Journal Pre-proofs AGS-30, an andrographolide derivative, suppresses tumor angiogenesis and growth in vitro and in vivo Jingjing Li, Feng Li, Fan Tang, Jinming Zhang, Renkai Li, Dekuan Sheng, Simon Ming-Yuen Lee, Guo-Chun Zhou, George Pak-Heng Leung PII: DOI: Reference:
S0006-2952(19)30393-4 https://doi.org/10.1016/j.bcp.2019.113694 BCP 113694
To appear in:
Biochemical Pharmacology
Received Date: Accepted Date:
30 September 2019 4 November 2019
Please cite this article as: J. Li, F. Li, F. Tang, J. Zhang, R. Li, D. Sheng, S.M-Y. Lee, G-C. Zhou, G.P-H. Leung, AGS-30, an andrographolide derivative, suppresses tumor angiogenesis and growth in vitro and in vivo, Biochemical Pharmacology (2019), doi: https://doi.org/10.1016/j.bcp.2019.113694
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 Published by Elsevier Inc.
1
AGS-30, an andrographolide derivative, suppresses tumor angiogenesis and growth in
2
vitro and in vivo
3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39
Jingjing Lia,1, Feng Lib,1, Fan Tangc,1, Jinming Zhangd, Renkai Lia, Dekuan Shengb, Simon Ming-Yuen Leec,*,Guo-Chun Zhoub,*, George Pak-Heng Leunga,* aDepartment
of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong,
China; bSchool
of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China; Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China; dCollege of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China. cState
1These
authors contributed equally to the paper.
*Corresponding author Dr. Simon Ming-Yuen Lee, N22 Research Building, State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, China Tel.: +853 88224695 E-mail address:
[email protected]
Dr. Guo-Chun Zhou, NO.30, South Puzhu Road, Pukou District, School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, China. Tel.: +86-15850780314 E-mail address:
[email protected]
Dr. George Pak-Heng Leung, 2/F, 21 Sassoon Road, Li Ka Shing Faculty of Medicine, Laboratory Block, Faculty of Medicine Buliding, Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong SAR, China Tel.: +852 39179024 E-mail address:
[email protected]
Key words: andrographolide derivative, anti-angiogenic, colon cancer, VEGF
40 1
41
Abstract
42
Poor bioavailability and limited efficacy are challenges associated with using
43
andrographolide as a therapeutic agent. We recently synthesized AGS-30, a new
44
andrographolide derivative, in our laboratory. In this study we investigated the potential
45
anti-tumor effect of AGS-30 and the underlying mechanisms, particularly those related to
46
angiogenesis. Results from our in vitro experiments showed that AGS-30 exerted
47
anti-angiogenic effects by inhibiting endothelial cell proliferation, migration, invasion, and
48
tube formation. Phosphorylation and activation of angiogenesis-related signaling molecules
49
(e.g., vascular endothelial growth factor [VEGF] receptor 2, mitogen-activated protein kinase
50
kinase 1/2, extracellular signal-regulated kinase 1/2, mechanistic target of rapamycin
51
[mTOR], protein kinase B [Akt], and p38) were markedly reduced by AGS-30. Meanwhile,
52
AGS-30 potently inhibited cell proliferation and phosphorylation of cell survival-related
53
proteins (e.g., Akt, mTOR, and ERK1/2) and decreased the expression of VEGF in HT-29
54
colon cancer cells. AGS-30 blocked microvessel sprouting in a rat aortic ring model and
55
blood vessel formation in zebrafish embryos and a mouse Matrigel plug model. Additionally,
56
AGS-30 suppressed tumor growth and angiogenesis in HT-29 colon cancer cell xenografts in
57
nude mice. These effects were not observed when same concentration of andrographolide, the
58
parent compound of AGS-30, was used. Thus, AGS-30 exerted a strong antitumor effect by
59
inhibiting tumor cell growth and angiogenesis and is a candidate compound for the treatment
60
of cancer.
61 62 2
63
1. Introduction
64
Natural products have great potential for drug development owing to their diversity,
65
complexity, and biocompatibility. The basic chemical structures of natural products can be
66
modified and can serve as a basis for the development of novel agents with improved
67
biological properties and safety profiles [1]. It has been reported that over 50% of approved
68
drugs in clinical use either originate or are derived from natural products [2] and as of the end
69
of 2014, nearly half of the 175 small-molecule compounds approved for the treatment of
70
cancer were natural product-based [3]. Most of the drugs that succeeded in clinical trials (e.g.,
71
sorafenib, imatinib, and gefitinib [4]) were chemically modified from lead compounds.
72 73
Angiogenesis is a basic biological process involving the sprouting and growth of new blood
74
vessels from the pre-existing vascular primordium [5] and plays a key role not only in the
75
regulation of various physiological activities (e.g., embryonic development, wound healing,
76
female reproductive cycle, etc.) but also in tumor growth and metastasis [6, 7]. Tumors that
77
grow beyond a size of 2 mm3 are dependent on oxygen and nutrients supplied by surrounding
78
newly formed blood vessels [8]. Anti-angiogenic therapy involves starving tumor cells by
79
targeting endothelial cells and blocking angiogenesis [9]. Most currently used anti-angiogenic
80
drugs are small-molecule compounds, which have advantages over monoclonal antibodies.
81
For instance, small molecules agents are less expensive and more convenient to administer
82
[10]. However, the development of drug resistance and limited efficacy limit their widespread
83
clinical application [11]. There is therefore a need for novel types of small molecule with
84
anti-angiogenic activity. 3
85
Diterpenoids are a class of bioactive natural compounds that are found in many medicinal
86
herbs and have great potential in the treatment of diseases involving excessive angiogenesis
87
such as cancer. For instance, oridonin, a tetracycline diterpenoid isolated from Rabdosia
88
rubenscens, was shown to induce apoptosis and inhibits the migration and invasion of highly
89
metastatic human breast cancer cells [12]. Oridonin suppressed tumor growth and metastasis
90
by blocking tumor angiogenesis via downregulation of vascular endothelial growth factor
91
(VEGF)-induced Jagged/Notch signaling [13]. We recently showed that oridonin not only
92
synergistically enhances the anti-tumor efficacy of doxorubicin against aggressive breast
93
cancer
94
doxorubicin-induced cardiotoxity in mice [14]. Another example of a natural anti-angiogenic
95
compound is triptolide, a diterpenoid epoxide isolated from Tripterygium wilfordii Hook F.
96
Triptolide was shown to inhibit angiogenesis and invasion of human anaplastic thyroid
97
carcinoma cells by blocking nuclear factor-κB signaling [15], and induced apoptosis while
98
suppressing the growth and angiogenesis of human pancreatic cancer cells via negative
99
regulation of cyclooxygenase 2 and VEGF expression [16]. These reports demonstrate the
100
through
proapoptotic
and
anti-angiogenic
signaling,
but
also
reduced
potential of diterpenoids as anti-cancer agents.
101 102
Andrographolide (Andro; Scheme 1) is a bioactive labdane diterpenoid. It was previously
103
reported that the Andro content of dried whole Andrographalis paniculata (Burn. f.) plant is
104
as high as 4%, suggesting that Andro is the most abundant diterpenoid in this medicinal plant
105
[17]. Andro is known to have anticancer [18, 19], anti-angiogenic [20, 21], anti-inflammatory
106
[22], antidiabetic [23], and neuroprotective [24] activities. Andro and A. paniculate extracts 4
107
inhibit tumor angiogenesis by suppressing pro-angiogenic molecules such as VEGF and nitric
108
oxide and enhancing the expression of anti-angiogenic factors such as interleukin 2 and
109
TIMP metallopeptidase inhibitor 1 [25]. Moreover, Andro can block tumor angiogenesis by
110
preventing VEGF-A-induced activation of VEGFR2 and downstream mitogen-activated
111
protein kinase (MAPK) signaling [20, 26]. However, despite the therapeutic potential
112
demonstrated by Andro, its development and clinical application are a challenge owing to its
113
poor water solubility, low potency, and limited therapeutic efficacy [27].
114 115
AGS-30 is a recently synthesized Andro derivative [28]. In this study, we investigated the
116
anticancer and anti-angiogenic effects of AGS-30 in vitro and in vivo and the underlying
117
mechanisms of action. Moreover, the present study also indicated that modification of the
118
C14 position in the chemical structure of Andro significantly enhanced its anticancer and
119
anti-angiogenic effects. Our findings provide a basis for the future development of Andro
120
derivatives as novel angiogenesis inhibitors for the treatment of cancer.
121
5
123
2. Materials and methods
124
2.1. Ethical statement
125
Ethics approval for animal experiments was obtained from the University of Macau and
126
Chengdu University of Traditional Chinese Medicine. All animal experiments were
127
conducted in compliance with Instructional Animal Care and Use Committee (IACUC).
128 129
2.2. Chemicals and reagents
130
AGS-30 was synthesized from Andro (Scheme 1) [28]. Andro, dimethyl sulfoxide (DMSO),
131
heparin, gelatin, collagen, protease, endothelial cell growth supplement (ECGS), Trypan blue,
132
paraformaldehyde, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide (MTT), and
133
Drabkin Reagent Kit 525 were purchased from Sigma-Aldrich (St. Louis, MO, USA).
134
Dulbecco’s Modified Eagle’s Medium (DMEM), fetal bovine serum (FBS), Hoechst 33342
135
dye, bicinchoninic acid assay kit (BCA), phenylmethylsulfonyl fluoride (PMSF), protease
136
inhibitor cocktail, phosphate-buffered saline (PBS), penicillin/streptomycin, and 0.25% (w/v)
137
trypsin containing 1 mM EDTA were from Invitrogen (Carlsbad, CA, USA). Growth
138
factor-reduced Matrigel was obtained from BD Biosciences (Franklin Lakes, NJ, USA).
139
VEGF-A was from R&D Systems (Minneapolis, MN, USA). The lactate dehydrogenase
140
(LDH) cytotoxicity assay kit was from Cayman Chemical (Ann Arbor, MI, USA). VEGF
141
receptor (VEGFR) tyrosine kinase inhibitor II (VRI) was from CalBiochem (San Diego, CA,
142
USA). A primary antibody against VEGF-A and a human VEGF enzyme-linked
143
immunosorbent assay (ELISA) kit were obtained from Abcam (Cambridge, MA, USA).
144
Other antibodies were purchased from Cell Signaling Technology (Danvers, MA, USA). All 6
145
chemicals were dissolved in appropriate solvent and stored at -20°C until use.
146 147
2.3. Cell culture
148
Human umbilical vein endothelial cells (HUVECs) and HT-29 human colon adenocarcinoma
149
cells were obtained from American Type Culture Collection (Manassas, VA, USA).
150
HUVECs were cultured in F-12K complete medium supplemented with 10 mM L-glutamine,
151
1 μg/mL hydrocortisone hemisuccinate, 50 μg/mL ascorbic acid, 100 µg/mL heparin, 30
152
µg/mL endothelial cell growth supplement, 10% heat-inactivated FBS, and 1%
153
penicillin/streptomycin. Cells from early passages (passages 38) were used in experiments.
154
HT-29 human colon cancer cells were cultured in DMEM supplemented with 10%
155
heat-inactivated FBS and 1% penicillin/streptomycin. Cells were cultured in tissue flasks
156
precoated with 0.1% gelatin at 37°C in a humidified atmosphere of 5% CO2.
157 158
2.4. Cell viability assay
159
HUVECs and HT-29 cells were seeded in 96-well plates at a density of 8 × 103 cells/well and
160
cultured for 24 h until the cells had attached. The cells were then treated with various
161
concentrations (020 µM) of Andro and AGS-30 in low-serum medium (containing 0.5%
162
FBS) for 24 or 48 h. Cell viability was evaluated with the MTT assay according to the
163
manufacturer’s protocol. Briefly, the medium was discarded and the cells were incubated
164
in MTT solution (at a final concentration of 0.5 mg/mL) for 4 h at 37°C. DMSO was then
165
added to lyse the cells and dissolve the violet formazan crystals that formed inside the
166
cells. Absorbance at 570 nm was measured using a SpectraMax M5 Multi-Mode Microplate 7
167
Reader (Molecular Devices, Sunnyvale, CA, USA).
168 169
2.5. Cell proliferation assay
170
HUVECs were seeded in a 48-well plate at a density of 3 × 104 cells/well and cultured for 24
171
h until they had attached; they were then cultured overnight in low-serum medium to induce
172
quiescence. The cells were treated with various concentrations of Andro and AGS-30 in
173
low-serum medium containing VEGF (20 ng/mL) for 48 h and then stained using Trypan
174
blue and observed under a microscope. Blue cells were considered non-viable.
175 176
2.6. Lactate dehydrogenase (LDH) assay
177
Cellular injury was assessed by measuring the activity of LDH released into the culture
178
medium upon cellular damage using a commercial kit according to the manufacturer’s
179
instructions. Absorbance at 490 nm was measured on a microplate reader.
180 181
2.7. Transwell migration and invasion assays
182
Cell migration and invasion assays were performed using HUVECs as previously described
183
[29]. Transwell inserts (8-μm pores) were obtained from BD Biosciences (Franklin Lakes, NJ,
184
USA). For the cell migration assay, the upper and lower sides of the Transwell membrane
185
were pre-coated with 0.1% collagen. In the invasion assay, the upper and lower sides of the
186
membrane were pre-coated with 100 μL Matrigel (20% in blank medium). HUVECs were
187
seeded in the Transwells at a density of 5 × 104 cells/well and cultured in low-serum medium
188
containing VEGF (20 ng/mL) and different concentrations of Andro and AGS-30. After 8
189
incubation for 24 h at 37°C, cells on the upper surface of the Transwell membrane were
190
removed using cotton swabs. The membranes were fixed with 4% paraformaldehyde for 15
191
min and then stained with Hoechst 33342 (10 μg/mL) for 15 min and mounted on microscope
192
slides, and images were captured on a fluorescence inverted microscope (Axiovert 200; Carl
193
Zeiss, Oberkochen, Germany) with a charge-coupled device camera (AxioCam HRC; Carl
194
Zeiss). Cell migration and invasion were quantified by counting the number of cells per insert
195
using ImageJ software (National Institutes of Health, Bethesda, MD, USA).
196 197
2.8. Tube formation assay
198
To evaluate angiogenic potential, 15-well microslides (ibidi, Fitchburg, WI, USA) were
199
coated with Matrigel (50% in blank medium) and incubated for 30 min at 37°C to induce
200
polymerization. HUVECs (2 × 105 cells/mL) were resuspended in low-serum medium with
201
different concentrations of Andro and AGS-30 in the presence of 20 ng/mL VEGF, and then
202
seeded on the Matrigel-coated slides. After incubation for 46 h, tube-like structures formed
203
that were connected at both ends, which were defined as endothelial cords. Images were
204
acquired before and after drug treatment on an inverted microscope. Tube formation was
205
quantified by counting the number of branch points in three randomly selected fields of view.
206 207
2.9. Western blot analysis
208
Total protein was extracted from cells using lysis buffer containing 1% PMSF and 1%
209
protease inhibitor. Lysates were centrifuged at 12,500 × g for 20 min at 4°C and the
210
supernatant was collected. Protein concentration was determined with the bicinchoninic acid 9
211
assay. Equal amounts of protein were resolved by sodium dodecyl sulfate–polyacrylamide gel
212
electrophoresis and transferred to a polyvinylidene difluoride membrane (Bio-Rad
213
Laboratories, Hercules, CA, USA) that was blocked with 5% non-fat milk in Tris-buffered
214
saline containing 0.1% Tween-20 (TBST) for 1 h. The membrane was then incubated
215
overnight at 4°C with primary antibodies against VEGF-A, VEGFR2, phosphorylated
216
(p-)VEGFR2 (Tyr1175), mechanistic target of rapamycin (mTOR), p-mTOR (Ser2448),
217
protein kinase B (Akt), p-Akt (Ser473), extracellular signal-regulated kinase (ERK)1/2,
218
p-ERK1/2 (Thr202/Tyr204), MAPK kinase (MEK)1/2, p-MEK1/2 (Ser217/221), p38, p-p38
219
(Thr180/Tyr182), and glyceraldehyde 3-phosphate dehydrogenase (GAPDH). All the primary
220
antibodies were diluted with TBST buffer at a ratio of 1:1000. After washing with PBS, the
221
membrane was incubated with horseradish peroxidase-conjugated secondary antibodies
222
(1:2000 dilution) for 1 h at room temperature. After multiple washes with PBS, proteins were
223
visualized by enhanced chemiluminescence. Images of protein bands were captured and
224
densitometric measurements of signal intensity were performed using the Molecular Imager
225
ChemiDoc XRS system (Bio-Rad Laboratories, Hercules, CA, USA).
226 227
2.10.
ELISA
228
HT-29 colon cancer cells were treated with AGS-30 (0–5 μM) for 48 h, and the levels of
229
secreted VEGF-A in the culture medium were measured and quantified using a commercial
230
ELISA kit according to manufacturer’s instructions.
231 232
2.11.
Zebrafish maintenance and embryo handling 10
233
The Tg(fli1:EGFP) transgenic zebrafish line expressing enhanced green fluorescent protein
234
(EGFP) in endothelial cells was provided by the Zebrafish Information Network (Eugene, OR,
235
USA). Wild-type zebrafish were purchased from a local pet store. The zebrafish were
236
maintained in a controlled environment at 28.5°C on a 14:10-h light/dark cycle and were fed
237
twice a day with brine shrimp and occasionally with general tropical fish food. Embryos were
238
generated through natural mating and reared in embryo medium at 28.5°C. Dead, unfertilized,
239
or abnormally shaped embryos were removed at 4 h post-fertilization (hpf).
240 241
2.12.
Morphological observation of zebrafish embryos
242
Morphological observation of zebrafish embryos was performed as previously described [30].
243
At 24 hpf, Tg(fli-1:EGFP) embryos (12 per group) were distributed in 12-well plates. The
244
groups were treated with different concentrations of AGS-30 (3–30 µM) or Andro (300 µM)
245
dissolved in embryo medium for 24 h. Embryos treated with DMSO (0.1%) served as the
246
control, and those treated with 250 ng/mL VRI were the positive control. The embryos were
247
observed for morphological changes and images were captured with a spinning disk confocal
248
microscope system (Olympus, Tokyo, Japan). Intact and defective intersegmental vessels
249
(ISVs) in each zebrafish embryo were counted as previously described [27].
250 251
2.13.
Rat aortic ring angiogenesis assay
252
A 48-well plate was precoated with 100 µL Matrigel per well and polymerized at 37°C for 30
253
min. Aortic rings were isolated from 6- to 8-week-old male Sprague-Dawley rats and cut into
254
1.5-mm pieces that were rinsed at least three times in cold PBS and then transferred to a well 11
255
and overlaid with 100 µl of Matrigel for sealing. Serum-free culture medium containing
256
VEGF (100 ng/ml), with or without Andro or AGS-30 (5 μM), was added to the wells. The
257
medium was changed every 2 days. After 10 days, microvessel growth was photographed on
258
an inverted microscope and the number of branching sites was quantified with ImageJ
259
software.
260 261
2.14.
Mouse Matrigel plug angiogenesis assay
262
Growth factor-reduced Matrigel (0.5 mL) containing heparin (100 U) and VEGF (250 ng)
263
with or without Andro and AGS-30 was subcutaneously injected into the ventral area of
264
8-week-old male C57BL/6 mice (n = 5). After 14 days, mice were sacrificed and the Matrigel
265
plugs were removed. To quantify the formation of functional blood vessels, the hemoglobin
266
concentration in the plug was measured using Drabkin Reagent Kit 525.
267 268
2.15.
Inhibition of tumor growth in vivo
269
The subcutaneous dorsum of male nude mice (4-weeks-old; 16 g) was inoculated with HT-29
270
human colon cancer cells (1 × 106 cells/mouse). Tumors were allowed to grow to
271
approximately 200 mm3, and the xenografted mice were then randomly assigned to one of
272
three groups (n = 5 each) that were intraperitoneally injected with saline, Andro (2 mg/kg), or
273
AGS-30 (2 mg/kg) once every 2 days for 20 days. Body weight and tumor volume ([width]2
274
× [length]/2) were recorded every 2 days. After treatment, mice were sacrificed by overdose
275
of barbiturates administered intraperitoneally and then photographed, primary tumors and
276
major organs were excised and weighted. Tumor tissue was fixed with 4% (v/v) 12
277
formaldehyde and cut into 6-μm sections. Tumors and major organs sections were stained
278
with hematoxylin and eosin (H&E) to examine tissue architecture. The sections were also
279
labeled with primary antibodies against VEGF, p-VEGFR2, and CD31 to evaluate
280
angiogenesis at the tumor site.
281 282
2.16.
Statistical analysis
283
Data are expressed as mean ± SD of at least three independent experiments and were
284
analyzed with Prism v.5.0 software (GraphPad, La Jolla, CA, USA). The difference between
285
groups was evaluated by one-way analysis of variance and P < 0.05 was considered
286
significant.
287 288 289
13
291
3. Results
292
3.1. Effects of AGS-30 on the viability of HUVECs and HT-29 cancer cells
293
Cell viability was evaluated with the MTT assay in HUVECs and HT-29 colon cancer cells
294
treated with various concentrations (0–20 μM) of Andro or AGS-30 for 24 or 48 h. The
295
half-maximal inhibitory concentration (IC50) of AGS-30 in HT-29 cancer cells after 24 and
296
48 h of treatment was 8.72 ± 3.35 and 4.13 ± 2.25 μM, respectively (Fig. 1A). IC50 values for
297
Andro were much higher at these time points (> 40 μM). Meanwhile, the viability of
298
HUVECs was decreased by treatment for 24 and 48 h with AGS-30 at concentrations greater
299
than 5 μM, but was unaffected by Andro in the concentration range of 0–20 μM (Fig. 1B). To
300
further evaluate the toxicity of Andro and AGS-30, we examined LDH levels in HUVECs
301
and observed no cellular damage upon treatment with 1.25–5 μM AGS-30 and 5–20 μM
302
Andro, respectively (Fig. 1C). These concentrations were therefore considered as safe for
303
HUVECs and were used in subsequent experiments evaluating angiogenesis.
304 305
3.2. Effects of AGS-30 on VEGF-induced proliferation
306
Endothelial cell proliferation plays a critical role in angiogenesis. We therefore examined the
307
effects of Andro and AGS-30 on endothelial cell proliferation by counting cell numbers.
308
HUVEC proliferation was increased by 38% in the presence of VEGF (20 ng/mL) (Fig. 1D).
309
This effect was reversed by AGS-30 and Andro in a concentration-dependent manner; the
310
lowest effective concentrations of AGS-30 and Andro were 2.5 and 20 μM, respectively.
311
These data demonstrated that AGS-30 more potently inhibited HUVEC proliferation than
312
Andro. 14
314
3.3. Effects of AGS-30 on VEGF-induced endothelial cell migration, invasion, and tube
315
formation
316
The effects of AGS-30 and Andro on endothelial cell migration and invasion were evaluated
317
with the Transwell assay. The migratory and invasive capacities of HUVECs were increased
318
by 145% and 210%, respectively, in the presence of VEGF (20 ng/mL) (Fig. 2A, B). This
319
effect was reversed by AGS-30 in a concentration-dependent manner, with the lowest
320
effective concentration at 2.5 μM (Fig. 2D, E). However, VEGF-induced endothelial cell
321
migration and invasion was unaffected by Andro (5 μM).
322 323
We next investigated the effect of AGS-30 and Andro on the construction of cord-like
324
networks by HUVECs. After treatment for 6 h, endothelial cells in the control and 20 ng/mL
325
VEGF-treated groups formed capillary-like structures and cord-like networks (Fig. 2C).
326
However, VEGF-induced tube formation was abolished by AGS-30 in a dose-dependent
327
manner, whereas Andro (5 μM) had no effect (Fig. 2F). These data suggested that AGS-30
328
had more potent anti-angiogenic activity than Andro in vitro.
329 330
3.4. Effect of AGS-30 on VEGFR2-mediated angiogenesis signaling
331
To investigate the anti-angiogenic mechanisms of AGS-30, we examined the effect of
332
AGS-30 on the expression levels of key proteins involved in VEGFR2-mediated
333
angiogenesis signaling by western blotting. The phosphorylation of VEGFR2, mTOR, Akt,
334
MEK1/2, ERK1/2, and p38 in HUVECs was increased by VEGF (50 ng/mL) (Fig. 3A).
335
However, this was abrogated by AGS-30 in a concentration-dependent manner (1.25–5 μM 15
336
(Fig. 3C–F). In addition, the VEGF-induced phosphorylation of VEGFR2 and p38 was
337
inhibited by AGS-30 at concentrations as low as 1.25 μM (Fig. 3B, G). However, overall
338
expression of these proteins was unaltered by AGS-30.
339 340
3.5. Effects of AGS-30 on cancer cell proliferation and survival pathways
341
To investigate the mechanism underlying the effect of AGS-30 on cancer cell proliferation
342
and survival, we examined the expression levels of proteins in cancer cell proliferation and
343
survival pathways. The total amounts of these proteins were unaffected by AGS-30. However,
344
mTOR
345
concentration-dependent manner 48 h after treatment with AGS-30 (1.25–5 μM) (Fig. 4A).
346
Meanwhile, p-ERK1/2 and p-Akt were downregulated by AGS-30 (5 μM), although the
347
levels were unaltered by AGS-30 concentrations of 1.25 and 2.5 μM (Fig. 4B, D).
phosphorylation
in
HT-29
colon
cancer
cells
was
reduced
in
a
348 349
3.6. Effect of AGS-30 on VEGF expression in HT-29 cells
350
VEGF plays a critical role not only in angiogenesis but also in tumor growth. To clarify the
351
effect of AGS-30 on VEGF expression in HT-29 cells, we assessed intra- and extracellular
352
VEGF levels in HT-29 cells by western blotting and ELISA, respectively. VEGF levels in
353
HT-29 colon cancer cell lysates and culture medium were decreased by AGS-30 treatment in
354
a concentration-dependent manner (Fig. 4E, F).
355 356
3.7. Effect of AGS-30 on blood vessel formation in zebrafish model
357
To investigate the anti-angiogenic activity of AGS-30 in vivo, we examined the effect of 16
358
AGS-30 on blood vessel formation in a transgenic zebrafish model. Zebrafish embryos at 24
359
h hpf were treated with vehicle (0.1% DMSO), the positive control VRI (250 ng/mL), Andro
360
(300 μM), or AGS-30 (3–30 μM) for 24 h. Intact ISVs were observed in the control and
361
Andro groups (Fig. 5A, C), whereas defective ISVs were present in the VRI and
362
AGS-30-treated groups (Fig. 5B). ISV formation in the AGS-30 group was markedly
363
inhibited even at a concentration as low as 3 μM (Fig. 5D–F). Quantitative analysis revealed
364
that the number of intact ISVs decreased with increasing AGS-30 concentration (Fig. 5M).
365
We analyzed the anti-angiogenic phenotype of embryos (i.e., with incomplete ISVs) in each
366
group and found that in the VRI-treated group, the growth of intact ISVs was reduced, with
367
nearly 90% showing an anti-angiogenic phenotype. On the other hand, the fraction of ISVs
368
exhibiting the phenotype in the AGS-30-treated group was increased from 42% to 72% when
369
the concentration was increased from 3 to 30 μM (Fig. 5N). Blood vessel formation in the
370
zebrafish model was unaffected by Andro, even a very high concentration (300 μM). These
371
results confirmed that AGS-30 had more potent anti-angiogenic activity than Andro in vivo.
372 373
3.8. Effects of AGS-30 on VEGF-induced angiogenesis ex vivo and in vivo
374
To investigate whether AGS-30 blocks angiogenesis ex vivo, we evaluated the effect of
375
AGS-30 on microvessels sprouting in a rat aortic ring model. Microvessels sprouting was
376
induced by treatment with VEGF (100 ng/mL) for 10 days, resulting in the formation of a
377
complete network of microvessels surrounding aortic rings. This was abolished in the
378
presence of 5 μM AGS-30 (Fig. 6B) but was unaffected by 5 μM Andro treatment.
379 17
380
The anti-angiogenic activity of AGS-30 in vivo was assessed using a mouse Matrigel plug
381
model. Matrigel plugs containing different compounds were implanted in mice; those
382
containing VEGF (250 ng) were dark red and filled with blood vessels after 14 days (Fig. 6C),
383
suggesting that abundant microvessels were formed in the plugs. In contrast, Matrigel plugs
384
containing VEGF and 5 μM AGS-30 were light red and nearly transparent, indicating that
385
fewer blood vessels had formed. To quantify functional blood vessel formation, we measured
386
hemoglobin levels in the Matrigel plugs and found that they were reduced by AGS-30 and
387
Andro by 74% and 36%, respectively, compared to the VEGF-treated group (Fig. 6D).
388 389
3.9. Effects of AGS-30 on tumor growth in a nude mouse model
390
Based on the in vitro and in vivo data suggesting that AGS-30 has anti-angiogenic activity,
391
we further investigated the anti-tumor efficacy of AGS-30 and Andro in nude mice bearing
392
HT-29 colon cell-derived tumors in which tumor growth is closely associated with
393
angiogenesis. Average tumor volume in the control group exceeded 1,300 mm3 after 20 days;
394
treatment with 2 mg/kg of AGS-30 and Andro reduced tumor weight by 52% and 21%,
395
respectively (Fig. 7B). The inhibitory effect of AGS-30 on tumor volume was greater than
396
that of Andro (Fig. 7C). However, neither AGS-30 nor Andro affected the body weight of
397
mice (Fig. 7D).
398 399
We performed a histological analysis in order to verify the mechanisms underlying the
400
anti-angiogenic and anti-tumor effects of AGS-30 in the mouse xenograft model. H&E
401
staining showed no necrosis in tumors of both control and Andro-treated mice, whereas large 18
402
areas of necrosis were observed in tumors of mice treated with AGS-30 (Fig. 8A). A
403
quantitative analysis showed that the necrotic area was increased nearly 2 fold in the
404
AGS-30-treated group compared to the control and Andro-treated groups (Fig. 8B). Blood
405
vessels in tumors were labeled with an antibody against CD31, an endothelial cell marker.
406
The immunohistochemical analysis revealed that VEGF expression was slightly reduced by
407
Andro, while a marked decreased was observed in microvessel density and VEGFR2
408
phosphorylation (Fig. 8A). A quantitative analysis showed that microvessel density and
409
VEGF and p-VEGFR2 levels were decreased by 82%, 77%, and 91%, respectively, in the
410
presence of AGS-30 (Fig. 8C–E).
411 412
Finally, we investigated the toxicity of AGS-30 and Andro in vivo by examining
413
morphological changes in the major organs. No significant damage was observed in the heart,
414
liver, spleen, lung, and kidney of mice treated with AGS-30 or Andro (Fig.9).
19
416
4. Discussion
417
VEGFR2 is an important drug target for angiogenesis inhibitors; to date, a large number of
418
drugs targeting VEGFR2 have been approved by the U.S. Food and Drug Administration
419
(FDA) including monoclonal antibodies, small molecule compounds, and microRNAs [31].
420
Sorafenib, a small molecule drug that acts on multiple kinases, was shown to inhibit tumor
421
angiogenesis by suppressing VEGFR-mediated signaling [32]. In the present study, we
422
showed that AGS-30, a 14β-(2'-chlorophenoxy) derivative of andrographolide, potently
423
inhibited tumor angiogenesis by blocking VEGFR2-induced endothelial cell proliferation,
424
migration, invasion, and tube formation. Previous studies have demonstrated that both
425
Akt/mTOR- and MEK/ERK-dependent signaling pathways regulate cell survival and
426
proliferation in endothelial and cancer cells [33-36], and that p38 MAPK regulates
427
angiogenesis by modulating endothelial cell permeability and migration [37, 38]. The present
428
study showed that AGS-30 decreased VEGF-induced phosphorylation of Akt, mTOR,
429
MEK1/2, ERK1/2, and p38 MAPK in endothelial cells. Sorafenib, a small molecule drug,
430
inhibited tumor growth and angiogenesis by blocking RAF/MEK/ERK-dependent signaling
431
in hepatocellular carcinoma [39]. Interestingly. we found that AGS-30 also inhibited the
432
activation of Akt, mTOR, and ERK1/2 in HT-29 cells. In addition to the blockade of
433
VEGFR2-mediated angiogenesis in endothelial cells, such inhibition of Akt/mTOR and
434
ERK1/2-dependent signaling in colon cancer cells may directly contribute to the anticancer
435
effect of AGS-30. The primary target of AGS-30 on HT-29 colon cancer cells is not known
436
and further study is required. It is well established that receptor tyrosine kinases (RTKs) are a
437
class of cell membrane receptors that play important roles in the regulation of cell survival, 20
438
proliferation and migration by activating the downstream PI3K/Akt/mTOR and MAPK/ERK
439
signaling pathways. We speculate that AGS-30 not only inhibits VEGFR2 but also other
440
RTKs, such as epidermal growth factor receptors (EGFR), which are found in cancer cells.
441
To support this notion, previous studies have demonstrated the potential blocking activities of
442
Andro on EGFR in cancer cells [40]. In addition, the inhibition of cancer cell proliferation via
443
the direct blocking of Akt/mTOR and ERK pathways is not impossible.
444 445
VEGF plays a critical role in tumor angiogenesis, growth, and metastasis. Preclinical and
446
clinical studies have shown that solid tumors including colon cancer express high levels of
447
VEGF in the tumor microenvironment that stimulate endothelial cells for angiogenesis [41].
448
VEGF is the most important angiogenic factor in the progression of human colon cancer and
449
its overexpression has been linked to tumor metastasis and poor prognosis [42]. Bevacizumab,
450
an anti-angiogenic agent targeting VEGF, was first approved by the FDA in 2004 and is used
451
as first-line therapy for metastatic colorectal cancer [43]. In the present study, we
452
demonstrated that VEGF is overexpressed in HT-29 colon cancer cells. Interestingly, VEGF
453
was potently suppressed by AGS-30 in these cells as well as in the tumor tissue of nude mice.
454
Thus, AGS-30 showed promise as an inhibitor of tumor angiogenesis not only through
455
blockade of VEGFR2-mediated signaling but also by reducing VEGF expression in tumor
456
cells.
457 458
The in vitro and in vivo anti-angiogenic activities of Andro have been previously
459
demonstrated.
Andro
was
shown
to
inhibit 21
tumor
angiogenesis
by
blocking
460
VEGF/VEGFR2-mediated MAPK signaling [20]; it also blocked VEGF-induced endothelial
461
cell migration and tube formation at 2.5 μM and cell proliferation at 7.5 μM [20]. However, 5
462
μM Andro had no effect on endothelial cells in our study. The reason is unclear but it is likely
463
due to the different sources of Andro. In the earlier study, Andro was isolated from
464
Andrographis paniculata by the authors [20]. However, we used Andro (purity ≧ 98%)
465
purchased commercially. The purity of Andro used in Shen et.al. was not known and we
466
cannot completely exclude the possibility that it might contain other bioactive ingredients that
467
could enhance the anti-angiogenic activity. However, our findings are in good agreement
468
with another report that Andro has anti-angiogenic activity in HUVECs at concentrations
469
higher than 25 μM and that Andro had no effects on blood vessel formation in zebrafish even
470
at a concentration as high as 300 μM [44].
471 472
Rat aortic rings and Matrigel plugs are two classic models for evaluating angiogenesis ex vivo
473
and in vivo, respectively [45]. To date, there are no reports on the anti-angiogenic effects of
474
Andro or its derivatives in the rat aortic ring model. Our results showed that 5 μM Andro had
475
no effects in this model whereas AGS-30 suppressed VEGF-induced microvessel sprouting.
476
In previous work, Andro at a concentration of 5 μM decreased blood vessel formation by
477
20% in the Matrigel plug model; moreover, Andro (3 mg/kg) blocked tumor growth and
478
reduced tumor weight by approximately 30% in nude mice [19]. These data are consistent
479
with our results, which demonstrated that 5 μM Andro inhibited VEGF-induced angiogenesis
480
by 15% in the Matrigel plug model and that 2 mg/kg Andro decreased tumor weight by 20%.
481
Importantly, we found that AGS-30 had a more potent anti-angiogenic effect in the Matrigel 22
482
plug assay and anti-tumor effects in nude mice bearing human colon tumors than its parent
483
compound Andro. AGS-30 suppressed tumor growth and decreased tumor weight by 55%
484
compared to the control group, whereas body weight was unaffected by treatment with
485
AGS-30 for 20 days. Additionally, AGS-30 did not cause any organ damage in mice,
486
highlighting the safety of this compound. The results of the immunohistochemical analysis
487
showed that AGS-30 inhibited blood vessel formation and VEGF and p-VEGFR2 expression
488
in the tumor microenvironment. The large necrotic areas observed in the tumor tissue of
489
AGS-30-treated mice may have resulted from the decreased supply of oxygen and nutrients
490
to tumor cells caused by the inhibition of angiogenesis.
491 492
In our previous study, AGP-40, a 14-phenoxy andrographolide acetal derivative of Andro,
493
was shown to exert anti-angiogenic activities in vitro and in vivo; the lowest effective
494
concentration of AGP-40 in both HUVECs and zebrafish was 10 μM [46], which is 3- to
495
4-fold
496
3,19-acetonylidene-14α-(2-methoxymethoxy) andrographolide, was shown to inhibit
497
VEGF-induced endothelial cell proliferation, migration, invasion, and tube formation at a
498
concentration as low as 2.5 μM [27], which is similar to AGS-30. However, the average
499
effective concentration of 3,19-acetonylidene-14α-(2-methoxymethoxy) andrographolide for
500
inhibiting angiogenesis in zebrafish was 103.9 ± 5.9 μM, which is much higher than that of
501
AGS-30 (7.5 ± 2.3 μM). These data indicated that AGS-30 was the most potent
502
anti-angiogenic and anticancer agent among all Andro derivatives tested to date.
higher
than
what
was
observed
503 23
here.
Another
derivative
of
Andro,
504
In conclusion, a new derivative of Andro, AGS-30, was synthesized by modifying the 14
505
position of Andro. In vitro and in vivo studies showed that this derivative was a more potent
506
anti-angiogenic and antitumor agent than its parent compound. AGS-30 strongly suppressed
507
tumor cell growth and proliferation by inhibiting Akt/mTOR and ERK-dependent pathways.
508
In addition, AGS-30 reduced VEGF expression in tumor cells and inhibited VEGF-induced
509
endothelial cell proliferation, migration, invasion, and tube formation by blocking
510
VEGFR2-mediated signaling. These findings highlighted the therapeutic potential of AGS-30
511
in the treatment of cancer.
512 513
5. Competing interests
514
The authors declare that they have no conflicts of interest.
24
516
6. References
517
[1] I. Paterson, E.A. Anderson, The renaissance of natural products as drug candidates,
518
Science 310(5747) (2005) 451-453.
519
[2] M.S. Butler, Natural products to drugs: natural product-derived compounds in clinical
520
trials, Nat Prod Rep 25(3) (2008) 475-516.
521
[3] D.J. Newman, G.M. Cragg, Natural Products as Sources of New Drugs from 1981 to 2014,
522
Journal of natural products 79(3) (2016) 629-661.
523
[4] S. Hoelder, P.A. Clarke, P. Workman, Discovery of small molecule cancer drugs:
524
Successes, challenges and opportunities, Mol Oncol 6(2) (2012) 155-176.
525
[5] J. Folkman, Angiogenesis in Cancer, Vascular, Rheumatoid and Other Disease, Nature
526
medicine 1(1) (1995) 27-31.
527
[6] J. Folkman, Fighting cancer by attacking its blood supply, Scientific American 275(3)
528
(1996) 150-4.
529
[7] J. Folkman, A new link in ovarian cancer angiogenesis: lysophosphatidic acid and
530
vascular endothelial growth factor expression, Journal of the National Cancer Institute 93(10)
531
(2001) 734-5.
532
[8] F.A. Eskens, Angiogenesis inhibitors in clinical development; where are we now and
533
where are we going?, British journal of cancer 90(1) (2004) 1-7.
534
[9] J. Folkman, M. Bach, J.W. Rowe, F. Davidoff, P. Lambert, C. Hirsch, et al., Tumor
535
Angiogenesis - Therapeutic Implications, New Engl J Med 285(21) (1971) 1182-&.
536
[10] K. Imai, A. Takaoka, Comparing antibody and small-molecule therapies for cancer,
537
Nature reviews. Cancer 6(9) (2006) 714-27. 25
538
[11] R.N. Gacche, R.J. Meshram, Angiogenic factors as potential drug target: Efficacy and
539
limitations of anti-angiogenic therapy, Bba-Rev Cancer 1846(1) (2014) 161-179.
540
[12] S.P. Wang, Z.F. Zhong, J.B. Wan, W. Tan, G.S. Wu, M.W. Chen, et al., Oridonin
541
Induces Apoptosis, Inhibits Migration and Invasion on Highly-Metastatic Human Breast
542
Cancer Cells, Am J Chinese Med 41(1) (2013) 177-196.
543
[13] Y. Dong, T. Zhang, J.J. Li, H.Y. Deng, Y.J. Song, D. Zhai, et al., Oridonin Inhibits
544
Tumor Growth and Metastasis through Anti-Angiogenesis by Blocking the Notch Signaling,
545
Plos One 9(12) (2014).
546
[14] J. Li, Y. Wu, D. Wang, L. Zou, C. Fu, J. Zhang, et al., Oridonin synergistically enhances
547
the anti-tumor efficacy of doxorubicin against aggressive breast cancer via pro-apoptotic and
548
anti-angiogenic effects, Pharmacol Res 146 (2019) 104313.
549
[15] W. Zhu, Y. Ou, Y. Li, R. Xiao, M. Shu, Y. Zhou, et al., A small-molecule triptolide
550
suppresses angiogenesis and invasion of human anaplastic thyroid carcinoma cells via
551
down-regulation of the nuclear factor-kappa B pathway, Molecular pharmacology 75(4)
552
(2009) 812-9.
553
[16] J.X. Ma, Y.L. Sun, Y.Q. Wang, H.Y. Wu, J. Jin, X.F. Yu, Triptolide induces apoptosis
554
and inhibits the growth and angiogenesis of human pancreatic cancer cells by downregulating
555
COX-2 and VEGF, Oncology research 20(8) (2013) 359-68.
556
[17] H.Y. Cheung, C.S. Cheung, C.K. Kong, Determination of bioactive diterpenoids from
557
Andrographis paniculata by micellar electrokinetic chromatography, J Chromatogr A 930(1-2)
558
(2001) 171-6.
559
[18] J. Zhou, G.D. Lu, C.S. Ong, C.N. Ong, H.M. Shen, Andrographolide sensitizes cancer 26
560
cells to TRAIL-induced apoptosis via p53-mediated death receptor 4 up-regulation,
561
Molecular cancer therapeutics 7(7) (2008) 2170-80.
562
[19] Y.L. Peng, Y. Wang, N. Tang, D.D. Sun, Y.L. Lan, Z.L. Yu, et al., Andrographolide
563
inhibits breast cancer through suppressing COX-2 expression and angiogenesis via
564
inactivation of p300 signaling and VEGF pathway, J Exp Clin Canc Res 37 (2018).
565
[20] K. Shen, L. Ji, B. Lu, C. Xu, C. Gong, G. Morahan, et al., Andrographolide inhibits
566
tumor
567
Chemico-biological interactions 218 (2014) 99-106.
568
[21] T.G. Blanchard, R. Lapidus, V. Banerjee, A.C. Bafford, S.J. Czinn, H. Ahmed, et al.,
569
Upregulation of RASSF1A in Colon Cancer by Suppression of Angiogenesis Signaling and
570
Akt Activation, Cellular physiology and biochemistry : international journal of experimental
571
cellular physiology, biochemistry, and pharmacology 48(3) (2018) 1259-1273.
572
[22] A.A. Abu-Ghefreh, H. Canatan, C.I. Ezeamuzie, In vitro and in vivo anti-inflammatory
573
effects of andrographolide, Int Immunopharmacol 9(3) (2009) 313-8.
574
[23] Z. Yu, B. Lu, Y. Sheng, L. Zhou, L. Ji, Z. Wang, Andrographolide ameliorates diabetic
575
retinopathy by inhibiting retinal angiogenesis and inflammation, Biochimica et biophysica
576
acta 1850(4) (2015) 824-31.
577
[24] M.Y. Yang, Q.L. Yu, Y.S. Huang, G. Yang, Neuroprotective effects of andrographolide
578
derivative CX-10 in transient focal ischemia in rat: Involvement of Nrf2/AE and
579
TLR/NF-kappaB signaling, Pharmacol Res 144 (2019) 227-234.
580
[25] K. Sheeja, C. Guruvayoorappan, G. Kuttan, Antiangiogenic activity of Andrographis
581
paniculata extract and andrographolide, Int Immunopharmacol 7(2) (2007) 211-221.
angiogenesis
via
blocking
VEGFA/VEGFR2-MAPKs
27
signaling
cascade,
582
[26] K. Kajal, A.K. Panda, J. Bhat, D. Chakraborty, S. Bose, P. Bhattacharjee, et al.,
583
Andrographolide binds to ATP-binding pocket of VEGFR2 to impede VEGFA-mediated
584
tumor-angiogenesis, Scientific reports 9(1) (2019) 4073.
585
[27] D.K. Sheng, J.J. Li, K. Wang, Y.R. Peng, S. Li, Y.C. Sun, et al., Differential in vitro and
586
in vivo anti-angiogenic activities of acetal and ketal andrographolide derivatives in HUVEC
587
and zebrafish models, Rsc Adv 6(105) (2016) 102831-102842.
588
[28] F. Li, X.M. Li, D.K. Sheng, S.R. Chen, X. Nie, Z.Y. Liu, et al., Discovery and
589
preliminary SAR of 14-aryloxy-andrographolide derivatives as antibacterial agents with
590
immunosuppressant activity, Rsc Adv 8(17) (2018) 9440-9456.
591
[29] J. Zhang, J. Li, Z. Shi, Y. Yang, X. Xie, S.M. Lee, et al., pH-sensitive polymeric
592
nanoparticles for co-delivery of doxorubicin and curcumin to treat cancer via enhanced
593
pro-apoptotic and anti-angiogenic activities, Acta Biomater 58 (2017) 349-364.
594
[30] J. Li, J. Zhang, L. Zou, S.M. Lee, C. Yang, S.W. Seto, et al., Pro-angiogenic effects of
595
Ilexsaponin A1 on human umbilical vein endothelial cells in vitro and zebrafish in vivo,
596
Phytomedicine : international journal of phytotherapy and phytopharmacology 36 (2017)
597
229-237.
598
[31] K.M. Cook, W.D. Figg, Angiogenesis inhibitors: current strategies and future prospects,
599
CA: a cancer journal for clinicians 60(4) (2010) 222-43.
600
[32] L. Adnane, P.A. Trail, I. Taylor, S.M. Wilhelm, Sorafenib (BAY 43-9006, Nexavar (R)),
601
a dual-action inhibitor that targets RAF/MEK/ERK pathway in tumor cells and tyrosine
602
kinases VEGFR/PDGFR in tumor vasculature, Method Enzymol 407 (2006) 597-+.
603
[33] J. Baum, B. Johnson, S. Adams, J. Tang, S. Iadevaia, B. Schoeberl, et al., MM-141, a 28
604
Novel Bispecific Antibody Co-targeting IGF-1R and ErbB3, Inhibits PI3K/Akt/mTOR
605
Pro-survival Signaling in Preclinical Cancer Models, Eur J Cancer 48 (2012) 97-97.
606
[34] J. Karar, A. Maity, PI3K/AKT/mTOR Pathway in Angiogenesis, Frontiers in molecular
607
neuroscience 4 (2011) 51.
608
[35] C. Montagut, J. Settleman, Targeting the RAF-MEK-ERK pathway in cancer therapy,
609
Cancer letters 283(2) (2009) 125-134.
610
[36] R. Srinivasan, T. Zabuawala, H. Huang, J.Y. Zhang, P. Gulati, S. Fernandez, et al., Erk1
611
and Erk2 Regulate Endothelial Cell Proliferation and Migration during Mouse Embryonic
612
Angiogenesis, Plos One 4(12) (2009).
613
[37] J.S. Mudgett, J.X. Ding, L. Guh-Siesel, N.A. Chartrain, L. Yang, S. Gopal, et al.,
614
Essential role for p38 alpha mitogen-activated protein kinase in placental angiogenesis, P
615
Natl Acad Sci USA 97(19) (2000) 10454-10459.
616
[38] A. Cuenda, S. Rousseau, P38 MAP-Kinases pathway regulation, function and role in
617
human diseases, Bba-Mol Cell Res 1773(8) (2007) 1358-1375.
618
[39] L. Liu, Y. Cao, C. Chen, X. Zhang, A. McNabola, D. Wilkie, et al., Sorafenib blocks the
619
RAF/MEK/ERK pathway, inhibits tumor angiogenesis, and induces tumor cell apoptosis in
620
hepatocellular carcinoma model PLC/PRF/5, Cancer research 66(24) (2006) 11851-8.
621
[40] Y. Tan, K.H. Chiow, D. Huang, S.H. Wong, Andrographolide regulates epidermal
622
growth factor receptor and transferrin receptor trafficking in epidermoid carcinoma (A-431)
623
cells, Brit J Pharmacol 159(7) (2010) 1497-1510.
624
[41] L.M. Ellis, Y. Takahashi, W. Liu, R.M. Shaheen, Vascular endothelial growth factor in
625
human colon cancer: biology and therapeutic implications, The oncologist 5 Suppl 1 (2000) 29
626
11-5.
627
[42] A. Ahluwalia, M.K. Jones, S. Szabo, A.S. Tarnawski, Aberrant, ectopic expression of
628
VEGF and VEGF receptors 1 and 2 in malignant colonic epithelial cells. Implications for
629
these cells growth via an autocrine mechanism, Biochemical and biophysical research
630
communications 437(4) (2013) 515-20.
631
[43] N. Ferrara, K.J. Hillan, H.P. Gerber, W. Novotny, Discovery and development of
632
bevacizumab, an anti-VEGF antibody for treating cancer, Nature reviews. Drug discovery 3(5)
633
(2004) 391-400.
634
[44] B. Huang, Y. Peng, J. Li, S. Li, Y. Sun, D. Wang, et al., An andrographolide derivative
635
AGP-26b exhibiting anti-angiogenic activity in HUVECs and zebrafish via blocking the
636
VEGFA/VEGFR2 signaling pathway, Molecular bioSystems 13(3) (2017) 525-536.
637
[45] C.A. Staton, S.M. Stribbling, S. Tazzyman, R. Hughes, N.J. Brown, C.E. Lewis, Current
638
methods for assaying angiogenesis in vitro and in vivo, Int J Exp Pathol 85(5) (2004) 233-48.
639
[46] J. Li, Y. Peng, S. Li, Y. Sun, J.Y. Chan, G. Cui, et al., Anti-angiogenic activity of a new
640
andrographolide derivative in zebrafish and HUVECs, Eur J Pharmacol 789 (2016) 344-353.
641
30
643
Scheme 1 O HO
O
HO a
HO
H
HO
644
Andrographolide (Andro)
Cl
O
O
O b
O
H
O
H O
O Intermediate
645
31
AGS-30
O O
647
Figure 1
648
32
650
Figure 2
651 652
33
654
Figure 3
655
34
657
Figure 4
658
35
660
Figure 5
661 662 663 664 36
665
Figure 6
666
37
668
Figure 7
669
38
671
Figure 8
672
39
674
Figure 9
675
40
677
Scheme 1. Synthesis of AGS-30. Reagents and conditions: (a) previously described; (b)
678
anhydrous tetrahydrofuran, intermediate (1.0 eq.), 2-chlorophenol (1.5 eq.), diisopropyl
679
azodicarboxylate (1.5 eq.), triphenylphosphine (1.5 eq.), 0°C to room temperature.
680 681 682
Figure 1. Effect of AGS-30 on the viability of HT-29 colon cancer cells and HUVECs. (A,
683
B) HT-29 cells (A) and HUVECs (B) were treated with various concentrations of Andro and
684
AGS-30 (0–20 μM) for 24 and 48 h, and cell viability was evaluated with the MTT assay. (C)
685
Cellular damage in HUVECs detected with the LDH assay. (D) HUVECs were starved in
686
low-serum medium (0.5%) for 24 h and then left untreated (control) or treated with VEGF
687
(20 ng/mL) in the presence of various concentrations of Andro (5–20 μM) and AGS-30
688
(1.25–5 μM) for 48 h. Viable cell number was quantified by Trypan Blue staining. Data are
689
presented as a percentage of the control group (mean ± SD of three independent experiments).
690
#P
< 0.05 vs. control; *P < 0.05 vs. VEGF treatment.
691 692 693
Figure 2. Effect of AGS-30 on VEGF-induced endothelial cell migration, invasion, and
694
tube formation. (A) Transwell migration assay. HUVECs were left untreated (control) or
695
treated with VEGF (20 ng/mL) in the presence of Andro (5 μM) or various concentrations of
696
AGS-30 (1.25–5 μM) for 24 h. The migration of HUVECs was detected by nuclear staining
697
of cells on the lower side of the polycarbonate membrane coated with collagen in the
698
Transwell system using 10 μg/mL Hoechst 33342. (B) Transwell invasion assay. HUVECs 41
699
were left untreated (control) or treated with VEGF (20 ng/mL) in the presence of Andro (5
700
μM) or various concentrations of AGS-30 (1.25–5 μM) for 24 h. The invasion of HUVECs
701
was detected by nuclear staining as described above. (C) HUVECs seeded on microslides
702
coated with Matrigel were left untreated (control) or treated with VEGF (20 ng/mL) in the
703
presence of Andro (5 μM) and various concentrations of AGS-30 (1.25–5 μM) for 6 h. (D–F)
704
Quantitative analysis of HUVEC migration (D), invasion (E), and tube formation (F). Scale
705
bar: 200 μm. Data are presented as percentage of the control group (mean ± SD of three
706
independent experiments). #P < 0.05 vs. control; *P < 0.05 vs. VEGF treatment.
707 708 709
Figure 3. Effect of AGS-30 on VEGFR2-mediated angiogenesis. HUVECs were cultured
710
in low-serum medium (0.5%) for 3 h and then treated with DMSO (0.1%, vehicle control) or
711
different concentrations of AGS-30 (1.25–5 μM) for 8 h, then stimulated with VEGF (50
712
ng/mL) for 15 min. (A) Expression levels of the major proteins involved in
713
VEGFR2-mediated angiogenesis in HUVECs as determined by Western blotting. (B–G)
714
Quantitative analysis of protein expression levels. Data are presented as a percentage of the
715
control group (mean ± SD of three independent experiments). #P < 0.05 vs. control; *P < 0.05
716
vs. VEGF treatment.
717 718 719 720 42
721
Figure 4. Effect of AGS-30 on the expression of VEGF and other proteins related to
722
colon cancer cell survival and proliferation. (A) HT-29 human colon cancer cells were
723
treated with various concentrations of AGS-30 (1.25–5 μM) for 48 h, and the expression
724
levels of p-Akt, Akt, p-mTOR, mTOR, p-ERK1/2, ERK1/2, and intracellular VEGF were
725
detected by Western blotting. (B–E) Quantitative analysis of protein levels. (F) Extracellular
726
VEGF levels of HT-29 colon cancer cells were measured by ELISA. Data are presented as a
727
percentage of the control group (mean ± SD of three independent experiments). *P < 0.05 vs.
728
control group.
729 730
Figure 5. Effect of AGS-30 on blood vessel formation in a transgenic zebrafish model.
731
(A–F) At 24 hpf, Tg(fli-1: EGFP) zebrafish embryos were treated with 0.1% DMSO (vehicle
732
control), 250 ng/mL VRI (positive control), 300 μM Andro, or various concentrations of
733
AGS-30 (3–30 μM) for 24 h. The embryos were photographed under an inverted fluorescence
734
microscope. (G–L) Magnified views of panels A to F. Yellow arrows indicate intersegmental
735
vessels (ISVs), dorsal aorta (DA), and dorsal longitudinal anastomotic vessel (DLAV); white
736
asterisk indicates ISV loss. (M) Quantitative analysis of blood vessel loss in zebrafish,
737
performed by counting the number of defective and intact ISVs in each embryo at 48 hpf. *P
738
< 0.05 vs. defective ISVs in control group; #P < 0.05 vs. intact ISVs in control group. (N)
739
Quantitative analysis of the anti-angiogenic phenotype of zebrafish. Embryos with any
740
incomplete ISVs were defined as exhibiting an anti-angiogenic phenotype. Scale bar: 500 μm.
741
Data are presented as the mean ± SD of ≥ 12 independent experiments *P < 0.05 vs. control.
742 43
743
Figure 6. Effect of AGS-30 on angiogenesis ex vivo and in vivo. (A) Representative
744
photographs of microvessel sprouting in rat aortic rings. The rings were embedded in
745
Matrigel and treated with 5 µM Andro and AGS-30 in the presence of VEGF (100 ng/mL)
746
for 10 days. Microvessel growth was recorded with an inverted microscope and the number
747
of sprouting microvessels was quantified with ImageJ software. Aortic rings received DMSO
748
(0.1%) and VEGF (100 ng/mL) served as negative control and positive control, respectively.
749
Scale bar: 100 µm. (B) Quantitative analysis of sprouting microvessels in each group. Data
750
are presented as a percentage of the control group (mean ± SD of three independent
751
experiments). $P < 0.05 vs. control; *P < 0.05 vs. VEGF treatment; #P < 0.05 vs. Andro. (C)
752
Representative photographs of blood vessel formation in Matrigel plugs. The 8-week-old
753
C57BL/6N female mice (n = 5) were subcutaneously injected with 0.5 mL of growth
754
factor-reduced Matrigel containing VEGF (250 ng) and heparin (150 Unit) with or without 5
755
µM Andro and AGS-30. After 14 days, the mice were sacrificed and the plugs were removed
756
and photographed. Scale bar: 2 mm. (D) Quantitative analysis of hemoglobin levels in
757
Matrigel plugs. To quantify the formation of functional blood vessels, the amount of
758
hemoglobin was measured using Drabkin reagent. Data are presented as a percentage of the
759
control group (mean ± SD of five independent experiments). $P < 0.05 vs. control; *P < 0.05
760
vs. VEGF treatment; #P < 0.05 vs. Andro.
761 762 763 764 44
765
Figure 7. Effect of AGS-30 on tumor growth in colon tumor cell-derived xenograft mice.
766
The subcutaneous dorsum of nude mice (4-weeks-old; 16 g) was inoculated with HT-29
767
colon cancer cells. Tumors were allowed to grow to approximately 200 mm3, and the mice
768
were intraperitoneally injected once every 2 days for 20 days with saline, 2 mg/kg Andro, or
769
AGS-30. (A) Images of excised tumors obtained at the end of the experiment. (B) Tumor
770
weight measured at the end of the experiment. (C) Time course of tumor growth progression.
771
(D) Time course of body weight. Data are presented as mean ± SD of five independent
772
experiments. *P < 0.05 vs. saline; #P < 0.05 vs. Andro.
773 774 775
Figure 8. Immunohistochemical analysis of tumor tissues. (A) Tumor tissue (n = 5) was
776
fixed with 4% (v/v) formaldehyde and cut into 6-μm sections that were stained with H&E to
777
evaluate tissue architecture. The sections were labeled with anti-CD31 antibody to visualize
778
blood vessels and with anti-VEGF and anti-p-VEGFR2 antibodies to evaluate tumor
779
angiogenesis. (B–E) Quantitative analysis of necrotic area, blood vessel formation, and
780
VEGF and p-VEGFR2 expression in each group. Data were quantified with ImageJ software.
781
*P
< 0.05 vs. saline; #P < 0.05 vs. Andro.
782 783 784
Figure 9. Hematoxylin and eosin staining of major organs in mice treated with saline,
785
Andro and AGS-30.
786 45