Quercetin induces growth arrest through activation of FOXO1

advertisement
1
Quercetin induces growth arrest through activation of FOXO1
2
transcription factor in EGFR-overexpressing oral cancer cells
3
4
Chun-Yin Huang a, Chien-Yi Chan a,†, I-Tai Chou a,†, Chia-Hsien Lien a, Hsiao-Chi Hung a,
5
Ming-Fen Lee b,*
6
7
a
Department of Nutrition, China Medical University, Taichung, Taiwan, ROC
8
b
Department of Nutrition and Health Sciences, Chang Jung Christian University, Tainan,
9
Taiwan, ROC
10
11
Correspondence: Dr. Ming-Fen Lee, Department of Nutrition and Health Sciences, Chang
12
Jung Christian University, 396 Sec. 1 Changrong Rd., Gueiren Dist., Tainan, Taiwan. Tel:
13
+886 6 2785123 x3308; Fax: +886 6 2785926, E-mail: leemf@mail.cjcu.edu.tw
14
15
†
These authors contributed equally.
16
17
Running title: Quercetin, FOXO1, EGFR, and oral cancer
18
19
Keywords: EGFR; FOXO1; oral cancer; quercetin
1
20
21
Funding sources: This work was supported by the grants, CMU97-303 (China Medical
22
University) and NSC98-2320-B-039-021-MY3 (National Science Council) to Chun-Yin
23
Huang, and NSC98-2320-B-309-002-MY3 and NSC101-2320-B-309-001 (National Science
24
Council) to Ming-Fen Lee.
25
2
26
Abstract
27
28
The squamous cell carcinomas of the head and neck (SCCHN) with aberrant epidermal
29
growth factor receptor (EGFR) signaling are often associated with poor prognosis and low
30
survival. Therefore, efficient inhibition of the EGFR signaling could intervene with the
31
development of malignancy. Quercetin appears anti-tumorigenesis, but the underlying
32
mechanism remains unclear in oral cancer. Fork-head box O (FOXO) transcription factors,
33
Akt downstream effectors, are important regulators of cell growth. Here, we hypothesized
34
that FOXO1 might be crucial in quercetin-induced growth inhibition in
35
EGFR-overexpressing oral cancer. Quercetin treatment suppressed cell growth by inducing
36
G2 arrest and apoptosis in EGFR-overexpressing HSC-3 and TW206 oral cancer cells.
37
Quercetin inhibited EGFR/Akt activation with a concomitant induction of FOXO1 activation.
38
FOXO1-knockdown attenuated quercetin-induced p21 and FasL expression and subsequent
39
G2 arrest and apoptosis, respectively. Likewise, quercetin suppressed tumor growth in HSC-3
40
xenograft mice. Taken together, our data indicate that quercetin is an effective anti-cancer
41
agent and that FOXO1 is crucial in quercetin-induced growth suppression in
42
EGFR-overexpressing oral cancer.
43
44
3
45
1. Introduction
46
47
The invasive squamous cell carcinomas of the head and neck (SCCHN) frequently
48
overexpress epidermal growth factor receptor (EGFR) [1], leading to unfavorable clinical
49
outcome–high recurrence and low survival rates [2, 3]. Aberrant EGFR/PI3K/Akt signaling
50
pathway is often responsible for the malignant phenotype [4]. Although several
51
EGFR-targeting drugs are clinically available [5-7], their wide application is limited by the
52
resistance to the drugs and mutations in the EGFR downstream effectors in some patients [8].
53
Therefore, to improve therapeutic efficacy, new anti-cancer agent should be considered
54
accordingly.
55
56
Quercetin (3,3’,4’,5,7-pentahydroxyflavone) is one of the major dietary flavonoids
57
found in a wide range of fruits, vegetables, and beverages [9]. Among those, onion, apple,
58
and red wine are good sources of quercetin. The antioxidant, anti-inflammatory, as well as
59
anti-proliferative and apoptosis aspects of quercetin have been widely investigated [9]. A
60
phase I clinical trial indicated that quercetin can be safely administered and its plasma levels
61
are sufficient to inhibit lymphocyte tyrosine kinase activity [10]. Consumption of quercetin
62
from onions and apples was inversely associated with lung cancer risk, supporting the
63
chemopreventive effect of quercetin [11]. The chemotherapeutic efficacy of quercetin has
4
64
also been demonstrated in many types of cancer [12-18]. Quercetin exerts its anti-cancer
65
property mainly through induction of growth arrest in G1 or G2 [14, 15, 17, 19], apoptosis
66
[15, 16, 20], and inhibition of angiogenesis [13] as evidenced by down-regulating the
67
expression of oncogenes (HER2 [21], H-ras, K-ras, c-myc [22], or COX-2 [13] ) and mutant
68
p53 [23], or up-regulating cell cycle control proteins (p21WAF1 and p27KIP1) [18, 19, 24].
69
In addition, quercetin inhibits several tyrosine and serine–threonine kinases, whose activities
70
are linked to cell surface receptors-transduced survival pathways (PI3K/Akt/PKB) [25-27].
71
Nonetheless, the effect of quercetin on oral cancers remains uncertain. Quercetin seems to
72
induce apoptosis in some human oral cancer cell lines [28-30]; however, the underlying
73
mechanisms are still elusive.
74
75
Fork-head box O (FOXO) transcription factors share a conserved “fork-head box”
76
DNA-binding domain, which comprise four members in mammals: FOXO1, FOXO3a,
77
FOXO4, and FOXO6 [31, 32]. FOXO factors are involved in a wide range of biological
78
processes, including cell cycle arrest, apoptosis, DNA repair, glucose metabolism, oxidative
79
stress resistance, and longevity [32]. The biological activity of FOXO factors is highly
80
dependent on their intracellular trafficking, mediated by post-translational modifications in
81
phosphorylation, acetylation, or ubiquitination [33]. In addition, FOXO factors share highly
82
conserved sites within and nearby their DNA binding domains for phosphorylation by the
5
83
survival PI3K/Akt signaling pathway. The growth factors-mediated Akt phosphorylation on
84
these conserved residues negatively regulates FOXO expression and activity [34]. In this
85
regard, FOXO factors are implicated in pathogenesis including aging, diabetes, immune
86
disease, infertility, neurodegeneration, and cancer [35]. Among FOXO factors, Increased
87
p-FOXO1 or decreased FOXO1 expression is often associated with tumorigenesis [36].
88
FOXO1 may exert its tumor suppression function through its transcription-dependent
89
expression on growth arrest and apoptosis-related genes, including p15, p19, NOXA, FasL,
90
TRAIL, and Bim [37-39]. The promoters of these genes comprise the consensus sequence
91
T/C-G/A-A-A-A-C-A-A recognized by FOXO1 [35]. Alternatively, FOXO1 can mediate
92
cancer cell death through induction of autophagy which is independent of its transcriptional
93
activity [40]. Similarly, FOXO1 inhibits Runx2-mediated cell migration and invasion
94
independent of its transcriptional activity [41]. Together, FOXO1 appears to be a potential
95
therapeutic target of anti-cancer reagents [42-46].
96
97
Aberrant EGFR expression and PI3K mutation are often observed in invasive SCCHN;
98
however, the role of transcription factor FOXO1 as a downstream target of activated
99
EGFR/PI3K/Akt signaling axis in oral cancer still remains unclear. Therefore, in the present
100
study, we aimed to investigate the anti-cancer efficacy of quercetin in human oral cancer cell
101
lines with elevated EGFR expression, and to further identify the crucial role of FOXO1 factor
6
102
in quercetin-mediated growth suppression.
7
103
2. Materials and methods
104
105
2.1 Reagents, antibodies, and plasmids
106
107
All chemicals including quercetin were purchased from Sigma (St. Louis, MO) and
108
antibodies were purchased from Cell Signaling Technology (Beverly, MA), respectively,
109
unless specified otherwise. PVDF membrane and ECL detection reagents were from Perkin
110
Elmer Life Sciences, Inc. The annexin V-FITC apoptosis detection kit was purchased from
111
BD Biosciences (San Jose, CA). Antibody against cyclin D1 was purchased from Santa Cruz
112
Biotechnology (Santa Cruz, CA) and anti-α-tubulin antibody was purchased from Abcam
113
(Cambridge, MA). FOXO1-specific siRNA and the Lipofectamine RNAiMAX reagent were
114
purchased from Invitrogen (Grand Island, NY). The pGL3-FHRE-luc plasmid was obtained
115
from Addgene (Cambridge, USA; Addgene database plasmid 1789) and the PolyJet In Vitro
116
DNA Transfection Reagent was purchased from SignaGen Laboratories (Ijamsville, MD).
117
The NE-PER Nuclear and Cytoplasmic Extraction Reagents were purchased from Thermo
118
Fisher Scientific (Rockford, IL).
119
120
2.2 Cell culture and treatment
121
8
122
HSC-3 and TW206 human oral squamous carcinoma and HGF human gingival fibroblasts
123
cells were kind gifts of Dr. Hsin-Ling Yang at China Medical University (Taichung, Taiwan).
124
HSC-3 and HGF cells were maintained in DMEM-F12 and DMEM (Invitrogen), respectively,
125
supplemented with 10% fetal bovine serum (FBS) and 1% antibiotic-antibimycotic (Gibco).
126
All cells were cultured with 5% CO2 at 37°C.
127
128
2.3 Cell viability assay
129
130
Cell viability was measured by trypan blue method. Cells were allowed to grow in 6-well
131
culture plates and then treated with various concentrations of quercetin (0-50 μM) for
132
indicated hours. Then, cells were trypsinized and viable cells were stained with trypan blue
133
for hemocytometric counting.
134
135
2.4 Western blotting
136
137
Cells were washed with cold PBS and lysed in RIPA buffer containing 150 mM NaCl, 10
138
mM Tris (pH 7.2), 0.1% SDS, 1% Triton X-100, 1% deoxycholate, 5 mM EDTA, and
139
protease/phosphatase inhibitors. Protein concentration was determined by BCA protein assay,
140
and denatured proteins were separated in 8% to 15% SDS-PAGE, and transferred onto PVDF
9
141
membranes. Nonspecific binding was blocked with 5% milk in TBST buffer (20 mM Tris
142
base, 140 mM NaCl, pH 7.6, 0.1% Tween-20) for 1 h, followed by incubation with primary
143
antibodies at 4°C overnight and secondary antibodies at room temperature for 1 h. Blots were
144
visualized by ECL detection reagents.
145
146
2.5 Cell cycle analysis
147
148
HSC-3 and TW206 cells were seeded onto 6-well plates and serum-starved for 18 h followed
149
by quercetin treatment in growth media for 24 h. Cells were then trypsinized, washed with
150
PBS, and fixed in cold 70% ethanol at -20 °C. The fixed cells were collected and stained in a
151
solution containing 0.5 ml of 4 μg/ml of PI, 0.5 mg/ml of RNase, and 1% Triton X-100 for 30
152
min at 4 °C. DNA content of these cells was analyzed using a FACScan flow cytometer
153
(Becton Dickinson).
154
155
2.6 Colony formation assay
156
157
After incubation with the indicated concentration of quercetin, cells were washed with PBS,
158
fixed with 10% formalin (Mallinckrodt Chemicals) for 10 min, and stained with 0.05%
159
crystal violet (Panreac Quimica S.A.U.) for 30 min. Dishes were subjected to colony
10
160
counting and optical density measurement at 540 nm.
161
162
2.7 Apoptosis analysis
163
164
Cells were seeded onto 6-well plates in growth media and treated with quercetin for 24 h.
165
Cells were then trypsinized, washed with PBS, and resuspended in binding buffer (10 mM
166
Hepes/NaOH, pH 7.4, 140 mM NaCl, and 2.5 mM CaCl2) at a concentration of 1×106
167
cells/ml. One hundred microliters of the lysis solution were transferred to a polystyrene tube,
168
followed by 5 μl of annexin V-FITC and 5 μl of PI. The mixtures were sheltered from light
169
and incubated for 15 min at room temperature. Finally, 400 μl of 1× binding buffer was
170
added to each tube and cells were analyzed using a FACScan flow cytometer (Becton
171
Dickinson).
172
173
2.8 Reporter assay
174
175
The pGL3-FHRE-luc and β-galactosidase plasmids with or without FOXO1 siRNA were
176
co-transfected into HSC-3 cells for 24 h, followed by another 24 h of quercetin treatment.
177
Cell lysates were collected in lysis solution (Applied Biosystems, Carlsbad, CA) according to
178
the manufacturer’s protocol. The Dual Light® System (Applied Biosystems) was used to
11
179
quantify luciferase and β-galactosidase activities.
180
181
2.9 Xenograft mice
182
183
Seven-week-old male nude mice were purchased from National Laboratory Animal Center
184
(Taipei, Taiwan). The animal study protocol, IACUC 98-71-N, was reviewed and approved
185
by the Institutional Animal Care and Use Committee (IACUC) of the China Medical
186
University. The animals were given free access to autoclaved food and water during the study.
187
After 2-week of acclimation, animals were implanted subcutaneously with HSC-3 cells (3 X
188
106) suspended with liquid Matrigel to a final volume of 100 μl into the flank regions and
189
randomly assigned into the control or quercetin group. Tumor size was recorded every other
190
day for 40 days. Two weeks after the implantation, quercetin (3 mg/kg/day) or PBS was
191
given via gavage feeding. Tumor volume = ½(length × width2).
192
193
2.10 Statistical analysis
194
195
Data are expressed as mean ± SD from at least three independent experiments. Statistical
196
significance was analyzed using Student’s t test or Duncan’s multiple range tests. Results
197
were considered significantly different at p < 0.05.
12
198
3
Results
199
200
3.1 Quercetin inhibits cell growth in human oral cancer cells
201
202
The growth-inhibitory effect of quercetin has been shown in various types of cancer; however,
203
its efficacy on oral squamous cancer cells remains elusive. To examine the effect of quercetin
204
on oral cancer, we exposed two human tongue squamous carcinoma cell lines, HSC-3 and
205
TW206 [47], and a control, human gingival fibroblast HGF cells, to various concentrations of
206
quercetin. Quercetin potently inhibited HSC-3 cell growth as compared with TW206 and
207
HGF cells (Fig. 1A). The IC50 of quercetin in HSC-3 cells was around 20 μM, much less
208
than that of TW206 (45 μM). These results indicate that the metastatic HSC-3 cell line is
209
more susceptible to quercetin than the other two cell lines tested. Indeed, quercetin
210
supplementation for 7 days significantly suppressed HSC-3 colony formation in a
211
dose-dependent manner with an IC50 of 7 μM (Fig. 1B). These data support quercetin as a
212
potential anti-cancer reagent in oral squamous carcinomas.
213
214
Cell cycle arrest and apoptosis are two main factors responsible for growth suppression.
215
Quercetin has been shown to cause growth arrest in various cancer cells [14, 15, 17, 19]. To
216
investigate the effect of quercetin on cell proliferation in oral cancer, we performed flow
13
217
cytometric analysis for cell cycle distribution upon quercetin addition. In both HSC-3 and
218
TW206 cells, quercetin (20 μM) treatment for 24 h resulted in a significant increase of cell
219
numbers in G2/M phase (Fig. 2A), with concomitant reductions of G2/M phase cyclins,
220
cyclin A and B1 [48], whereas quercetin showed no suppressive effect on G1 phase cyclin,
221
cyclin D1 (Fig. 2B). In addition, quercetin induced the expression of p21 but not negative G1
222
regulator, p15 [49] (Fig. 2B). These results demonstrate that quercetin is a potent inducer of
223
growth arrest in human oral squamous carcinoma cells.
224
225
Next, we evaluated the apoptotic efficacy of quercetin in oral cancer cells. Upon
226
quercetin treatment (0-30 μM), both HSC-3 and TW206 cells displayed an accumulation of
227
apoptotic cells in a dose-dependent manner (Fig. 2C). The apoptosis-inducing effect was
228
further confirmed by the observations of the condensed chromatin staining (Fig. 2D) and the
229
increased FasL and cleaved caspase 3 (Fig. 2E) upon quercetin treatment in HSC-3 cells.
230
However, quercetin had little effect on the expression of Bax, a pro-apoptotic member of the
231
Bcl-2 family (Fig. 2E). These results suggest the potential involvement of FasL-mediated
232
extrinsic apoptotic pathway in quercetin-mediated cell death of HSC-3. Taken together, both
233
growth arrest and apoptosis are responsible for the anti-cancer effect of quercetin in oral
234
squamous carcinoma cells.
235
14
236
3.2 Quercetin modulates the EGFR/Akt signaling axis
237
238
Aberrant EGFR activation is the key cause of oral cancer malignancy. Therefore, we studied
239
the effect of quercetin on EGFR signaling. In EGFR-overexpressing HSC-3 cells (Fig. 3A),
240
the protein levels of p-Y1086-EGFR and its downstream effector, p-Akt, were decreased
241
upon quercetin treatment in both time- (Fig. 3B) and dose-(Fig. 3C) dependent patterns,
242
whereas the p-Y1045-EGFR levels were gradually increased (Fig. 3B). It is noteworthy that
243
the phosphorylation at tyrosine 1045 is responsible for c-Cbl-mediated ubiquitination and
244
degradation of EGFR protein [50]. These results suggest quercetin as an important modulator
245
of the EGFR/Akt pathway.
246
247
3.3 Quercetin regulates FOXO1 activation
248
249
FOXO transcription factors play a role in the regulation of cell growth and apoptosis. The
250
activity of FOXO factors can be regulated by Akt-mediated nucleus exclusion and subsequent
251
degradation [51]. Therefore, we examined the effect of quercetin on FOXOs. While HSC-3
252
cells displayed increased FOXO1 protein levels in both dose- and time-dependent manners
253
(Fig. 4A-B) upon quercetin treatment, no change of FOXO3a expression was observed (Fig.
254
4B). In addition, quercetin reduced the phosphorylation of cytoplasmic FOXO1 at serine 256
15
255
(Fig. 4C), and induced the translocation of FOXO1 from cytoplasm to nucleus as shown by
256
Western blotting (Fig. 4C) and immunofluorescence (Fig. 4D) in HSC-3 cells. These results
257
suggest that quercetin induces the nuclear translocation of FOXO1 and, thereby,
258
transactivates FOXO1. Indeed, quercetin activated a luciferase reporter gene driven by three
259
fork-head responsive elements (FHREs) in HSC-3 cells, whereas such activation was
260
abolished upon FOXO1 knockdown (Fig. 4E). Since these FHREs were originally identified
261
in FasL promoter [52], our data indicate that quercetin activates FOXO1 and induces FasL
262
expression. Furthermore, as in the treatment of quercetin (Fig. 3A), the administration of
263
LY294002, a PI3K inhibitor, increased FOXO1 expression in a time-dependent manner in
264
HSC-3 cells (Fig. 4F). Taken together, FOXO1 is a downstream transducer of quercetin.
265
266
3.4 Quercetin mediates growth inhibition through FOXO1 activation
267
268
To verify if FOXO1 plays a key role in quercetin-mediated growth inhibition in oral cancer
269
cells, we suppressed FOXO1 expression by FOXO1-specific siRNA in the presence of
270
quercetin. FOXO1 knockdown relieved the suppression effect of quercetin on cell growth
271
(Fig. 5A) and colony formation (Fig. 5B). In particular, FOXO1 suppression abolished
272
quercetin-mediated G2/M arrest in both HSC-3 and TW206 cells (Fig. 5C). FOXO1
273
knockdown reduced quercetin-induced p21 expression (Fig. 5D); ectopic expression of
16
274
FOXO1 resulted in increased p21 expression (Fig. 5E). These results support the role of
275
FOXO1 in quercetin-mediated growth inhibition where the subsequent p21 expression might
276
be responsible for the G2 arrest.
277
278
We next examined the role of FOXO1 in quercetin-mediated apoptosis. FOXO1
279
knockdown alleviated quercetin-induced apoptosis (Fig. 6A) and reduced the expression
280
levels of FasL and cleaved caspase 3 (Fig. 6B) in HSC-3 cells. These results further suggest
281
that quercetin-induced cell death might, in part, through FOXO1-mediated FasL expression.
282
283
3.5 Quercetin inhibits tumor growth with concomitant FOXO1 induction in vivo
284
285
The anti-growth effect of quercetin on HSC-3 cells was further tested with xenograft mice.
286
Both tumor size and weight of HSC-3-engineered xenografts were significantly suppressed
287
upon quercetin administration as compared with the control (Fig. 7A-B). In addition,
288
quercetin-fed xenograft mice also exhibited higher FOXO1 expression than those of control
289
animals (Fig. 7C). Therefore, in consistent with the in vitro results, the in vivo data indicate
290
that quercetin inhibits tumor growth and induces FOXO1 expression.
291
17
292
4
Discussion
293
294
Aberrant activation of the EGFR/PI3K/Akt signaling pathway is often associated with cancer
295
malignancy [53]. Accordingly, this deregulated signaling pathway becomes a conceivable
296
therapeutic target in anti-cancer strategy. HSC-3 cell line, a cervical lymph node metastasis
297
originated from a tongue primary tumor, displays activated EGFR/PI3K/Akt signaling by
298
expressing augmented EGFR levels and a constitutively active PI3K mutant [53]. Quercetin
299
has been reported as a potent PI3K inhibitor [25], and the efficacy of quercetin inhibition on
300
tyrosine kinase may continue for 16 h [10]. In the current study, we observed
301
down-regulation of EGFR and Akt phosphorylation upon quercetin treatment for 12 h in
302
HSC-3 cells. It is possible that quercetin might compete with ATP for the binding to the
303
catalytic site of the PI3K [25]. Our data potentiate quercetin as an alternative regimen for oral
304
cancer patients carrying an aberrant EGFR/PI3K/Akt signaling axis.
305
306
Oral squamous cancer cells with EGFR overexpression appear susceptible to
307
anti-proliferation effect of quercetin. The concentration of quercetin to inhibit SCC-1483 cell
308
viability by half is about 40 μM [29], in contrast with 20 μM for EGFR-overexpressing
309
HSC-3 oral cancer cell line. In the current study, quercetin-induced G2 arrest is associated
310
with the induction of FOXO1-dependent p21 expression in HSC-3 human oral cancer cells.
18
311
Similar induction effect on p21 has also been reported in quercetin-treated breast and prostate
312
cancer cells [17, 18]. However, the concentration of quercetin to induce p21 expression in
313
PC-3 cells was much higher (50-100 μM) than that of HSC-3 cells.
314
315
The involvement of p21 in G2 arrest could result from either DNA damage or mitogen
316
starvation [49]. Seoane et al. proposed that, in response to TGF-β, FOXO1 forms a complex
317
with Smad 3/4, which further induces p21 expression through the binding of the Forkhead
318
binding element within the p21 promoter [54]. The link between FOXO1 and p21 was further
319
supported by the finding that FOXO1-mediated p21 expression is crucial in the antineoplastic
320
effect of calorie restriction [55].
321
322
Quercetin may exert apoptotic function in a FOXO1-dependent fashion. Here, we
323
demonstrated that quercetin treatment caused apoptosis in both HSC-3 and TW206 cells (Fig.
324
2C). This phenomenon was supported by increased levels of cleaved caspase 3 and FasL
325
upon quercetin addition (Fig. 2E). In particular, FOXO1 suppression alleviated
326
quercetin-mediated cell death and reduced the induction of FasL and cleaved caspase 3 (Fig.
327
6). Mutant p53 could block apoptosis, whereas quercetin can down-regulate mutant p53
328
protein levels [23]. Since HSC-3 carries a truncated p53 mutant [56], this could be an
329
alternative mechanism how quercetin facilitates cell death in HSC-3 cells.
19
330
331
In summary, our data support quercetin as an efficient anti-cancer agent in
332
EGFR-overexpressing oral cancers; the schematic mechanism is illustrated in Fig. 8. By
333
suppressing EGFR/Akt activation, quercetin stimulates FOXO1 activation and subsequently
334
induces p21 and FasL which link to G2 arrest and apoptosis, respectively. The current study
335
provides several lines of evidence both in vivo and in vitro to support quercetin as a potential
336
therapeutic agent for a subset of oral squamous carcinomas. Specifically, the mechanism
337
involving FOXO1 in the anti-cancer action of quercetin may be translated to the future
338
application as clinical stratagem in cure of oral cancer.
339
340
Conflict of interest
341
The authors declare no conflict of interest.
342
20
343
5
References
344
345
[1] Sheu JJ, Hua CH, Wan L, Lin YJ, Lai MT, Tseng HC, et al. Functional genomic analysis
346
identified epidermal growth factor receptor activation as the most common genetic event in
347
oral squamous cell carcinoma. Cancer Res. 2009;69:2568-76.
348
[2] Chung CH, Ely K, McGavran L, Varella-Garcia M, Parker J, Parker N, et al. Increased
349
epidermal growth factor receptor gene copy number is associated with poor prognosis in head
350
and neck squamous cell carcinomas. J Clin Oncol. 2006;24:4170-6.
351
[3] Temam S, Kawaguchi H, El-Naggar AK, Jelinek J, Tang H, Liu DD, et al. Epidermal
352
growth factor receptor copy number alterations correlate with poor clinical outcome in
353
patients with head and neck squamous cancer. J Clin Oncol. 2007;25:2164-70.
354
[4] Modjtahedi H, Essapen S. Epidermal growth factor receptor inhibitors in cancer treatment:
355
advances, challenges and opportunities. Anticancer Drugs. 2009;20:851-5.
356
[5] Burtness B. The role of cetuximab in the treatment of squamous cell cancer of the head
357
and neck. Expert Opin Biol Ther. 2005;5:1085-93.
358
[6] Burtness B, Goldwasser MA, Flood W, Mattar B, Forastiere AA. Phase III randomized
359
trial of cisplatin plus placebo compared with cisplatin plus cetuximab in metastatic/recurrent
360
head and neck cancer: an Eastern Cooperative Oncology Group study. J Clin Oncol.
361
2005;23:8646-54.
21
362
[7] Dobelbower MC, Russo SM, Raisch KP, Seay LL, Clemons LK, Suter S, et al. Epidermal
363
growth factor receptor tyrosine kinase inhibitor, erlotinib, and concurrent 5-fluorouracil,
364
cisplatin and radiotherapy for patients with esophageal cancer: a phase I study. Anticancer
365
Drugs. 2006;17:95-102.
366
[8] Ratushny V, Astsaturov I, Burtness BA, Golemis EA, Silverman JS. Targeting EGFR
367
resistance networks in head and neck cancer. Cell Signal. 2009;21:1255-68.
368
[9] Lamson DW, Brignall MS. Antioxidants and cancer, part 3: quercetin. Altern Med Rev.
369
2000;5:196-208.
370
[10] Ferry DR, Smith A, Malkhandi J, Fyfe DW, deTakats PG, Anderson D, et al. Phase I
371
clinical trial of the flavonoid quercetin: pharmacokinetics and evidence for in vivo tyrosine
372
kinase inhibition. Clin Cancer Res. 1996;2:659-68.
373
[11] Le Marchand L, Murphy SP, Hankin JH, Wilkens LR, Kolonel LN. Intake of flavonoids
374
and lung cancer. J Natl Cancer Inst. 2000;92:154-60.
375
[12] Vargas AJ, Burd R. Hormesis and synergy: pathways and mechanisms of quercetin in
376
cancer prevention and management. Nutr Rev. 2010;68:418-28.
377
[13] Xiao X, Shi D, Liu L, Wang J, Xie X, Kang T, et al. Quercetin suppresses
378
cyclooxygenase-2 expression and angiogenesis through inactivation of P300 signaling. PLoS
379
One. 2011;6:e22934.
380
[14] Vidya Priyadarsini R, Senthil Murugan R, Maitreyi S, Ramalingam K, Karunagaran D,
22
381
Nagini S. The flavonoid quercetin induces cell cycle arrest and mitochondria-mediated
382
apoptosis in human cervical cancer (HeLa) cells through p53 induction and NF-kappaB
383
inhibition. Eur J Pharmacol. 2010;649:84-91.
384
[15] Zhang Q, Zhao XH, Wang ZJ. Cytotoxicity of flavones and flavonols to a human
385
esophageal squamous cell carcinoma cell line (KYSE-510) by induction of G2/M arrest and
386
apoptosis. Toxicol In Vitro. 2009;23:797-807.
387
[16] Rubio S, Quintana J, Eiroa JL, Triana J, Estevez F. Acetyl derivative of quercetin
388
3-methyl ether-induced cell death in human leukemia cells is amplified by the inhibition of
389
ERK. Carcinogenesis. 2007;28:2105-13.
390
[17] Vijayababu MR, Kanagaraj P, Arunkumar A, Ilangovan R, Aruldhas MM, Arunakaran J.
391
Quercetin-induced growth inhibition and cell death in prostatic carcinoma cells (PC-3) are
392
associated with increase in p21 and hypophosphorylated retinoblastoma proteins expression.
393
J Cancer Res Clin Oncol. 2005;131:765-71.
394
[18] Choi JA, Kim JY, Lee JY, Kang CM, Kwon HJ, Yoo YD, et al. Induction of cell cycle
395
arrest and apoptosis in human breast cancer cells by quercetin. Int J Oncol. 2001;19:837-44.
396
[19] Beniston RG, Campo MS. Quercetin elevates p27(Kip1) and arrests both primary and
397
HPV16 E6/E7 transformed human keratinocytes in G1. Oncogene. 2003;22:5504-14.
398
[20] Wang P, Zhang K, Zhang Q, Mei J, Chen CJ, Feng ZZ, et al. Effects of quercetin on the
399
apoptosis of the human gastric carcinoma cells. Toxicol In Vitro. 2012;26:221-8.
23
400
[21] Jeong JH, An JY, Kwon YT, Li LY, Lee YJ. Quercetin-induced ubiquitination and
401
down-regulation of Her-2/neu. J Cell Biochem. 2008;105:585-95.
402
[22] Ranelletti FO, Maggiano N, Serra FG, Ricci R, Larocca LM, Lanza P, et al. Quercetin
403
inhibits p21-RAS expression in human colon cancer cell lines and in primary colorectal
404
tumors. Int J Cancer. 2000;85:438-45.
405
[23] Avila MA, Velasco JA, Cansado J, Notario V. Quercetin mediates the down-regulation of
406
mutant p53 in the human breast cancer cell line MDA-MB468. Cancer Res. 1994;54:2424-8.
407
[24] Casagrande F, Darbon JM. Effects of structurally related flavonoids on cell cycle
408
progression of human melanoma cells: regulation of cyclin-dependent kinases CDK2 and
409
CDK1. Biochem Pharmacol. 2001;61:1205-15.
410
[25] Matter WF, Brown RF, Vlahos CJ. The inhibition of phosphatidylinositol 3-kinase by
411
quercetin and analogs. Biochem Biophys Res Commun. 1992;186:624-31.
412
[26] Gulati N, Laudet B, Zohrabian VM, Murali R, Jhanwar-Uniyal M. The antiproliferative
413
effect of Quercetin in cancer cells is mediated via inhibition of the PI3K-Akt/PKB pathway.
414
Anticancer Res. 2006;26:1177-81.
415
[27] Sun ZJ, Chen G, Hu X, Zhang W, Liu Y, Zhu LX, et al. Activation of
416
PI3K/Akt/IKK-alpha/NF-kappaB signaling pathway is required for the apoptosis-evasion in
417
human salivary adenoid cystic carcinoma: its inhibition by quercetin. Apoptosis.
418
2010;15:850-63.
24
419
[28] Chowdhury SA, Kishino K, Satoh R, Hashimoto K, Kikuchi H, Nishikawa H, et al.
420
Tumor-specificity and apoptosis-inducing activity of stilbenes and flavonoids. Anticancer Res.
421
2005;25:2055-63.
422
[29] Kang JW, Kim JH, Song K, Kim SH, Yoon JH, Kim KS. Kaempferol and quercetin,
423
components of Ginkgo biloba extract (EGb 761), induce caspase-3-dependent apoptosis in
424
oral cavity cancer cells. Phytother Res. 2010;24 Suppl 1:S77-82.
425
[30] Haghiac M, Walle T. Quercetin induces necrosis and apoptosis in SCC-9 oral cancer
426
cells. Nutr Cancer. 2005;53:220-31.
427
[31] Kaestner KH, Knochel W, Martinez DE. Unified nomenclature for the winged
428
helix/forkhead transcription factors. Genes Dev. 2000;14:142-6.
429
[32] Greer EL, Brunet A. FOXO transcription factors at the interface between longevity and
430
tumor suppression. Oncogene. 2005;24:7410-25.
431
[33] Zhao Y, Wang Y, Zhu WG. Applications of post-translational modifications of FoxO
432
family proteins in biological functions. J Mol Cell Biol. 2011;3:276-282.
433
[34] Essaghir A, Dif N, Marbehant CY, Coffer PJ, Demoulin JB. The transcription of FOXO
434
genes is stimulated by FOXO3 and repressed by growth factors. J Biol Chem.
435
2009;284:10334-42.
436
[35] Maiese K, Chong ZZ, Shang YC. OutFOXOing disease and disability: the therapeutic
437
potential of targeting FoxO proteins. Trends Mol Med. 2008;14:219-27.
25
438
[36] Kim SY, Yoon J, Ko YS, Chang MS, Park JW, Lee HE, et al. Constitutive
439
phosphorylation of the FOXO1 transcription factor in gastric cancer cells correlates with
440
microvessel area and the expressions of angiogenesis-related molecules. BMC Cancer.
441
2011;11:264.
442
[37] Katayama K, Nakamura A, Sugimoto Y, Tsuruo T, Fujita N. FOXO transcription
443
factor-dependent p15(INK4b) and p19(INK4d) expression. Oncogene. 2008;27:1677-86.
444
[38] Valis K, Prochazka L, Boura E, Chladova J, Obsil T, Rohlena J, et al. Hippo/Mst1
445
stimulates transcription of the proapoptotic mediator NOXA in a FoxO1-dependent manner.
446
Cancer Res. 2011;71:946-54.
447
[39] Suhara T, Kim HS, Kirshenbaum LA, Walsh K. Suppression of Akt signaling induces
448
Fas ligand expression: involvement of caspase and Jun kinase activation in Akt-mediated Fas
449
ligand regulation. Mol Cell Biol. 2002;22:680-91.
450
[40] Zhao Y, Yang J, Liao W, Liu X, Zhang H, Wang S, et al. Cytosolic FoxO1 is essential for
451
the induction of autophagy and tumour suppressor activity. Nat Cell Biol. 2010;12:665-75.
452
[41] Zhang H, Pan Y, Zheng L, Choe C, Lindgren B, Jensen ED, et al. FOXO1 inhibits Runx2
453
transcriptional activity and prostate cancer cell migration and invasion. Cancer Res.
454
2011;71:3257-67.
455
[42] Boreddy SR, Pramanik KC, Srivastava SK. Pancreatic tumor suppression by benzyl
456
isothiocyanate is associated with inhibition of PI3K/AKT/FOXO pathway. Clin Cancer Res.
26
457
2011;17:1784-95.
458
[43] Chakrabarty A, Sanchez V, Kuba MG, Rinehart C, Arteaga CL. Feedback upregulation of
459
HER3 (ErbB3) expression and activity attenuates antitumor effect of PI3K inhibitors. Proc
460
Natl Acad Sci U S A. 2011.
461
[44] Chen Q, Ganapathy S, Singh KP, Shankar S, Srivastava RK. Resveratrol induces growth
462
arrest and apoptosis through activation of FOXO transcription factors in prostate cancer cells.
463
PLoS One. 2010;5:e15288.
464
[45] Roy SK, Chen Q, Fu J, Shankar S, Srivastava RK. Resveratrol Inhibits Growth of
465
Orthotopic Pancreatic Tumors through Activation of FOXO Transcription Factors. PLoS One.
466
2011;6:e25166.
467
[46] Wu Y, Shang X, Sarkissyan M, Slamon D, Vadgama JV. FOXO1A is a target for
468
HER2-overexpressing breast tumors. Cancer Res. 2010;70:5475-85.
469
[47] Hseu YC, Lee MS, Wu CR, Cho HJ, Lin KY, Lai GH, et al. The chalcone flavokawain B
470
induces G2/M cell-cycle arrest and apoptosis in human oral carcinoma HSC-3 cells through
471
the intracellular ROS generation and downregulation of the Akt/p38 MAPK signaling
472
pathway. J Agric Food Chem. 2012;60:2385-97.
473
[48] Pagano M, Pepperkok R, Verde F, Ansorge W, Draetta G. Cyclin A is required at two
474
points in the human cell cycle. Embo J. 1992;11:961-71.
475
[49] Foijer F, te Riele H. Check, double check: the G2 barrier to cancer. Cell Cycle.
27
476
2006;5:831-6.
477
[50] Ravid T, Sweeney C, Gee P, Carraway KL, 3rd, Goldkorn T. Epidermal growth factor
478
receptor activation under oxidative stress fails to promote c-Cbl mediated down-regulation. J
479
Biol Chem. 2002;277:31214-9.
480
[51] Zhao Y, Wang Y, Zhu WG. Applications of post-translational modifications of FoxO
481
family proteins in biological functions. J Mol Cell Biol. 2011;3:276-82.
482
[52] Brunet A, Bonni A, Zigmond MJ, Lin MZ, Juo P, Hu LS, et al. Akt promotes cell
483
survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell.
484
1999;96:857-68.
485
[53] Kozaki K, Imoto I, Pimkhaokham A, Hasegawa S, Tsuda H, Omura K, et al. PIK3CA
486
mutation is an oncogenic aberration at advanced stages of oral squamous cell carcinoma.
487
Cancer Sci. 2006;97:1351-8.
488
[54] Seoane J, Le HV, Shen L, Anderson SA, Massague J. Integration of Smad and forkhead
489
pathways in the control of neuroepithelial and glioblastoma cell proliferation. Cell.
490
2004;117:211-23.
491
[55] Yamaza H, Komatsu T, Wakita S, Kijogi C, Park S, Hayashi H, et al. FoxO1 is involved
492
in the antineoplastic effect of calorie restriction. Aging Cell. 2010;9:372-82.
493
[56] Sakai E, Tsuchida N. Most human squamous cell carcinomas in the oral cavity contain
494
mutated p53 tumor-suppressor genes. Oncogene. 1992;7:927-33.
28
495
Figure legends
496
497
Fig. 1. Quercetin inhibits cell growth in human oral cancer cell lines. (A). HGF (control) and
498
two human oral cancer cell lines, TW206 and HSC-3 cells, were treated with quercetin (0-50
499
μM) dissolved in DMSO for 24 h. The final DMSO concentration in the culture media was no
500
more than 0.3 %. Viable cell numbers were counted (*, p < 0.05 as compared with control, 0
501
μM). The cell number of each quercetin-untreated group was set as 1. (B). Effect of quercetin
502
on colony formation. HSC-3 cells were incubated with the indicated concentration of
503
quercetin for 7 days. Cell colonies were stained with crystal violet and quantified. Bars with
504
different letters indicate statistically significant difference from each other (p<0.05).
505
506
Fig. 2. Quercetin induces cell cycle arrest and apoptosis in human oral cancer cells. (A).
507
Effect of quercetin on cell cycle progression. Both HSC-3 and TW206 cells were treated with
508
quercetin (20 μM) for 24 h and then undergone flow cytometric analysis for cell cycle
509
distribution (*, p<0.05). (B). Effect of quercetin on cell cycle-related genes. HSC-3 cells were
510
treated with quercetin (0-20 μM) for 24 h, and the protein lysates were analyzed by Western
511
blotting with the indicated antibodies where α-tubulin served as a loading control. (C-E).
512
Effect of quercetin on apoptosis. Both HSC-3 and TW206 cells were subjected to annexin
513
V-FITC and PI staining after 24 h treatment of quercetin (0-30 μM). Annexin V-positive cells
29
514
were analyzed and quantified using a FACScan flow cytometer (*, p<0.05 as compared with
515
Q0, quercetin 0 μM) (C). HSC-3 cells after 24 h treatment of quercetin were stained with
516
DAPI (D) or analyzed for apoptosis-related gene expression by Western blotting with the
517
indicated antibodies where GAPDH served as a loading control (E).
518
519
Fig. 3.
520
HSC-3, TW206, and HGF cell lines were detected by Western blotting, where p38 served as
521
a loading control. Quercetin suppresses phosphorylation of EGFR and its downstream
522
effector, Akt, in time- (B) and dose-dependent (C) manners. HSC-3 cell lysates from each
523
respective treatment were extracted and analyzed by Western blotting with the indicated
524
antibodies. The experiments repeated three times with consistent results.
Effect of quercetin on EGFR/Akt signaling. (A) The endogenous levels of EGFR in
525
526
Fig. 4.
527
time- (A) and dose-dependent (B) manners. HSC-3 cell lysates from each respective
528
treatment were extracted and analyzed by Western blotting with antibodies against FOXO1,
529
FOXO3a, or α-tubulin. (C-D). Effect of quercetin on subcellular localization of FOXO1. (C).
530
HSC-3 cells were treated with quercetin (0 or 10 μM) for 24 h and then undergone
531
cytoplasmic and nuclear extraction. Protein lysates were subjected to Western blotting
532
analysis of p-FOXO1 and FOXO1 expression with α-tubulin and TBP as loading controls for
Quercetin induces FOXO1 activation. Quercetin induces FOXO1 expression in
30
533
cytoplasmic and nuclear portions, respectively. (D). HSC-3 cells were seeded onto a chamber
534
slide, treated with quercetin for 24 h, and then fixed and stained with an antibody against
535
FOXO1 (green). Nuclei were counterstained with DAPI (blue). (E). Quercetin induces
536
FOXO1 transactivation. HSC-3 cells, in triplicate, were cotransfected with
537
pGL3-FHRE-luciferase (0.25 μg) and β-galactosidase (0.05 μg) plasmids, in the presence or
538
absence of FOXO1 siRNA, for 24 h, and then treated with quercetin (0, 20 μM) for another
539
24 h. The luciferase activity was normalized to β-galactosidase for transfection efficiency and
540
presented as -fold activation. Each bar represents the mean + S.D. (*, p<0.05). (F). Ly294002
541
(LY), a PI3K inhibitor, mimics quercetin’s effect on FOXO1. HSC-3 cells were treated with
542
LY at the indicated time points, and cell lysates were then analyzed by Western blotting for
543
p-Akt and FOXO1 expression.
544
545
Fig. 5.
546
transfected with either scramble siRNA (Control) or FOXO1 siRNA were treated with
547
quercetin as indicated for 24 h (A) or 7 days (B) for the measurement of viable cells and
548
colony numbers, respectively (*, p<0.05 vs. control). (C). Effect of FOXO1 knockdown on
549
quercetin-mediated cell cycle arrest. Both HSC-3 and TW206 cells with FOXO1 knockdown
550
were treated with quercetin for 24 h and then analyzed for cell cycle distribution by a
551
FACScan flow cytometer. (D). Effect of FOXO1 knockdown on quercetin-induced p21
Role of FOXO1 in quercetin-mediated growth suppression. (A-B). HSC-3 cells
31
552
expression. HSC-3 cells were transfected with either FOXO1 or scramble siRNA for 24 h
553
followed by quercetin treatment for another 24 h. Cell lysates were analyzed for FOXO1 and
554
p21 expression by Western blotting. (E). FOXO1 mimics quercetin’s effect on p21. HSC-3
555
cells were transiently transfected with a Flag-FOXO1 expression construct, and cells lysates
556
were analyzed by Western blotting with anti-flag and anti-p21 antibodies.
557
558
Fig. 6. Role of FOXO1 in quercetin-induced apoptosis. HSC-3 cells undergone FOXO1
559
siRNA and/or 24 h of quercetin treatment were stained with annexin V-FITC and PI, analyzed
560
by a FACScan flow cytometer (*, p<0.05) (A), and subjected to Western blotting analysis
561
with the indicated antibodies (B).
562
563
564
Fig. 7.
565
mice were established, with or without quercetin treatment, for 35 days. (A) Tumor size was
566
recorded every other day (*, p < 0.05 as compared with control). (B) At the end of the study,
567
tumors were excised and weighed. The picture represents the average tumor growth in either
568
group (n=6). A two-tailed, unpaired t test was used to assess the differences of tumor weight
569
between the two groups (*, p<0.05). (C). Quercetin induces FOXO1 expression in HSC-3
570
implanted xenograft mice. At the end of the experiment, tumors were excised and
Quercetin suppresses tumor growth in vivo. (A-B). HSC-3 implanted xenograft
32
571
homogenized for protein extraction. Protein lysates were then subjected to Western blotting
572
analysis for FOXO1 expression.
573
574
Fig. 8.
575
carcinomas. Quercetin efficiently inhibits cell growth by targeting EGFR/Akt signaling in
576
EGFR-overexpressing oral cancer. The resulting FOXO1 activation leads to elevated
577
expression of p21 and FasL, and, subsequently, growth arrest in G2 phase and apoptosis.
Role of FOXO1 in quercetin-mediated growth suppression of oral squamous
33
Download