Accepted Manuscript Irisin stimulates cell proliferation and invasion by targeting the PI3K/AKT pathway in human hepatocellular carcinoma Guangjun Shi, Nan Tang, Jiantao Qiu, Deguo Zhang, Fei Huang, Yayu Cheng, Kun Ding, Weisheng Li, Ping Zhang, Xueying Tan PII:
S0006-291X(17)31682-0
DOI:
10.1016/j.bbrc.2017.08.148
Reference:
YBBRC 38423
To appear in:
Biochemical and Biophysical Research Communications
Received Date: 6 August 2017 Accepted Date: 25 August 2017
Please cite this article as: G. Shi, N. Tang, J. Qiu, D. Zhang, F. Huang, Y. Cheng, K. Ding, W. Li, P. Zhang, X. Tan, Irisin stimulates cell proliferation and invasion by targeting the PI3K/AKT pathway in human hepatocellular carcinoma, Biochemical and Biophysical Research Communications (2017), doi: 10.1016/j.bbrc.2017.08.148. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. 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.
ACCEPTED MANUSCRIPT
Irisin stimulates cell proliferation and invasion by targeting the PI3K/AKT
2
pathway in human hepatocellular carcinoma
3
Guangjun Shia, Nan Tanga, Jiantao Qiub, Deguo Zhanga, Fei Huanga, Yayu Chengc, Kun Dingc,
4
Weisheng Lic, Ping Zhangc,*, Xueying Tana,*
RI PT
1
5 6
a
7
University, Qingdao, 266000, China
8
b
9
266000, China
SC
Department of Hepatobiliary Surgery, The Affiliated Qingdao Municipal Hospital of Qingdao
M AN U
Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao,
10
c
11
Dao, China
12
* Corresponding authors1: Xueying Tan, Department of Hepatobiliary Surgery, The Affiliated
13
Qingdao Municipal Hospital of Qingdao University, Qing Dao, China; Telephone:
14
86-532-18562692818; E-mail address:
[email protected]
15
and
16
Ping Zhang, Department of Gynecology, The Affiliated Qingdao Municipal Hospital of Qingdao
17
University, Qing Dao, China; Telephone: 86-532-17685506626; E-mail address:
18
[email protected]
AC C
EP
TE D
Department of Gynecology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qing
19 20 1
Abbreviations: FNDC5, fibronectin type III domain-containing protein 5; HCC, hepatocellular carcinoma; IM,
modified irisin; INM: nonmodified irisin; Dox, doxorubicin.
1
ACCEPTED MANUSCRIPT Abstract
22
Irisin is a newly identified myokine that may be cancer-associated, and its impact on liver cancer is
23
unclear. To understand the roles of irisin in liver cancer, we investigated its effect in HepG2 and
24
SMCC7721 hepatocellular carcinoma cells, and the underlying mechanisms. We determined irisin
25
levels in liver tissues and serum samples obtained from patients by using real-time polymerase chain
26
reaction and enzyme-linked immunosorbent assay. Irisin levels in cancerous livers were significantly
27
upregulated compared with those in control livers, but serum irisin levels remained unchanged.
28
Additionally, we evaluated the effects of different concentrations of human recombinant modified and
29
active (glycosylated) irisin (IM) or human recombinant nonmodified irisin (INM) on cell migration,
30
proliferation, viability, and invasiveness. CCK8, transwell, and scratching assays demonstrated that
31
irisin significantly increased cell proliferation, invasion, and migration through activation of the
32
PI3K/AKT pathway. Irisin-induced cell proliferation, migration, and invasion were blocked by a PI3K
33
inhibitor (LY294002). Irisin also decreased the cytotoxicity of doxorubicin in HepG2 cells. These data
34
indicate that increased irisin levels may have protective roles in liver cancer cells through partial
35
activation of the PI3K/AKT pathway, which may facilitate liver cancer progression and decrease the
36
sensitivity to chemotherapy.
38 39
SC
M AN U
TE D
EP
AC C
37
RI PT
21
Keywords: Irisin, proliferation, doxorubicin, phosphorylated AKT
40 41 42 2
ACCEPTED MANUSCRIPT 1. Introduction
44
Irisin (FNDC5) is a recently identified adipokine/myokine [1] that has been widely studied worldwide.
45
After intense exercise, PGC1-α, a transcriptional coactivator of the PPARc nuclear receptor,
46
stimulates the expression of FNDC5, FNDC5 N-terminal domain (1-31 amino-acids (aa)) is
47
proteolytically cleaved, and irisin (32-143 aa) is formed and released into the blood. Irisin was
48
initially identified in skeletal muscles and adipose tissues [2], but its expression has been observed in
49
the brain, pancreas, liver, stomach [3], heart [4], spleen, and skin [5]. Additionally, the expression of
50
irisin was determined to increase in patients with metabolic disorder [6], cervical cancer, endometrial
51
cancer [7],cancer cachexia [8], and prior gestational diabetes mellitus [9], and determined to decrease
52
in type 2 diabetes patients [10, 11] and non-alcoholic fatty liver disease [12]. Irisin serum levels have
53
been shown to be decreased in breast cancer [13], and irisin was shown to have considerable
54
suppressive effects on malignant breast cancer cells [14]. Primary liver cancer was the sixth most
55
frequent cancer and the second leading cause of cancer-related death worldwide in 2015, accounting
56
for 810,500 deaths [15]. However, the detailed molecular mechanisms underlying liver cancer
57
development and progression remain unclear. Although a significant increase in irisin levels has been
58
detected in hepatocellular carcinoma (HCC) [16], no previous in vitro or in vivo experiments have
59
confirmed the potential role of irisin in HCC.
AC C
EP
TE D
M AN U
SC
RI PT
43
60
Here, we aimed to determine the expression levels of irisin in HCC tissues and serum samples,
61
and, for the first time, to characterize the effects of irisin on liver cancer and molecular mechanisms
62
underlying these effects.
63 64
2. Materials and Methods 3
ACCEPTED MANUSCRIPT 2.1. Samples
66
This study has been approved by the Ethics Committee of Qing Dao municipal hospital. Twenty
67
patients with HCC were recruited at the Qing Dao Municipal Hospital in 2016 and 2017.
68
Corresponding non-tumor liver specimens were used as the controls. The study was conducted in
69
collaboration with the Liver Center of the Qingdao Municipal Hospital. Only adult patients were
70
enrolled, and informed consent was obtained from all patients. Exclusion criteria were as follows: (1)
71
acute liver failure; (2) non-HCC liver neoplasms (i.e., cholangiocarcinoma, adenomatosis, hepatic
72
hemangioma); (3) acute and/or chronic kidney disease (defined as serum creatinine 1.5 mg/dL at
73
transplantation); (4) body-mass index (BMI) > 30 kg/m2; (5) autoimmune liver disease on active
74
steroid treatment; (6) hepatitis C virus (HCV)-related disease treated with interferon; (7) a history of
75
major cardiovascular events, such as myocardial infarction, unstable angina, or stroke; (8) diabetes; (9)
76
history of chemotherapy and/or radiotherapy; (10) occurrence of transfer; (11) polycystic ovary
77
syndrome; or (12) mental impairment.
78
2.2. Determination of human serum FNDC5/irisin level
79
The serum FNDC5/irisin level was determined using an enzyme-linked immunosorbent assay (ELISA)
80
kit (USCN Life Science, Wuhan, China) according to the manufacturer’s instructions. The absorbance
81
of the samples was measured at 450 nm (Bio-Tek ELX800, Winooski, USA).
82
2.3. Real-time quantitative polymerase chain reaction (qPCR)
83
Total RNA was extracted from tissues using Trizol (Invitrogen, California, USA). RNA was
84
reverse-transcribed using reverse transcriptase (Takara, Dalian, China). To determine irisin mRNA
85
levels, real-time qPCR was performed using an ABI 7500 Fast Real-Time PCR system (Applied
AC C
EP
TE D
M AN U
SC
RI PT
65
4
ACCEPTED MANUSCRIPT Biosystems, Foster City, CA, USA). The results were normalized to the levels of the internal reference,
87
glyceraldehyde-3-phosphate dehydrogenase (GAPDH).
88
2.4. Cell culture and treatments
89
HepG2 and SMMC7721 cells were cultured in Dulbecco's Modified Eagle Medium (DMEM,
90
Hyclone Laboratories, Utah, USA) supplemented with 10% fetal bovine serum (FBS; Excell Bio,
91
Shanghai, China) and penicillin (100 U/mL) and streptomycin (100 U/mL). The cells were transferred
92
into culture flasks and cultured at 37°C in a humidified atmosphere with 5% CO2. The cells were
93
treated for 24 h with either human recombinant modified (glycosylated) irisin (IM) from PlexBio (San
94
Francisco, CA, USA) or human recombinant nonmodified irisin (INM; Cayman Chemical, Ann Arbor,
95
MI, USA) at concentrations of 0.625 nM, 1.25 nM, 2.5 nM, 5 nM, 10 nM, or 20 nM, dissolved in
96
culture media.
97
2.5. Cell proliferation
98
Cell proliferation was assessed using the Cell Counting Kit 8 (CCK 8; 7-sea, Haimen, China). HepG2
99
and SMCC7721 cells were seeded into 96-well plates at 2000 cells/well in 200 µL of medium, and
100
treated with different concentrations of IM or INM (0.625, 1.25, 2.5, 5, 10, and 20 nM) or without
101
these molecules (N=8) for 24 h in a humidified atmosphere with 5% CO2. Subsequently, the cells
102
were then incubated with 10 µL of CCK8 per well at 37°C for 1 h. An ELISA reader was then used to
103
measure the optical density at 450 nm.
104
AC C
EP
TE D
M AN U
SC
RI PT
86
Furthermore, HepG2 and SMCC7721 cells were treated with IM (2.5 nM) for 0, 12, 24, 48, and
105
72 h (N=8), 10 µL of CCK8 solution was added to each well, and the cultures were incubated at 37°C
106
for 1 h. The optical density of each well was determined at 450 nm.
5
ACCEPTED MANUSCRIPT 2.6. Cell migration and invasion
108
Cell migration was assessed by scratching monolayers of cells cultured in six-well plates. HepG2 cells
109
were treated with or without 2.5 nM IM for 24 h prior to the introduction of the scratch. Subsequently,
110
the cells were washed three times with phosphate-buffered saline (PBS), and the medium was
111
removed from the wells and replaced with serum-free medium (N=5). Migration was observed after
112
24 h and imaged under a light microscope (Olympus, Tokyo, Japan). Cell invasion was evaluated
113
using Transwell insert chambers with an 8-µm pore size (Corning, Maine, USA) coated with Matrigel
114
(BD Biosciences, Bedford, MA), and 5×104 cells were seeded in the serum-free medium in the upper
115
chamber. The lower chamber was filled with DMEM with 10% FBS. After 24 h, the cells that had
116
traversed the membrane were fixed, stained in 0.1% crystal violet solution for 30 min, and counted
117
(N=5).
118
2.7. Western blot
119
Total protein was extracted from HepG2 cells. The protein concentration was determined using the
120
BCA method, as recommended by the manufacturer (Beyotime Biotechnology, China). The collected
121
proteins were boiled for 5 min and separated using SDS-PAGE (10%), after which they were
122
transferred to polyvinylidene difluoride (PVDF) membranes (Millipore, Bedford, MA, USA). The
123
PVDF membranes were blocked with 5% bovine serum albumin (BSA) for 2 h at 25°C and then
124
incubated with primary antibodies against AKT (1:1000) and phospho (p)-AKT (1:2000) (CST, Santa
125
Cruz Biotechnology, CA, USA), at 4°C overnight. After washing with phosphate-buffered saline with
126
Tween 20 (PBST) three times, the membranes were incubated with the secondary rabbit polyclonal
127
antibody (1:2000) (Abgent, Suzhou, China) for 1 h at 25°C. Labeled protein bands were revealed by
128
ECL detection reagent (Millipore, MA, USA). The obtained bands were quantified by determining the
AC C
EP
TE D
M AN U
SC
RI PT
107
6
ACCEPTED MANUSCRIPT band intensity in each group by ImageJ software.
130
2.8. Annexin V/propidium iodide (PI) staining
131
HepG2 cells were treated with medium or Dox (10 µM) with and without IM (2.5 nM) for 24 h. The
132
efficiency of Dox against HepG2 cells was determined by using annexin V/PI staining. HepG2 cells
133
were trypsinized, harvested, and washed with PBS two times, after which they were harvested by
134
centrifugation at 1000 rpm for 3 min. Apoptotic cells were detected using an Annexin V-FITC
135
apoptosis detection kit (Beyotime Biotechnology, Shanghai, China), according to the manufacturer’s
136
instructions. Briefly, 1×105 treated cells were collected and incubated with Annexin V-FITC and PI,
137
and analyzed using flow cytometry (BD FACS Calibur, BD, USA).
138
2.9. Statistical analyses
139
Data are presented as the mean ± standard deviation. Comparisons between the groups were
140
performed using one–way analysis of variance (ANOVA). Student’s t-test was performed to determine
141
the differences between two groups. P<0.05 was considered statistically significant.
142
EP
TE D
M AN U
SC
RI PT
129
3. Results
144
3.1. Irisin expression in HCC tissues
145
Serum irisin levels did not differ between HCC patients and controls (Fig. 1A). Real-time qPCR
146
analysis showed a significant increase in FNDC5 expression (over seven fold) in HCC tissues
147
compared with that in the controls (Fig. 1B).
148
3.2. Effects of irisin on cell proliferation, migration, and invasion
149
The viability of HepG2 and SMCC7721 cells was shown to increase following the application of all
AC C
143
7
ACCEPTED MANUSCRIPT investigated concentrations of IM or INM, compared with that of the control cells (Fig. 2A).
151
Furthermore, the CCK8 assay demonstrated that IM treatment (2.5 nM) led to a significant increase in
152
cell proliferation, compared with that of the control cells (Fig. 2B). Additionally, HepG2 cells treated
153
with irisin (2.5 nM) displayed significantly increased cell migration (Fig. 2C) and invasiveness (Fig.
154
2D), compared with those of the control cells.
155
3.3. Irisin-mediated activation of PI3K/AKT signaling
156
As shown in Figure 3A, compared with that in the control cells, p-AKT levels were shown to be
157
significantly increased at 5, 15, and 30 min following irisin treatment. AKT phosphorylation induced
158
by irisin was shown to be gradually suppressed by the PI3K inhibitor (40 µM) (Fig. 3B), whereas total
159
AKT levels did not decrease. To further examine this effect, HepG2 cells were treated with LY294002
160
(PI3K inhibitor) or without it in the presence of irisin. Irisin-induced HepG2 cell proliferation,
161
migration, and invasiveness were shown to be significantly reduced following LY294002 treatment
162
(Fig. 3C-3E).
163
3.4. Effect of irisin on cellular sensitivity to Dox
164
Dox treatment (10 µM) alone induced cell apoptosis, whereas addition of 2.5 nM IM significantly
165
inhibited Dox cytotoxicity. To investigate the potential mechanism underlying this effect, we treated
166
cells with LY294002 in the presence of irisin and Dox. Irisin significantly decreased the cytotoxicity
167
of Dox, whereas the PI3K inhibitor inhibited the effects of irisin (Fig. 4).
AC C
EP
TE D
M AN U
SC
RI PT
150
168 169
4. Discussion
170
Recent studies found that overexpression of irisin in the hepatic tissue of HCC patients is associated
8
ACCEPTED MANUSCRIPT with cancer [16]. However, the effects of irisin overexpression on HepG2 and SM7721 cell
172
proliferation and apoptosis have not been investigated. Consistent with the previously obtained results,
173
we found that FNDC5 levels increased in the liver of patients with HCC. Irisin has been reported to
174
significantly decrease the proliferation, viability, and migration of MDA-MB-231 and MCF-7 breast
175
cancer cells [14]. However, in endometrial, colon, thyroid, and esophageal cancer cells in vitro, irisin
176
was shown to have no effect on cell proliferation, adhesion, or colony formation relative to the
177
findings in the control samples [17]. In contrast to the previous findings [14, 17], the findings
178
obtained in this study suggest that post-translational modification of irisin and INM promote liver
179
cancer cell proliferation, migration, and invasion. The discrepancy between our findings and those
180
obtained previously may be due to differences in the cell lines used, and suggest that the effects of
181
irisin may be tissue- and cell-specific.
182
Many studies have shown that the PI3K/AKT pathway is involved in the development and
183
progression of several cancers, including glioblastoma and ovarian, pancreatic, and liver cancers.
184
PI3K/AKT has also been shown to regulate the expression of downstream target proteins, including
185
mTOR [18, 19], NF-κB [20, 21], P70S6K [22], and Bax to regulate cell proliferation and migration.
186
Further, AKT expression was found to be increased in liver cancer tissue and liver cancer cells in vitro
187
[23], and was shown to be essential in the development and progression of liver cancer. Irisin has been
188
shown to bind to as-yet unidentified receptors on the cell surface, activating p38 mitogen-activated
189
protein kinase (p38MAPK), ERK mitogen-activated protein kinase (ERK-MAPK), and AKT to
190
promote the browning of white adipocytes [24, 25], cell survival [26, 27], and pancreatic β cell
191
proliferation [28]. In this study, we demonstrated that irisin significantly increases PI3K/AKT
192
pathway activity in HepG2 cells. After 5 min, 15 min, or 30 min of treatment with 2.5 nM irisin, the
AC C
EP
TE D
M AN U
SC
RI PT
171
9
ACCEPTED MANUSCRIPT levels of AKT phosphorylation were significantly higher than those in the control group. Following
194
addition of the PI3K inhibitor LY294002, the levels of AKT protein phosphorylation declined, as did
195
cell proliferation, migration, and invasion. This indicates that the PI3K/AKT pathway plays an
196
important role in irisin-induced cell proliferation, migration, and invasion.
197
RI PT
193
The PI3K/AKT pathway was previously shown to inhibit the initiation of apoptosis through the inhibition of caspase-9 phosphorylation [29], phosphorylation of Bad protein [30], inhibition of
199
glycogen synthase kinase-3 β (p-GSK-3 β) activity [31], and regulation of the expression of
200
transcription factors such as Forkhead [32] and NF-κB [33]. In addition, the PI3K/Akt signaling
201
pathway participates in the multidrug resistance of human hepatocellular carcinoma [34]. Previous
202
studies have demonstrated that irisin inhibits high glucose-induced apoptosis of endothelial cells [26]
203
and pancreatic β cells [28]. Therefore, the present study examined whether irisin inhibited the
204
apoptosis of HepG2 cells induced by Dox and whether the PI3K/AKT pathway was involved in the
205
process. We demonstrated that the number of apoptotic cells increased significantly after intervention
206
with Dox compared with that in the control group. HepG2 cell apoptosis significantly reduced in the
207
irisin-treated group compared with that in the Dox group, and treatment with a PI3K inhibitor reduced
208
this effect. Our results show that irisin can decrease the cytotoxicity of Dox in HepG2 cells through
209
the PI3K/AKT pathway.
M AN U
TE D
EP
AC C
210
SC
198
In conclusion, the data presented in this study show that irisin regulates cell proliferation and
211
migration of human liver cancer cells in vitro. We demonstrated that irisin activates the PI3K/AKT
212
signaling pathway, promoting cell proliferation and decreasing the sensitivity to Dox. This indicates
213
that the PI3K/AKT signaling pathway plays a central role in the irisin-induced proliferation of HepG2
214
cells. Therefore, irisin may represent a novel potential target for future cancer therapies and its roles 10
ACCEPTED MANUSCRIPT 215
should be further investigated.
216
Acknowledgments
218
We would like to thank Editage [www.editage.cn] for English language editing.
RI PT
217
219
Funding
221
This study was supported by research grants from the National Natural Science Fund of China (NO.
222
81601617); Fundamental Research on Application of Qingdao Science and Technology Bureau (NO.
223
12-1-4-16-(5)-jch).
M AN U
SC
220
AC C
EP
TE D
224
11
ACCEPTED MANUSCRIPT References:
226
[1] P. Bostrom, J. Wu, M.P. Jedrychowski, A. Korde, L. Ye, J.C. Lo, K.A. Rasbach, E.A. Bostrom, J.H. Choi,
227
J.Z. Long, S. Kajimura, M.C. Zingaretti, B.F. Vind, H. Tu, S. Cinti, K. Hojlund, S.P. Gygi, B.M. Spiegelman, A
228
PGC1-alpha-dependent myokine that drives brown-fat-like development of white fat and thermogenesis, Nature,
229
481 (2012) 463-468.
230
[2] J.M. Moreno-Navarrete, F. Ortega, M. Serrano, E. Guerra, G. Pardo, F. Tinahones, W. Ricart, J.M.
231
Fernandez-Real, Irisin is expressed and produced by human muscle and adipose tissue in association with
232
obesity and insulin resistance, The Journal of clinical endocrinology and metabolism, 98 (2013) E769-778.
233
[3] S. Aydin, T. Kuloglu, M.R. Ozercan, S. Albayrak, S. Aydin, U. Bakal, M. Yilmaz, M. Kalayci, M. Yardim, M.
234
Sarac, A. Kazez, H. Kocdor, B. Kanat, I.H. Ozercan, M. Gonen, M. Bilgen, F. Balgetir, Irisin
235
immunohistochemistry in gastrointestinal system cancers, Biotechnic & histochemistry : official publication of
236
the Biological Stain Commission, 91 (2016) 242-250.
237
[4] T. Kuloglu, S. Aydin, M.N. Eren, M. Yilmaz, I. Sahin, M. Kalayci, E. Sarman, N. Kaya, O.F. Yilmaz, A.
238
Turk, Y. Aydin, M.H. Yalcin, N. Uras, A. Gurel, S. Ilhan, E. Gul, S. Aydin, Irisin: a potentially candidate marker
239
for myocardial infarction, Peptides, 55 (2014) 85-91.
240
[5] S. Aydin, T. Kuloglu, S. Aydin, M.N. Eren, A. Celik, M. Yilmaz, M. Kalayci, I. Sahin, O. Gungor, A. Gurel,
241
M. Ogeturk, O. Dabak, Cardiac, skeletal muscle and serum irisin responses to with or without water exercise in
242
young and old male rats: cardiac muscle produces more irisin than skeletal muscle, Peptides, 52 (2014) 68-73.
243
[6] K.H. Park, L. Zaichenko, M. Brinkoetter, B. Thakkar, A. Sahin-Efe, K.E. Joung, M.A. Tsoukas, E.V.
244
Geladari, J.Y. Huh, F. Dincer, C.R. Davis, J.A. Crowell, C.S. Mantzoros, Circulating irisin in relation to insulin
245
resistance and the metabolic syndrome, The Journal of clinical endocrinology and metabolism, 98 (2013)
246
4899-4907.
AC C
EP
TE D
M AN U
SC
RI PT
225
12
ACCEPTED MANUSCRIPT [7] T. Kuloglu, O. Celik, S. Aydin, I. Hanifi Ozercan, M. Acet, Y. Aydin, G. Artas, A. Turk, M. Yardim, G. Ozan,
248
M. Hanifi Yalcin, N. Kocaman, Irisin immunostaining characteristics of breast and ovarian cancer cells, Cellular
249
and molecular biology (Noisy-le-Grand, France), 62 (2016) 40-44.
250
[8] D. Us Altay, E.E. Keha, S. Ozer Yaman, I. Ince, A. Alver, B. Erdogan, S. Canpolat, U. Cobanoglu, A.
251
Mentese, Investigation of the expression of irisin and some cachectic factors in mice with experimentally
252
induced gastric cancer, QJM : monthly journal of the Association of Physicians, 109 (2016) 785-790.
253
[9] M. Kuzmicki, B. Telejko, D. Lipinska, J. Pliszka, M. Szamatowicz, J. Wilk, M. Zbucka-Kretowska, P.
254
Laudanski, A. Kretowski, M. Gorska, J. Szamatowicz, Serum irisin concentration in women with gestational
255
diabetes, Gynecological endocrinology : the official journal of the International Society of Gynecological
256
Endocrinology, 30 (2014) 636-639.
257
[10] T. Kurdiova, M. Balaz, M. Vician, D. Maderova, M. Vlcek, L. Valkovic, M. Srbecky, R. Imrich, O.
258
Kyselovicova, V. Belan, I. Jelok, C. Wolfrum, I. Klimes, M. Krssak, E. Zemkova, D. Gasperikova, J. Ukropec,
259
B. Ukropcova, Effects of obesity, diabetes and exercise on Fndc5 gene expression and irisin release in human
260
skeletal muscle and adipose tissue: in vivo and in vitro studies, The Journal of physiology, 592 (2014)
261
1091-1107.
262
[11] T. Ebert, H. Stepan, S. Schrey, S. Kralisch, J. Hindricks, L. Hopf, M. Platz, U. Lossner, B. Jessnitzer, S.
263
Drewlo, M. Bluher, M. Stumvoll, M. Fasshauer, Serum levels of irisin in gestational diabetes mellitus during
264
pregnancy and after delivery, Cytokine, 65 (2014) 153-158.
265
[12] H.J. Zhang, X.F. Zhang, Z.M. Ma, L.L. Pan, Z. Chen, H.W. Han, C.K. Han, X.J. Zhuang, Y. Lu, X.J. Li,
266
S.Y. Yang, X.Y. Li, Irisin is inversely associated with intrahepatic triglyceride contents in obese adults, Journal
267
of hepatology, 59 (2013) 557-562.
268
[13] X. Provatopoulou, G.P. Georgiou, E. Kalogera, V. Kalles, M.A. Matiatou, I. Papapanagiotou, A. Sagkriotis,
AC C
EP
TE D
M AN U
SC
RI PT
247
13
ACCEPTED MANUSCRIPT G.C. Zografos, A. Gounaris, Serum irisin levels are lower in patients with breast cancer: association with disease
270
diagnosis and tumor characteristics, BMC cancer, 15 (2015) 898.
271
[14] N.P. Gannon, R.A. Vaughan, R. Garcia-Smith, M. Bisoffi, K.A. Trujillo, Effects of the exercise-inducible
272
myokine irisin on malignant and non-malignant breast epithelial cell behavior in vitro, International journal of
273
cancer, 136 (2015) E197-202.
274
[15] Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249
275
causes of death, 1980-2015: a systematic analysis for the Global Burden of Disease Study 2015, Lancet
276
(London, England), 388 (2016) 1459-1544.
277
[16] M. Gaggini, M. Cabiati, S. Del Turco, T. Navarra, P. De Simone, F. Filipponi, S. Del Ry, A. Gastaldelli, G.
278
Basta, Increased FNDC5/Irisin expression in human hepatocellular carcinoma, Peptides, 88 (2017) 62-66.
279
[17] H.S. Moon, C.S. Mantzoros, Regulation of cell proliferation and malignant potential by irisin in
280
endometrial, colon, thyroid and esophageal cancer cell lines, Metabolism: clinical and experimental, 63 (2014)
281
188-193.
282
[18] J.X. Zhao, Y.W. Yuan, C.F. Cai, D.Y. Shen, M.L. Chen, F. Ye, Y.J. Mi, Q.C. Luo, W.Y. Cai, W. Zhang, Y.
283
Long, Y. Zeng, G.D. Ye, S.Y. Yang, Aldose reductase interacts with AKT1 to augment hepatic AKT/ mTOR
284
signaling and promote hepatocarcinogenesis, Oncotarget, (2017).
285
[19] H. Zhu, Q. Liu, J. Tang, Y. Xie, X. Xu, R. Huang, Y. Zhang, K. Jin, B. Sun, Alpha1-ACT Functions as a
286
Tumour Suppressor in Hepatocellular Carcinoma by Inhibiting the PI3K/AKT/mTOR Signalling Pathway via
287
Activation of PTEN, Cellular physiology and biochemistry : international journal of experimental cellular
288
physiology, biochemistry, and pharmacology, 41 (2017) 2289-2306.
289
[20] H. Chen, Y. Huang, J. Huang, L. Lin, G. Wei, Gigantol attenuates the proliferation of human liver cancer
290
HepG2 cells through the PI3K/Akt/NF-kappaB signaling pathway, Oncology reports, 37 (2017) 865-870.
AC C
EP
TE D
M AN U
SC
RI PT
269
14
ACCEPTED MANUSCRIPT [21] B. Hu, M. Sun, J. Liu, G. Hong, Q. Lin, The preventative effect of Akt knockout on liver cancer through
292
modulating NF-kappaB-regulated inflammation and Bad-related apoptosis signaling pathway, International
293
journal of oncology, 48 (2016) 1467-1476.
294
[22] N. Babchia, A. Calipel, F. Mouriaux, A.M. Faussat, F. Mascarelli, The PI3K/Akt and mTOR/P70S6K
295
signaling pathways in human uveal melanoma cells: interaction with B-Raf/ERK, Investigative ophthalmology
296
& visual science, 51 (2010) 421-429.
297
[23] B. Tang, F. Tang, Z. Wang, G. Qi, X. Liang, B. Li, S. Yuan, J. Liu, S. Yu, S. He, Upregulation of
298
Akt/NF-kappaB-regulated inflammation and Akt/Bad-related apoptosis signaling pathway involved in hepatic
299
carcinoma process: suppression by carnosic acid nanoparticle, International journal of nanomedicine, 11 (2016)
300
6401-6420.
301
[24] J.I. Castillo-Quan, From white to brown fat through the PGC-1alpha-dependent myokine irisin:
302
implications for diabetes and obesity, Disease models & mechanisms, 5 (2012) 293-295.
303
[25] Y. Zhang, R. Li, Y. Meng, S. Li, W. Donelan, Y. Zhao, L. Qi, M. Zhang, X. Wang, T. Cui, L.J. Yang, D.
304
Tang, Irisin stimulates browning of white adipocytes through mitogen-activated protein kinase p38 MAP kinase
305
and ERK MAP kinase signaling, Diabetes, 63 (2014) 514-525.
306
[26] H. Song, F. Wu, Y. Zhang, Y. Zhang, F. Wang, M. Jiang, Z. Wang, M. Zhang, S. Li, L. Yang, X.L. Wang, T.
307
Cui, D. Tang, Irisin promotes human umbilical vein endothelial cell proliferation through the ERK signaling
308
pathway and partly suppresses high glucose-induced apoptosis, PloS one, 9 (2014) e110273.
309
[27] W.Y. So, P.S. Leung, Irisin ameliorates hepatic glucose/lipid metabolism and enhances cell survival in
310
insulin-resistant human HepG2 cells through adenosine monophosphate-activated protein kinase signaling, The
311
international journal of biochemistry & cell biology, 78 (2016) 237-247.
312
[28] S. Liu, F. Du, X. Li, M. Wang, R. Duan, J. Zhang, Y. Wu, Q. Zhang, Effects and underlying mechanisms of
AC C
EP
TE D
M AN U
SC
RI PT
291
15
ACCEPTED MANUSCRIPT irisin on the proliferation and apoptosis of pancreatic beta cells, PloS one, 12 (2017) e0175498.
314
[29] S.B. Bratton, G. Walker, S.M. Srinivasula, X.M. Sun, M. Butterworth, E.S. Alnemri, G.M. Cohen,
315
Recruitment, activation and retention of caspases-9 and -3 by Apaf-1 apoptosome and associated XIAP
316
complexes, The EMBO journal, 20 (2001) 998-1009.
317
[30] J. Sakamaki, H. Daitoku, K. Ueno, A. Hagiwara, K. Yamagata, A. Fukamizu, Arginine methylation of
318
BCL-2 antagonist of cell death (BAD) counteracts its phosphorylation and inactivation by Akt, Proceedings of
319
the National Academy of Sciences of the United States of America, 108 (2011) 6085-6090.
320
[31] Y.F. Xian, Z.X. Lin, Q.Q. Mao, J.N. Chen, Z.R. Su, X.P. Lai, P.S. Ip, Isorhynchophylline Protects PC12
321
Cells Against Beta-Amyloid-Induced Apoptosis via PI3K/Akt Signaling Pathway, Evidence-based
322
complementary and alternative medicine : eCAM, 2013 (2013) 163057.
323
[32] S.R. Boreddy, K.C. Pramanik, S.K. Srivastava, Pancreatic tumor suppression by benzyl isothiocyanate is
324
associated with inhibition of PI3K/AKT/FOXO pathway, Clinical cancer research : an official journal of the
325
American Association for Cancer Research, 17 (2011) 1784-1795.
326
[33] H. Sui, S.F. Pan, Y. Feng, B.H. Jin, X. Liu, L.H. Zhou, F.G. Hou, W.H. Wang, X.L. Fu, Z.F. Han, J.L. Ren,
327
X.L. Shi, H.R. Zhu, Q. Li, Zuo Jin Wan reverses P-gp-mediated drug-resistance by inhibiting activation of the
328
PI3K/Akt/NF-kappaB pathway, BMC complementary and alternative medicine, 14 (2014) 279.
329
[34] L. Cheng, S. Luo, C. Jin, H. Ma, H. Zhou, L. Jia, FUT family mediates the multidrug resistance of human
330
hepatocellular carcinoma via the PI3K/Akt signaling pathway, Cell death & disease, 4 (2013) e923.
SC
M AN U
TE D
EP
AC C
331
RI PT
313
332 333 334 16
ACCEPTED MANUSCRIPT 335
Figure legends
336
Figure 1. Hepatic FNDC5 levels in hepatocellular carcinoma patients and controls. A: Plasma irisin
338
levels did not different between HCC patients and controls. B: Heptic FNDC5/Irisin mRNA is
339
over-expressed in HCC patients respect to the controls. *** P<0.001, compared with the controls.
340
Figure 2. Effects of irisin on proliferation and migration of HepG2 and SMCC7721 cells. A: Viability
341
of HepG2 and SMCC7721 cells treated with medium, or with different concentrations of modified
342
irisin (IM) or nonmodified irisin (INM) ranging from 0.625 to 20 nM (N=8 in each group). B: Optical
343
density at 450 nm, measured in the CCK8 assay, following treatment with IM (2.5 nM) at 24, 48, and
344
72 h (N=8 in each group). C and D: Migration and invasiveness of the HepG2 cells treated as
345
indicated (N=5 in each group). * P<0.05, ** P<0.01 and *** P<0.001, compared with the control. #
346
P<0.05, compared with the control.
347
Figure 3. Effects of irisin on adipocyte proliferation, migration, and invasion through the PI3K/AKT
348
pathway. A: Phosphorylated and total AKT levels in HepG2 cells treated with phosphate-buffered
349
saline (PBS; control) or r-irisin (2.5 nmol/L) at the indicated time points (N=3). B: Phosphorylated
350
and total AKT levels in HepG2 cells pretreated with LY294002 at the indicated concentrations,
351
followed by irisin treatment (N=3). C-E: Inhibition of the PI3K/AKT pathway reduces cell
352
proliferation (N=8), migration (N=5), and invasion (N=5). * P<0.05, ** P<0.01, and *** P<0.001,
353
compared with the control. # #P<0.01, and ### compared with the irisin group.
354
Figure 4. Effects of irisin on HepG2 cell apoptosis induced by doxorubicin. HepG2 cells were treated
355
with medium or doxorubicin (10 µM) with or without modified irisin (IM; 2.5 nM) or LY294002 for
AC C
EP
TE D
M AN U
SC
RI PT
337
17
ACCEPTED MANUSCRIPT 356
24 h (N=3). ** P<0.01, compared with the control. # P<0.05, compared with the Dox group. + P<0.05,
357
compared with the Dox + irisin group.
359
RI PT
358
Figure 1
M AN U
SC
360
AC C
EP
TE D
361
18
ACCEPTED MANUSCRIPT Figure 2
M AN U
SC
RI PT
362
EP
365
AC C
364
TE D
363
19
ACCEPTED MANUSCRIPT Figure 3
M AN U
SC
RI PT
366
EP
369
AC C
368
TE D
367
20
ACCEPTED MANUSCRIPT 370
Figure 4
374 375 376 377 378
EP
373
AC C
372
TE D
M AN U
SC
RI PT
371
379 380
21
ACCEPTED MANUSCRIPT
Highlights:
RI PT
SC M AN U TE D EP
• •
Irisin levels are upgraded in patients with hepatocellular carcinoma. Irisin induces HEPG2 and SMCC7721 cell proliferation, migration, invasiveness, and inhibit appotosis of HEPG2 cell induced by Dox. Irisin induces the activation of PI3K/AKT pathway. Irisin has protective roles in liver cancer cells, inducing disease progression.
AC C
• •
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
Conflict of Interest Guangjun Shi declares that he has no conflict of interest. Nan Tang Shi declares that he has no conflict of interest. Jiantao Qiu declares that he has no conflict of interest. Deguo Zhang declares that he has no conflict of interest. Fei Huang declares that he has no conflict of interest. Kun Ding declares that she has no conflict of interest. Weisheng Li declares that he has no conflict of interest. Ping Zhang declares that she has no conflict of interest. Xueying Tan declares that he has no conflict of interest.