in mice.
633
Hepatology (Baltimore, Md) 2016;63(4):1340-55.
634
40.
Taniuchi K, Cerny RL, Tanouchi A, Kohno K, Kotani N, Honke K, Saibara T, Hollingsworth
635
MA. Overexpression of GalNAc-transferase GalNAc-T3 promotes pancreatic cancer cell
636
growth. Oncogene 2011;30(49):4843-54.
637
41.
Willoughby JLS, Chan A, Sehgal A, Butler JS, Nair JK, Racie T, Shulga-Morskaya S,
638
Nguyen T, Qian K, Yucius K, Charisse K, van Berkel TJC, Manoharan M, Rajeev KG, Maier
639
MA, Jadhav V, Zimmermann TS. Evaluation of GalNAc-siRNA Conjugate Activity in
640
Pre-clinical Animal Models with Reduced Asialoglycoprotein Receptor Expression.
641
Molecular therapy : the journal of the American Society of Gene Therapy 2018;26(1):105-14.
642
42.
Miura K, Yang L, van Rooijen N, Ohnishi H, Seki E. Hepatic recruitment of macrophages
643
promotes nonalcoholic steatohepatitis through CCR2. American journal of physiology
644
Gastrointestinal and liver physiology 2012;302(11):G1310-21. 37
645
43.
Weiler SME, Pinna F, Wolf T, Lutz T, Geldiyev A, Sticht C, Knaub M, Thomann S, Bissinger
646
M, Wan S, Rossler S, Becker D, Gretz N, Lang H, Bergmann F, Ustiyan V, Kalin TV, Singer
647
S, Lee JS, Marquardt JU, Schirmacher P, Kalinichenko VV, Breuhahn K. Induction of
648
Chromosome Instability by Activation of Yes-Associated Protein and Forkhead Box M1 in
649
Liver Cancer. Gastroenterology 2017;152(8):2037-51.e22.
650
651
38
652
Table 1. Designs of the GalNAc-siRNA conjugates.
653
Compound
Strand
Sequence (5' to 3')
GalNAc-siCcl2
S
UGAAUGAGUAGCAGCAGGUGAGUGG-(GalNAc)
AS
CCACUCACCUGCUGCUACUCAUUCATT
S
GGUCUGAUCACUGCUCGGU-(GalNAc)
AS
ACCGAGCAGUGAUCAGACCTT
GalNAc-siControl
654 655
S, sense strand; AS, antisense strand; GalNAc, triantennary N-acetylagalactosamine.
656
657
39
658
FIGURE LEGENDS
659
Figure 1. Spontaneous liver injury and fibrosis in hepatocyte-specific FoxM1 transgenic mice.
660
(A) Schematic illustration of strategy for generating hepatocyte-specific FoxM1 transgenic (TG)
661
mice. WT, wild-type. (B) Western blot analysis showing FoxM1 protein expression in the liver,
662
kidney, and heart of WT and TG mice after 3 days of doxycycline (DOX) treatment at 8 weeks.
663
FoxM1 protein expression was liver- and DOX-specific in the TG mice. (C) Representative images
664
of FoxM1 staining in liver sections of WT and TG mice after 3 days of DOX treatment at 8 weeks.
665
Scale bar: 100 µm (original magnification, ×200). (D) Quantification of FoxM1-positive area in liver
666
sections of WT and TG mice after 3 days of DOX treatment at 8 weeks (WT, n=8; TG, n=8). (E)
667
Serum ALT levels in WT and TG mice after 13 weeks of DOX treatment since birth (WT, n = 8; TG,
668
n = 8). (F) Representative images of H&E (top) and TUNEL (bottom) staining in liver sections of
669
WT and TG mice after 13 weeks of DOX treatment since birth. Scale bar: 200 µm (original
670
magnification, ×100). (G) Quantification of the number of TUNEL-positive hepatocytes in liver
671
sections of WT and TG mice after 13 weeks of DOX treatment since birth (WT, n = 8; TG, n = 8).
672
(H) Representative views of picrosirius red (top) and α-smooth muscle actin (bottom) staining in
673
liver sections of WT and TG mice after 13 weeks of DOX treatment since birth. Scale bar: 200 µm
674
(original magnification, ×100). (I) Quantification of picrosirius red positive area in the livers of WT
675
and TG mice after 13 weeks of DOX treatment since birth (WT, n = 8; TG, n = 8). (J) Gene
676
expression of ColIα1 and ColIα2 in the livers of WT and TG mice after 13 weeks of DOX treatment 40
677
since birth (WT, n = 8; TG, n = 8). Data are expressed as individual values and mean ± SD, **p <
678
0.01. Significance was calculated using an unpaired Student t test.
679
Figure 2. Reversible liver inflammation with macrophage recruitment induced by short-term
680
hepatocyte-specific overexpression of FoxM1. (A) Schematic illustration of short-term hepatic
681
overexpression of FoxM1. Hepatic overexpression of FoxM1 was induced by three days of
682
doxycycline (DOX) treatment (DOX on) and repressed by three subsequent days of DOX removal
683
(DOX off). Pretreatment is denoted as DOX (-). (B) Western blot analysis showing FoxM1 protein
684
expression in the livers of wild-type (WT) and transgenic (TG) mice at indicated time points. (C)
685
Serum ALT levels of WT and TG mice at indicated time points (WT, n = 8 at 8 wk; TG, n = 8 at 8
686
wk). (D) Representative images of TUNEL staining in liver sections of WT and TG mice. Top,
687
DOX(-); middle, DOX on; bottom, DOX off. Scale bar: 200 µm (original magnification, ×100). (E)
688
Quantification of the number of TUNEL-positive hepatocytes in liver sections of WT and TG mice at
689
indicated time points (WT, n = 8 at 8 wk; TG, n = 8 at 8 wk). (F) Representative images of H&E
690
(left) and F4/80 (right) staining in liver sections of WT and TG mice. Top, DOX (-); second, DOX
691
on; third, DOX on (enlarged view of boxed region of second panel); bottom, DOX off. Scale bar:
692
100 µm (original magnification, ×200). (G) Quantification of the number of CD11b+F4/80+
693
macrophages in the livers of WT and TG mice at indicated time points (WT, n = 3 at 8 wk; TG, n = 3
694
at 8 wk). (H) Quantification of hepatic gene expression of F4/80, Cd68, Nos2, Tnfα, Ccl2, Ccr2,
695
Ccl5, Arg1, and Cd206 in WT and TG mice after 3 days of DOX treatment (WT, n = 8 at 8 wk; TG, n 41
696
= 8 at 8 wk). (I) Quantification of serum CCL2 levels in WT and TG mice at indicated time points
697
(WT, n = 8 at 8 wk; TG, n = 8 at 8 wk). (J) Western blot analysis showing FoxM1 protein expression
698
in the livers of WT and TG mice after 1 day of DOX treatment. (K) Serum ALT levels of WT and TG
699
mice after 1 day of DOX treatment (WT, n = 8 at 8 wk; TG, n = 8 at 8 wk). (L) Gene expression of
700
Ccl2 (left) and serum CCL2 (right) of WT and TG mice after 1 day of DOX treatment (WT, n = 8 at
701
8 wk; TG, n = 8 at 8 wk). (M) Representative images of FoxM1 (top) and TUNEL (bottom) staining
702
in liver sections of TG mice. Left, DOX (-); middle, DOX low dose; right, DOX on. (N)
703
Quantification of FoxM1-positive area in liver sections of TG mice. DOX (-), n=8; DOX low dose,
704
n=6; DOX on, n=8 at 8 wk. (O) Quantification of the number of TUNEL-positive hepatocytes in
705
liver sections of TG mice. DOX (-), n=8; DOX low dose, n=6; DOX on, n=8 at 8 wk. (P) Serum ALT
706
levels of TG mice. DOX (-), n=8; DOX low dose, n=6; DOX on, n=8 at 8 wk. (Q) Ccl2 gene
707
expression in livers of TG mice. DOX (-), n=8; DOX low dose, n=6; DOX on, n=8 at 8 wk. Data are
708
expressed as individual values and mean ± SD, **p < 0.01. Significance was calculated using
709
one-way ANOVA test (C, E, G, I, N, O, P, Q), or Student t test (H, K, L).
710
Figure 3. Direct regulation of CCL2 by FoxM1 in hepatocytes. (A) Quantification of gene
711
expression of FoxM1 (left) and Ccl2 (right) in FoxM1 siRNA-transfected BNL-CL2 cells (n = 3 per
712
group). (B) Quantification of CCL2 levels in supernatant of FoxM1 siRNA-transfected BNL-CL2
713
cells at indicated time points after transfection (n = 3 per group). (C) Quantification of gene
714
expression of FoxM1 (left) and Ccl2 (right) in FoxM1 siRNA-transfected AML12 cells (n = 3 per 42
715
group). (D) Quantification of CCL2 levels in supernatant of FoxM1 siRNA-transfected AML12 cells
716
at indicated time points after transfection (n = 3 per group).
717
(Luc) reporter constructs containing −2468/+67 bp murine Ccl2 promoter and its deletion mutants
718
(−1401/+67 bp and −1136/+67 bp) and quantification of transcriptional activities induced by
719
co-transfection of T7-FoxM1 expression vector (T7-FoxM1) compared with CMV-empty vector
720
(Mock) (n=3 per group). A 6xCDX2 promoter LUC construct was used as a positive control. (F)
721
Quantification of chromatin immunoprecipitation assay to show direct binding of FoxM1 protein to
722
murine Ccl2 promoter DNA using two independent antibodies against FoxM1 (K19 and C20)
723
compared with IgG control (n = 3 per group). Data are expressed as individual values and mean ±
724
SD, **p < 0.01. Significance was calculated using an unpaired Student t test.
725
Figure 4. Hepatocytes are the major source of CCL2 production in TG mice. (A) Quantification
726
of Ccl2 gene expression in hepatocytes (left) and nonparenchymal cells (NPCs; right) from WT and
727
TG mice after 3 days of DOX treatment (n=3, each group). (B) Western blot analysis of FoxM1
728
protein expression in hepatocytes and NPCs of WT and TG mice.
729
Figure 5. FoxM1 induction has a modest effect on cell death in cultured hepatocytes in vitro.
730
(A) Western blot analysis showing FoxM1 protein expression in isolated primary hepatocytes of TG
731
mice treated with 100 ng/ml doxycycline (DOX). (B and C) Quantification of gene expression of
732
Ccl2 in primary hepatocytes from WT (B) and TG (C) mice treated with 100 ng/ml DOX (n = 4 per
43
(E) Schematic illustration of luciferase
733
group). (D and E) Quantification of CCL2 levels in supernatant of primary hepatocytes from WT (D)
734
and TG (E) mice treated with 100 ng/ml DOX (n = 4 per group). (F and G) WST assay of primary
735
hepatocytes from WT (F) and TG (G) mice treated with 100 ng/ml DOX (n = 4 per group). (H and I)
736
Caspase 3/7 activity in primary hepatocytes from WT (H) and TG (I) mice treated with 100 ng/ml
737
DOX (n = 4 per group). (J and K) Quantification of gene expression of Ccna2 in primary
738
hepatocytes from WT (J) and TG (K) mice treated with 100 ng/ml DOX (n = 4 per group). (L and M)
739
Quantification of gene expression of Ccnb1 in primary hepatocytes from WT (L) and TG (M) mice
740
treated with 100 ng/ml DOX (n = 4 per group). (N and O) Quantification of gene expression of Skp2
741
in primary hepatocytes of WT (N) and TG (O) mice treated with 100 ng/ml DOX (n = 4 per group).
742
Data are expressed as individual values and mean ± SD, * p < 0.05, **p < 0.01. Significance was
743
calculated using an unpaired Student t test.
744
Figure 6. Hepatocyte-specific CCL2 inhibition reduces liver injury in TG mice. (A)
745
Quantification of hepatic gene expression of Ccl2 (left) and serum CCL2 levels (right) in TG mice
746
treated with GalNAc-siControl (control) or GalNAc-siCcl2 (control, n = 4 at 8 wk; GalNAc-siCcl2,
747
n = 4 at 8 wk). (B) Representative images of H&E staining of TG mice treated with control or
748
GalNAc-siCcl2. Scale bar: 200 µm (original magnification, ×100). (C) Serum ALT levels of TG
749
mice treated with control or GalNAc-siCcl2 (control, n = 4 at 8 wk; GalNAc-siCcl2, n = 4 at 8 wk).
750
(D) Representative images of TUNEL staining of TG mice treated with control or GalNAc-siCcl2.
751
Scale bar: 200 µm (original magnification, ×100). (E) Quantification of the number of 44
752
TUNEL-positive hepatocytes in liver sections from control and GalNAc-siCcl2 group mice (control,
753
n = 4 at 8 wk; GalNAc-siCcl2, n = 4 at 8 wk). (F) Representative images of F4/80 staining of TG
754
mice treated with control or GalNAc-siCcl2 (bottom, enlarged views of the boxed regions in the top
755
image). Scale bar: 100 µm (original magnification, ×200). (G) Representative images of picrosirius
756
red staining in liver sections from TG mice treated with control or GalNAc-siCcl2. Scale bar: 200
757
µm (original magnification, ×100). (H) Quantification of picrosirius red positive area (%) in the
758
livers of control and GalNAc-siCcl2 group mice (control, n = 4 at 8 wk; GalNAc-siCcl2, n = 4 at 8
759
wk). (I) Gene expression of ColIα1 (left) and ColIα2 (right) in the livers of control and
760
GalNAc-siCcl2 group mice (control, n = 4 at 8 wk; GalNAc-siCcl2, n = 4 at 8 wk). Data are
761
expressed as individual values and mean ± SD, **p < 0.01. Significance was calculated using an
762
unpaired Student t test.
763
Figure 7. Macrophage recruitment is required for spontaneous liver inflammation in
764
transgenic mice. (A) Representative images of F4/80 staining in liver sections of 3-day DOX-treated
765
wild-type (WT) and transgenic (TG) mice injected with PBS or clodronate liposomes. Top, WT
766
mice; middle, TG mice; bottom, enlarged views of boxed region of middle panels. Scale bar: 200 µm
767
(original magnification, ×100). (B) Quantification of hepatic gene expression of F4/80 (left) and
768
Cd68 (right) in 3-day DOX-treated WT and TG mice injected with PBS or clodronate liposomes.
769
(PBS-treated WT mice, n = 4 at 8 wk; PBS-treated TG mice, n = 4 at 8 wk; clodronate
770
liposome–treated WT mice, n = 4 at 8 wk; clodronate liposome–treated WT mice, n = 4 at 8 wk). (C) 45
771
Quantification of serum ALT levels in 3 days DOX-treated WT and TG mice injected with PBS or
772
clodronate liposomes (PBS-treated WT mice, n = 4 at 8 wk; PBS-treated TG mice, n = 4 at 8 wk;
773
clodronate liposome–treated WT mice, n = 4 at 8 wk; clodronate liposome–treated WT mice, n = 4 at
774
8 wk). (D) Representative images of TUNEL staining in liver sections of 3-day DOX-treated WT and
775
TG mice injected with PBS or clodronate liposomes. Scale bar: 200 µm (original magnification,
776
×100). (E) Quantification of the number of TUNEL-positive hepatocytes in liver sections of 3-day
777
DOX-treated WT and TG mice injected with PBS or clodronate liposomes (PBS-treated WT mice, n
778
= 4 at 8 wk; PBS-treated TG mice, n = 4 at 8 wk; clodronate liposome–treated WT mice, n = 4 at 8
779
wk; clodronate liposome–treated WT mice, n = 4 at 8 wk). Data are expressed as individual values
780
and mean ± SD, ** p < 0.01. Significance was calculated using one-way ANOVA test.
781
Figure 8. Long-term hepatocyte-specific overexpression of FoxM1 causes hepatocarcinogenesis
782
with significant fibrosis in the background liver. (A) Gross appearance of livers in wild-type (WT)
783
and transgenic (TG) mice after 12 months of doxycycline (DOX) treatment since birth. Arrowheads
784
indicate liver tumors in TG mice. (B) Representative images of H&E (top), Arginase-1 (middle) and
785
Glypican-3 (bottom) staining of WT and TG mice after 12 months of DOX treatment since birth.
786
Non-tumor (NT) lesions in WT mice (left), non-tumor lesions in TG mice, denoted as TG(NT)
787
(middle), and tumor lesions in TG mice, which are denoted as TG(T) (right). Scale bar: 100 µm
788
(original magnification, ×400). (C) Quantification of tumor numbers (left), maximal size (mm,
789
middle), tumor incidence (%, right) in WT and TG mice after 12 months of DOX treatment since 46
790
birth (WT mice, n = 10; TG mice, n = 10). (D) Representative images of Ki-67 staining of WT and
791
TG mice after 12 months of DOX treatment since birth. Scale bar: 200 µm (original magnification,
792
×100). (E) Quantification of the number of Ki-67 positive hepatocytes in liver sections of NT
793
lesions in WT mice, NT lesions in TG mice, denoted as TG(NT), and tumor lesions in TG mice,
794
which are denoted as TG(T). (WT mice, n = 10; TG mice, n = 10). (F) Representative images of
795
picrosirius red staining of WT and TG mice after 12 months of DOX treatment since birth. NT,
796
non-tumor, T, tumor. Scale bar: 200 µm (original magnification, ×100). (G) Quantification of
797
picrosirius red positive area (%) in liver sections of WT and TG mice (WT mice, n = 10; TG mice, n
798
= 10). (H) Quantification of ColIα1 and ColIα2 gene expression in the livers of WT and TG mice
799
(WT mice, n = 10; TG mice, n = 10). Data are expressed as individual values and mean ± SD, NT,
800
non-tumor; T, tumor. * p < 0.05, ** p < 0.01. Significance was calculated using one-way ANOVA
801
test (E), or Student t test (C, G, H).
802
Figure 9. Increased expression of FoxM1 in liver sections of patients with chronic liver diseases
803
and murine model of liver injury. (A) Representative images of FoxM1 staining in liver sections of
804
patients with chronic liver diseases. Scale bar: 100 µm (original magnification, ×400).
805
Quantification of the FoxM1-positive area was indicated on the lower left. NL, normal liver; CHB,
806
chronic hepatitis B; CHC, chronic hepatitis C; NASH, nonalcoholic steatohepatitis; LC, liver
807
cirrhosis; HCC, hepatocellular carcinoma. (B) Quantification of hepatic FoxM1 expression (left) and
808
serum ALT levels (right) in C57BL/6 mice fed a high-fat and high-cholesterol (HFHC) diet for 8 47
809
weeks or treated with carbon tetrachloride (CCl4) for 4 weeks. Mice fed a normal diet (ND) were
810
used as the control (ND, n = 6 at 14 wk; HFHC, n = 6 at 14 wk; CCl4, n = 4 at 14 wk). (C)
811
Representative images of FoxM1 staining (left) and quantification of FoxM1-positive area (right) in
812
liver sections of C57BL/6 mice fed a ND or a HFHC diet or treated with CCl4. ND-fed mice were
813
used as the control. Scale bar: 100 µm (original magnification, ×200). Quantification positive rate
814
was indicated on the lower left. Data are expressed as individual values and mean ± SD, **p < 0.01.
815
Significance was calculated using an unpaired Student t test.
816
Figure 10. FoxM1 inhibitor reduces liver inflammation in mice fed a high-fat, high-cholesterol
817
diet. (A) Serum ALT levels of C57BL/6 mice fed a high-fat, high-cholesterol diet for 8 weeks and
818
treated with vehicle (control) or thiostrepton for 2 weeks (control, n = 8 at 14 wk; thiostrepton, n = 8
819
at 14 wk). (B) Representative images of H&E staining of control and thiostrepton groups. Scale bar:
820
200 µm (original magnification, ×100). (C) Representative images of TUNEL staining of control
821
and thiostrepton groups. Scale bar: 200 µm (original magnification, ×100). (D) Quantification of the
822
number of TUNEL-positive hepatocytes in liver sections of control and thiostrepton group (control, n
823
= 8; thiostrepton, n = 8). (E) Representative images of F4/80 staining of C57BL/6 mice treated with
824
vehicle or thiostrepton (bottom, enlarged views of top boxed regions). Scale bar: 100 µm (original
825
magnification, ×200). (F) Quantification of F4/80 and Ccl2 gene expression in the livers of control
826
and thiostrepton group (control, n = 8; thiostrepton, n = 8). (G) Serum CCL2 levels of control and
827
thiostrepton group (control, n = 8; thiostrepton, n = 8). (H) Representative images of Ki-67 staining 48
828
in control and thiostrepton group (control, n = 8; thiostrepton, n = 8). Scale bar: 100 µm (original
829
magnification, ×200). (I) Quantification of the number of Ki-67 positive hepatocytes in liver sections
830
of control and thiostrepton group (control, n = 8; thiostrepton, n = 8). Data are expressed as
831
individual values and mean ± SD, ** p < 0.01. Significance was calculated using an unpaired
832
Student t test.
833
Figure 11. FoxM1 inhibitor reduces liver inflammation in transgenic mice. (A) Serum ALT
834
levels of TG mice treated with vehicle (control) or thiostrepton every 3 days from 3 days before the
835
experiment (control, n = 8 at 8 wk; thiostrepton, n = 8 at 8 wk). Hepatic overexpression of FoxM1
836
was induced by 3 days of doxycycline treatment. (B) Representative images of H&E staining of TG
837
mice treated with vehicle or thiostrepton. Scale bar: 100 µm (original magnification, ×200). (C)
838
Representative images of TUNEL staining of TG mice treated with vehicle or thiostrepton. Scale bar:
839
200 µm (original magnification, ×100). (D) Quantification of the number of TUNEL-positive
840
hepatocytes in liver sections of control and thiostrepton group (control, n = 8 at 8 wk; thiostrepton, n
841
= 8 at 8 wk). (E) Representative images of F4/80 staining of TG mice treated with vehicle or
842
thiostrepton (bottom, enlarged views of top boxed regions). Scale bar: 100 µm (original
843
magnification, ×200). (F) Quantification of hepatic gene expression of F4/80 and Ccl2 in TG mice
844
treated with vehicle or thiostrepton (control, n = 8 at 8 wk; thiostrepton, n = 8 at 8 wk). (G) Serum
845
CCL2 levels in TG mice treated with vehicle or thiostrepton (control, n = 8 at 8 wk; thiostrepton, n =
846
8 at 8 wk). (H) Representative images of Ki-67 staining in control and thiostrepton group (control, n 49
847
= 8; thiostrepton, n = 8). Scale bar: 100 µm (original magnification, ×200). (I) Quantification of the
848
number of Ki-67 positive hepatocytes in liver sections of control and thiostrepton group (control, n =
849
8; thiostrepton, n = 8). Data are expressed as individual values and mean ± SD, * p < 0.05, ** p <
850
0.01. Significance was calculated using an unpaired Student t test.
50
TetO7-FoxM1 Rosa26-LSL-rtTA
FoxM1 DOX (-)
TG mice TetO7-FoxM1 Rosa26-LSL-rtTA Alb-Cre
D
TG
DOX (+)
DOX (-)
WT
200
100
WT
40 20 0
WT
TG
G
TG
** 4 WT TG 2
0 WT
TG
WT WT TG
I WT
TG
Picrosirius red positive area (%)
H αSMA Picrosirius red
WT TG
60
** 8
4
0
WT
TG
TG
J Relative expression of mRNA /Gapdh (fold of control)
0
F
80
TUNEL-positive hepatocytes (/field)
WT TG
GAPDH **
H&E
**
GAPDH FoxM1
DOX (+)
TUNEL
Serum ALT (U/L)
E
Liver Kidney Heart WT TG WTTG WTTG
Alb-Cre
WT mice TetO7-FoxM1 Rosa26-LSL-rtTA
C
B
Rosa26-LSL-rtTA
FoxM1-positive area (%)
Figure 1 A TetO7-FoxM1
**
WT TG
40
** 20
0
Col1α1
Col1α2
Figure 2 A DOX (-) 8 weeks♂ WT or TG
DOX on
DOX(+) 3 days
DOX (-)
B
DOX off
DOX on
DOX off
FoxM1 WT
DOX(-) 3 days
GAPDH FoxM1
C
**
**
TG GAPDH
**
Serum ALT (U/L)
600 WT TG
400 200
0 DOX (-) TUNEL WT
TG
DOX off
E
DOX (-) DOX on DOX off
DOX (-)
DOX on
DOX off
**
40
** WT TG
20
0 DOX (-) H&E WT
F4/80 TG
WT
TG
DOX on
DOX off
G ** Macrophages (x106 cells / g liver)
F
** TUNEL-positive hepatocytes (/field)
D
DOX on
WT TG **
**
0.6
0.4
0.2
0
DOX (-)
DOX on DOX off
H
Relative expression of mRNA / Gapdh (fold of control)
Figure 2 **
600 400
WT TG
** 200 **
**
**
**
**
0 F4/80
Nos2
Tnfα
Ccl2
Ccr2
CcI5
Arg1
Cd206
**
** Serum CCL2 (pg/mL)
I
Cd68 **
600
WT TG
400 200
0
DOX (-)
DOX on
J
DOX off
DOX 1day WT
TG
FoxM1 GAPDH
30 20 10 0
L
DOX 1day
10
**
8 6 4 2 0
DOX 1day
200 Serum CCL2 (pg/mL)
WT TG
Ccl2 mRNA (fold of control)
Serum ALT (U/L)
K
WT TG
150 100 50 0
DOX 1day
Figure 2
DOX (-)
DOX low dose
DOX on
M FoxM1
80
**
**
60 40 20 0 DOX DOX DOX (-) low dose on
Serum ALT (U/L)
P 600
**
**
400 200
0
DOX DOX DOX (-) low dose on
Q
Ccl2 mRNA (fold of control)
O FoxM1-positive area (%)
N
TUNEL-positive hepatocytes (/field)
TUNEL
40
**
**
30 20 10 0 DOX DOX DOX (-) low dose on **
**
400
200
0 DOX DOX DOX (-) low dose on
BNL-CL2 **
BNL-CL2 **
1
** 6000 4000 2000 0 0 24 48 72 Time post transfection (h)
0 AML12 **
D
AML-12 **
AML-12
1
CCL2 (pg/mL)
Ccl2 mRNA (fold of control)
Foxm1 mRNA (fold of control)
** 1
Control siFoxm1
**
Ccl2 mRNA (fold of control)
Foxm1 mRNA (fold of control)
1
0
C
BNL-CL2
B CCL2 (pg/mL)
Figure 3 A
30000
Control siFoxm1
**
20000 10000 0
0
0
0 24 48 72 Time post transfection (h) FoxM1 (K19) FoxM1 (C20) IgG
F Mock T7-FoxM1
FoxM1 binding site
+67
-1401
**
Luc +67 Luc -1136
+67 **
6xCDX2 promoter 0
2
4
6
8
Luciferase fold induction
Relative ratio to IgG Control
Luc -2468
** **
**
Mouse Ccl2 promoter construct
E
2
1
0 Ccl2 Control promoter primer
Ccl2 mRNA (fold of control)
Figure 4 12 A
B
** WT TG
8
Hepatocytes WT FoxM1 GAPDH
4
0
Hepatocytes
NPCs
TG
NPCs WT
TG
Figure 5 A
WT
DOX (-)
DOX (+)
TG DOX DOX (-) (+)
FoxM1 GAPDH
B
C
WT
TG **
Ccl2 mRNA (fold of control)
Ccl2 mRNA (fold of control)
DOX(-) DOX(+)
1.2 0.8 0.4
3 2 1 0
0 WT
D
DOX(-) DOX(+)
TG **
E DOX(-) DOX(+)
4000
2000
CCL2 (pg/mL)
CCL2 (pg/mL)
12000
4000
0
0 WT *
F
G
TG * 1
DOX(-) DOX(+)
WST assay (fold of control)
WST assay (fold of control)
1
0
DOX(-) DOX(+)
8000
DOX(-) DOX(+)
0
Skp2 mRNA (fold of control)
L 0 WT
1
N
6
4
2
0 DOX(-) DOX(+)
0 WT
2
WT
DOX(-) DOX(+) Ccna2 mRNA (fold of control)
Caspase 3/7 activity (fold of control) 1.2
DOX(-) DOX(+)
1 Ccnb1 mRNA (fold of control)
Ccna2 mRNA (fold of control)
J 1.6
DOX(-) DOX(+)
0.8
0.4 Caspase 3/7 activity (fold of control)
WT
Skp2 mRNA (fold of control)
Ccnb1 mRNA (fold of control)
Figure 5 H I
K
M
0
8 TG **
2
O
12
8
4
0
DOX(-) DOX(+)
1
0 TG
1 DOX(-) DOX(+)
0 TG **
** 12
8 DOX(-) DOX(+)
4
0
TG
*
DOX(-) DOX(+)
** 1.5 1 0.5
0
400
control GalNAc-siCcl2
200
0 GalNAc-siCcl2
C
TUNEL
GalNAc-siCcl2
F4/80
0 ** 16 12 8 4 0
control
GalNAc-siCcl2
control GalNAc-siCcl2
500
E control
**
1000
TUNEL-positive hepatocytes (/field)
D
F
1500 Serum ALT (U/L)
control
H&E
B
**
CCL2 (pg/L)
Ccl2 mRNA (fold of control)
Figure 6 A
control GalNAc-siCcl2
Picrosirius red positive area (%)
control
H
control GalNAc-siCcl2
0.6
0.4
0.2
0 2
1
0 Col1α1 Relative expression of mRNA /Gapdh (fold of control)
G
Relative expression of mRNA /Gapdh (fold of control)
Picrosirius red
Figure 6
GalNAc-siCcl2
I *
1.2
0.8
0.4
0 Col1α2
control GalNAc-siCcl2
Figure 7 A
PBS
B
Clodronate
WT TG
WT
TG
**
4
2
20
10
0
0
PBS
PBS Clodronate
**
Serum ALT (U/L)
600
D
** **
WT TG WT 400
200 TG 0 PBS Clodronate
Clodronate
E PBS
Clodronate
TUNEL-positive hepatocytes (/field)
C
**
** Cd68 mRNA (fold of control)
F4/80 mRNA (fold of control)
**
**
**
WT TG
** **
20
10
0
**
PBS Clodronate
Figure 8 A WT
TG
WT
Glypican-3
Arginase-1
H&E
B
TG(NT)
TG(T)
2
0
D Ki-67
TG T
F
WT
TG
T
NT 12
8
4
Tumor incidence (%)
**
0
WT
NT
Picrosirius red
12
10
8
6
4
2
0 100
G
** **
50
**
12
8
4
0
Col1α2 mRNA (fold of control)
4 **
Col1α1 mRNA (fold of control)
6
Maximal size (mm)
**
Ki-67 positive hepatocytes (/field)
8
Picrosirius red positive area (%)
Tumor number
Figure 8
C WT TG
100
0
E ** *
200
150 WT
TG (NT)
TG (T)
0
H
WT TG WT TG
8
6
4
2
0 **
Figure 9 **
B
CHB
0.4%
** Serum ALT (U/L)
NL
FoxM1
Foxm1 mRNA (fold of control)
A
** 400
60
40 20
300 200 100
0
0 ND HFHC CCl4
ND HFHC CCl4
24.0%
**
CHC
C
0.7% 33.6%
31.0% Normal HFHC CCl4
LC
42.0%
HCC
36.9%
42.2%
FoxM1-positive area (%)
** 20.0%
NASH
**
60 40 20 0 ND HFHC CCl4
Figure 10 A
B
**
control
control thiostrepton
80 60
thiostrepton
H&E
Serum ALT (U/L)
100
40 20 0
D thiostrepton
TUNEL
control
TUNEL-positive hepatocytes (/field)
C
E
control
thiostrepton
**
4
control thiostrepton
3 2 1 0
F
control thiostrepton ** 3 Ccl2 mRNA (fold of control)
F4/80
F4/80 mRNA (fold of control)
1.2
0.8
0.4
0
I control
150 100 50 0
Ki-67
Serum CCL2 (pg/mL)
H thiostrepton
1
0 30 Ki-67 positive hepatocytes (/field)
**
G
2
20
10
0
control thiostrepton
Figure 11 **
A
B
1500 1000 500 0
D thiostrepton
TUNEL
control
E
control
thiostrepton
control thiostrepton
** TUNEL-positive hepatocytes (/field)
C
thiostrepton
H&E
Serum ALT (U/L)
control control thiostrepton
2000
60 40
20
0
F
control thiostrepton
1.2 0.8 0.4
* Ccl2 mRNA (fold of control)
F4/80
F4/80 mRNA (fold of control)
*
0
2500
1500 1000 500 0
1 0
control thiostrepton
I control
2000
2
thiostrepton
Ki-67 positive hepatocytes (/field)
**
H
Ki-67
Serum CCL2 (pg/mL)
G
3
2
1
0