You are on page 1of 17

Microvascular Research 107 (2016) 17–33

Contents lists available at ScienceDirect

Microvascular Research

journal homepage: www.elsevier.com/locate/ymvre

Scopoletin, an active principle of tree tobacco (Nicotiana glauca) inhibits


human tumor vascularization in xenograft models and modulates ERK1,
VEGF-A, and FGF-2 in computer model
Yasser M. Tabana a,⁎, Loiy Elsir A. Hassan a, Mohamed B. Khadeer Ahamed b, Saad S. Dahham a,
Muhammad Adnan Iqbal b, Mohammed A.A. Saeed c, Md Shamsuddin S. Khan a, Doblin Sandai d,
Aman S. Abdul Majid e, Chern Ein Oon f, Amin Malik S.A. Majid a,⁎
a
EMAN Research and Testing Laboratory, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Pulau Pinang, Malaysia
b
EMAN Biodiscoveries Sdn. Bhd. Suite 126, Level 1, EUREKA Complex, Universiti Sains Malaysia (USM) Campus, Minden 11800, Penang, Malaysia
c
Department of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Universiti Sains Malaysia, Minden 11800, Pulau Pinang, Malaysia
d
Infectomics Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, 13200 Bertam, Penang, Malaysia
e
Department of Pharmacology, Quest International University, Perak, Malaysia
f
Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Minden 11800, Pulau Pinang, Malaysia

a r t i c l e i n f o a b s t r a c t

Article history: We recently reported the antineovascularization effect of scopoletin on rat aorta and identified its potential anti-
Received 10 August 2015 angiogenic activity. Scopoletin could be useful as a systemic chemotherapeutic agent against angiogenesis-
Revised 19 April 2016 dependent malignancies if its antitumorigenic activity is investigated and scientifically proven using a suitable
Accepted 24 April 2016
human tumor xenograft model. In the present study, bioassay-guided (anti-angiogenesis) phytochemical inves-
Available online 29 April 2016
tigation was conducted on Nicotiana glauca extract which led to the isolation of scopoletin. Further, anti-
Keywords:
angiogenic activity of scopoletin was characterized using ex vivo, in vivo and in silico angiogenesis models. Final-
Scopoletin ly, the antitumorigenic efficacy of scopoletin was studied in human colorectal tumor xenograft model using
Nicotiana glauca athymic nude mice. For the first time, an in vivo anticancer activity of scopoletin was reported and characterized
Xenograft model using xenograft models. Scopoletin caused significant suppression of sprouting of microvessels in rat aortic ex-
Antineovascularization plants with IC50 (median inhibitory concentration) 0.06 μM. Scopoletin (100 and 200 mg/kg) strongly inhibited
Angiogenic mitogens (59.72 and 89.4%, respectively) vascularization in matrigel plugs implanted in nude mice. In the tumor xenograft
model, scopoletin showed remarkable inhibition on tumor growth (34.2 and 94.7% at 100 and 200 mg/kg, respec-
tively). Tumor histology revealed drastic reduction of the extent of vascularization. Further, immunostaining
of CD31 and NG2 receptors in the histological sections confirmed the antivascular effect of scopoletin in tumor
vasculature. In computer modeling, scopoletin showed strong ligand affinity and binding energies toward the fol-
lowing angiogenic factors: protein kinase (ERK1), vascular endothelial growth factor A (VEGF-A), and fibroblast
growth factor 2 (FGF-2). These results suggest that the antitumor activity of scopoletin may be due to its strong
anti-angiogenic effect, which may be mediated by its effective inhibition of ERK1, VEGF-A, and FGF-2.
© 2016 Elsevier Inc. All rights reserved.

Introduction 2012; Wang and Zheng, 2012). For centuries, natural products have
played a strategic role in maintaining human health, and they have
Targeting angiogenesis via suppressing angiogenic factors is one of been utilized as a major source of remedies in all human civilizations.
the most recent successful strategies for cancer treatment. Cancer cells Thus, researchers have been looking for angiogenesis inhibitors and
produce various angiogenic factors to initiate angiogenesis. Vascular promoters from natural products for the past 15 years (Wang et al.,
endothelial growth factor A (VEGFA), fibroblast growth factor 2 2004).
(FGF2) and extracellular signal-regulated kinases-1 (ERK-1) are com- Tree tobacco, botanically known as Nicotiana glauca (Solanaceae), is
monly expressed in most of the cancers and their expression levels a plant widely used in traditional medicine to treat various infectious
have been correlated with tumorigenesis and metastasis (Cao et al., and inflammatory diseases. N. glauca is native to central northwest
Argentina and Bolivia, however later on the plant has invaded Central
⁎ Corresponding authors.
and North America (Mexico, southern US states including California)
E-mail addresses: Yasser.tabana@hotmail.com (Y.M. Tabana), and Africa, specifically Morocco, South Africa and Namibia (Florentine
aminmalikshah@gmail.com (A.M.S.A. Majid). and Westbrooke, 2005; Ibrahim, 2012). The plant can also be found in

http://dx.doi.org/10.1016/j.mvr.2016.04.009
0026-2862/© 2016 Elsevier Inc. All rights reserved.
18 Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33

Fig. 1. Isolation of scopoletin. Schematic diagram showing the bioassay guided sequential extraction and fractionation of N. glauca leading to isolation of scopoletin.

Australia and is widely distributed from mesic to arid environments, angiogenesis-dependent malignancies, hence the anti-angiogenic prop-
including the Negev desert of Israel. Extracts from this plant have erty of scopoletin was needed to be confirmed using advanced and suit-
been reported to have dynamic biological activities and are commonly able animal models of angiogenesis and tumorigenesis. Based on this
used as a tonic, paste, or drink to treat general pain, boils, headaches, background, the present study was conducted to identify the active
piles, sores, and wounds. The extracts also were reported to have strong principle (scopoletin) of N. glauca extract which is responsible for its
antimicrobial and cytotoxic activities (Mdee et al., 2009). In a previous strong anti-angiogenic property. Furthermore, an attempt was made
study, we reported the strong anti-angiogenic effect of the n-hexane ex- to characterize the anti-angiogenic activity of scopoletin using ex vivo,
tract of N. glauca leaves on sprouting of blood vessels in the rat aorta in vivo and in computer modeling experimental systems. Finally, the
(Hassan et al., 2014). However, the active compound responsible for anti-angiogenic property of scopoletin was exploited to study the po-
the bioactivity of the herb was not known. tential in vivo anti-tumor activity using human tumor xenograft model.
Chemically, N. glauca is rich in coumarin-derived compounds.
Scopoletin is a 6-methoxy-7-hydroxycoumarin isolated from N. glauca.
Our previous study showed that scopoletin has promising anti- Materials and methods
angiogenic activity (Beh et al., 2012). Several other studies showed
that scopoletin possesses potent hepatoprotective (Kang et al., 1998), Chemicals, cell culture, and reagents
anti-oxidant (Shaw et al., 2003), and spasmolytic (Oliveira et al.,
2001) activities. Liu et al. (2001) reported that scopoletin inhibited The chemicals used in the cell culture study (penicillin/streptomycin
cell proliferation by inducing cell cycle arrest and increasing apoptosis (PS), phosphate buffered saline (PBS), diaminobenzidine, hematoxylin,
in human prostate tumor cells. Recently, scopoletin was reported to trypsin, avidin–biotin complex and MTS (3-(4,5-dimethylthiazol-2-yl)-
inhibit in vitro proliferation, migration and tube formation properties 5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium) were
of human endothelial cells (Pan et al., 2009). As scopoletin could procured from Sigma Aldrich (Darmstadt, Germany). Xylene, 3% H2O2,
prove to be useful as a systemic chemotherapeutic agent against antigen retrieval solution pH 6 and anti-rabbit biotinylated antibody
Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33 19

Fig. 2. Anti-angiogenic effect of scopoletin. Photomicrographic images show inhibition of sprouting of microvessels in rat aortic explants. The aortic rings were photographed at 20×
magnification after 5 days of treatment with scopoletin and the standard reference, suramin. A: control. B: scopoletin (0.06 μM). C: scopoletin (0.12 μM). D: scopoletin (0.25 μM).
E: scopoletin (0.5 μM). F: suramin (0.5 μM). G: graphical representation of the anti-angiogenic activity of scopoletin in comparison with that of the standard reference, suramin. All
values are expressed as mean ± SEM (n = 8). *p b 0.05, **p b 0.01.
20 Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33

were purchased from (Dako, USA). CD34 primary antibody was procured model was approved with the approval reference number: USM/Animal
from (ABCAM, UK). Ethics Approval/2014/(94) (589).
The reagents for the ex vivo culture study (aprotinin, fibrinogen, bo-
vine serum albumin (BSA), thrombin, matrigel, suramin, and vincristine) Acute toxicity of scopoletin
also were purchased from Sigma. HCT 116 (human colon cancer) and
human umbilical vein endothelial cells (HUVECs), colon cancer (HCT The acute toxic effect of scopoletin was assessed using a single dose
116) and normal (CCD-18Co) cells were purchased from American administration test, which was employed using OECD guideline-425
Type Culture Collection (ATCC, Manassas, VA, USA) and grown in suitable (Khadeer Ahamed et al., 2010).
cell medium supplemented with 5% heat inactivated fetal bovine serum
(HIFBS), both purchased from ScienCell (St. Louis, MO, USA), and 1% PS. Ex vivo rat aortic ring assay
Other chemicals used were either high performance liquid chromatogra-
phy (HPLC) grade or analytical grade. Methanol and orthophosphoric The rat aortic ring assay was performed following (Al-Salahi et al.,
acid were procured from QRec (New Zealand) and acetonitrile HPLC 2013) with slight modifications. Different concentrations (0.03125,
grade was purchased from Merck (Darmstadt, Germany). Thin layer 0.0625, 0.125, 0.25, and 0.5 μM) of scopoletin were used to determine
chromatography (TLC) plates coated with 0.25 mm Silica Gel 60 F254 the IC50 (median inhibitory concentration) values using regression
were obtained from Merck. An ultraviolet (UV) fluorescence cabinet equation obtained from the concentration-activity curve. Suramin
CM-10 (New York, USA) was used to visualize chromatograms on the (0.03 to 0.5 μM) and DMSO (0.1%) were used as positive and negative
TLC plates. A Sony Cyber-shot DSC-T33 digital camera was used to obtain controls, respectively (Dahham et al., 2015a, 2015b; Nassar et al., 2011)
the TLC photographs.
MTS assay
Plant material
MTS assay was performed to assess the anti-proliferative effect of
The plant material was collected from the Botanical Garden at Kassala, scopoletin on human endothelial (HUVECs), normal colon (CCD-18Co)
Eastern Sudan during the period of March to July 2013 with permission and colon cancer (HCT 116) cells. The assay plate was read using Tecan
from the authorities of the garden. The taxonomic authentication of Infinite® 200 PRO microplate reader (HIDEX, Finland) at 570 nm absor-
N. glauca was performed by Dr. Wail Alsadig at the Medicinal and bance. DMSO (0.1%) was used as a negative control. Suramin and 5-
Aromatic Plants Research Institute, National Center for Research, fluorouracil were used as positive controls for HUVEC and HCT 116
Sudan. A voucher specimen (voucher reference number: MAPRI/NB- cells, respectively. Dose–response curves were plotted and IC50 values
53b) was deposited at the herbarium of the institute (Tabana et al., were calculated using the regression equation (Dahham et al., 2015a,
2015). All experiments were carried out in the EMAN (Experimental 2015b; Tabana et al., 2015). The selectivity index (SI), which indicates
Medicine & Advanced Natureceutical) Lab at Universiti Sains Malaysia the cytotoxic selectivity of the compound against HUVECs and its ineffec-
(USM) with permission from the director of the laboratory. The present tive toward the other cell types, was determined from the ratio of
work did not involve any special locations or activities that required the IC50 of scopoletin on colon fibroblasts (CCD-18Co) versus IC50 on
permission. Furthermore, the present study did not involve endangered HUVECs.
or protected species.
In vivo matrigel plug assay

Isolation and structural confirmation of scopoletin NU/Nu immunocompromised nude mice were injected subcutane-
ously with 0.3 ml of matrigel supplemented with 100 μl of HCT-116
Isolation of scopoletin was carried out using bioassay guided frac- cells (106 × 1 cells/ml) near the abdominal midline (Passaniti et al.,
tionation of n-hexane extract of N. glauca (Fig. 1). The details of extrac- 1992). Mice were then treated orally with 100 and 200 mg/kg of
tion, fractionation and column chromatographic procedures are given in scopoletin (n = 6) daily for 7 days. Vehicle (0.1% Tween 80) and imatin-
the Supplementary information. The structure of scopoletin was ib (100 mg/kg) were used as the negative and positive controls, respec-
confirmed using FT-IR and NMR spectroscopic techniques (refer to the tively. At the end of the experiment, all the animals were euthanized
Supplementary information). and matrigel plugs were carefully excised and sectioned at 5 μm thick-
ness, then stained with hematoxylin and eosin (H&E). The number of
Experimental animals blood vessels in all sections was counted.

Healthy adult female Sprague Dawley (SD) rats (225–250 g) were In vivo assessment of tumor angiogenesis in nude mouse xenograft model
used in the acute toxicity study. For the rat aortic ring assay,
8–10 week old healthy male SD rats (200–230 g) were used. The rats Human colorectal tumors were grown in athymic (Ncr-Nu/Nu)
were obtained from the Animal House Facility, USM. For the xenograft nude mice by injecting HCT-116 cells (107 cells/150 μl DMEM) sub-
model, either sex of athymic NCR-nu/nu (nude) mice (8–12 weeks cutaneously on the dorsal side of the animals (Ahamed et al.,
old) was used. All experiments were conducted according to the guide- 2012). Scopoletin was administered to mice orally once daily in
lines of the Institutional Animal Ethics Committee. The study protocol three different doses: 50, 100, and 200 mg/kg (n = 10). Tween 80
was approved by the Animal Ethics Committee, USM [approval refer- (0.1%) and imatinib (100 mg/kg) were used as negative and positive
ence number: PPSG/07(A)/044/(2010) (61)]. The tumor xenograft controls, respectively. The animals from the negative control group

Fig. 3. H&E stained cross-sections taken from matrigel plugs implanted subcutaneously in representative groups of animals. A: Section of the matrigel from the negative control group
shows high vascularization (arrows) (original magnification of 20×) and more proliferation of HCT-116 cells compared to the other treated groups. B: Magnified section (40×) of a rep-
resentative matrigel plug from the negative control group shows abundance of blood vessels (indicated by the arrows). C: Section (20×) of a matrigel plug harvested from a scopoletin
(100 mg/kg)-treated animal demonstrates significant reduction in vascularization. D: Higher magnification (40 ×) of a section of the matrigel plug harvested from a scopoletin
(100 mg/kg)-treated animal reveals drastic reduction of blood vessels in the matrigel implant. E: A representative matrigel implant from a scopoletin (200 mg/kg)-treated mouse shows
complete inhibition of blood vessels (original magnification of 20×). F: Magnified image (40×) of the matrigel section showing the strong anti-angiogenic effect of scopoletin (200 mg/kg)
treatment. Most of the matrigel implant from this group showed complete inhibition of neovascularization. G: Matrigel section (20×) showing the anti-angiogenic effect of the standard
reference drug, imatinib (100 mg/kg). H: Magnified (40×) photomicrographic image of the matrigel section shows the potent anti-angiogenic effect of imatinib. I: Graphical representa-
tion of the effect of scopoletin on the mean blood vessel count in matrigel sections (* = p b 0.05, ** = p b 0.01, n = 6, values are mean ± SEM of 10 low power microscopic fields).
Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33 21
22 Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33

Fig. 4. In vivo antitumor and anti-angiogenic activities of scopoletin determined using a human tumor xenograft model in athymic nude mice bearing HCT-116 tumors at day 28 post-
inoculation. A: Animals from the vehicle-treated group (negative control). B: Animal treated with 50 mg/kg scopoletin. C: Animals treated with 100 mg/kg scopoletin. D: Animals
treated with 200 mg/kg scopoletin. E: Animals treated with 100 mg/kg imatinib. F: Tumor morphology of the representative groups of animals: (a) negative control, (b) 50 mg/kg of
scopoletin, (c) 100 mg/kg scopoletin, (d) 200 mg/kg scopoletin, and (e) 100 mg/kg imatinib (arrows indicate prompt and well-developed blood vessels in the control, and arrowheads
indicate the reduction of tumor vasculature).

were euthanized when the tumor volume reached the maximum Tools for the molecular docking study
level of 1000 mm3 in size. The tumor-bearing animals were treated
until either the tumor volume reached 1000 mm3 or for 21 days. Ligand crystal structures of scopoletin and suramin were
Tumor tissue was collected from the euthanized animals for histo- downloaded from the ChemSpider database. Protein 3D structures of
logical studies. The number of blood vessels in all tissue sections mitogen-activated protein kinase (ERK1/2) (Kinoshita et al., 2008), vas-
was counted. cular endothelial growth factor A (VEGF-A) (Mandal and Kent, 2011),
and fibroblast growth factor 2 (FGF-2) were downloaded from the
Visualization of tumor vasculature in immunohistochemistry RCSB protein data bank. Suramin was used as the positive control in
this docking study.
The paraffin sections were deparaffinized at 60 °C for 10 min
followed by rehydration in xylene and a series of graded alcohol. In-
Molecular docking study
hibition for endogenous peroxidase was carried out using 3% H2O2
(Dako). Antigen retrieval was performed using antigen retrieval so-
Specific active site-target and automated docking systems were used
lution pH 6 (Dako) for 15 min using the microwave. Sections were
to determine the binding affinity and ligand efficiency of scopoletin
then incubated with 15% bovine serum albumin (SIGMA) for 1 h at
with the major angiogenic mitogens ERK1/2, VEGF-A, and FGF-2. The
room temperature followed by CD34 primary antibody (ABCAM)
activity of scopoletin was compared with that of the standard reference,
overnight at 4 °C. The slides were rinsed with PBS and incubated
suramin. The two different methods were employed using different al-
with anti-rabbit biotinylated antibody (Dako) for 30 min. Sections
gorithms to analyze the comparative activity of scopoletin and suramin.
were then rinsed and incubated with standard avidin–biotin com-
plex (ABC; SIGMA) for 30 min. This is followed by incubation with di-
aminobenzidine (SIGMA) for 5 min and hematoxylin (SIGMA) for LeadIT — FlexX
1 min. Sections were finally mounted in mounting media (Dako)
and imaged using a brightfield microscope. BioSolveIT's LeadIT is a specific active site-target program that is
In addition, pericytes were identified by labeling with affinity- used to dock the compounds as a flexible function of the FlexX algo-
purified rabbit polyclonal antibodies against the NG2 proteoglycan rithm (Böhm, 1998). This program detects the binding site according
according to the manufacturer's instruction (Merck, Germany). to a reference ligand by superimposition of the experimented ligand.
Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33 23

Fig. 5. Illustration of the tumor growth profile in the respective test groups. A: Graphical illustration of the dose-dependent suppression effect of scopoletin on HCT-116 cell tumorigenesis
in nude mice in comparison to the negative and positive controls (Scop = scopoletin; PC = imatinib; NC = vehicle-treated negative control; the numbers in brackets represent the dose in
mg/kg). All values are expressed as mean ± SEM (n = 10). B: Effect of scopoletin on the average body weight of treated animals in comparison to the negative control (HD = high dose
(200 mg/kg) of scopoletin; MD = mid-dose (100 mg/kg) of scopoletin; LD = low dose (50 mg/kg) of scopoletin; PC = positive control (imatinib at 100 mg/kg); NC = vehicle-treated
negative control). All values are mean ± SEM (n = 10).

The active site was defined by selecting the residue of the protein Statistical analysis
with 10 Å at the center of the ligand. The top 10 poses were selected
to analyze the free binding energy (ΔG) of the protein–ligand com- All results are expressed as the mean ± standard error (± SEM).
plex and the ligand efficiency using Hyde assessment (Rarey et al., Statistical differences between the test compounds and the negative
1996). control were analyzed by one-way analysis of variance (ANOVA)
followed by Tukey's multiple comparison test using IBM SPSS software
(Version 20). Differences with p b 0.05 and b0.01 were considered to
SYBYL Surflex Dock be statistically significant.

Surflex Dock is an automated docking program that is used to simu- Results and discussion
late the interaction mode between compounds and protein bio-markers
in SYBYL-X 2.1. The active sites of the protein were selected as the LD50 value of scopoletin is N2000 mg/kg in SD rats
amino acids containing atoms within 6 Å. Five scoring functions of
Dock (DScore) (Meng et al., 1992), Gold (GScore) (Jones et al., 1997), The acute toxicity test showed that scopoletin did not produce
PMF (Pscore) (Muegge and Martin, 1999), ChemScore (Eldridge et al., treatment-related mortality at the limit test dose (2000 mg/kg). Oral
1997), and Consensus (CSCORE) were selected to calculate the correla- administration of scopoletin in rats did not produce any considerable
tion. CSCORE was used to find the correlation with other scores. Only toxic behavioral changes such as apathy, hyperactivity, or morbidity.
the topmost pose was selected to compute the correlation with the Body weight, respiration rate, and heart rate in all treated rats were
reported binding affinity. normal. These results suggest that scopoletin is safe at dose level of
24 Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33
Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33 25

Fig. 7. Photomicrographic images display immunofluorescence staining of differentiation molecule CD34 and the chondroitin sulfate proteoglycan NG2 receptors in the tumor histological
sections. A) Photomicrographic image showing NG2 immunofluorescence staining in negative control group (magnification ×400 μm). B) Photomicrographic image of tissue section from
imatinib treated animal showing NG2 immunofluorescence staining (magnification ×400 μm). C) Photomicrographic image showing NG2 immunofluorescence staining in scopoletin
(100 mg/kg) treated group (magnification × 200 μm). D) Photomicrographic image showing NG2 immunofluorescence staining in scopoletin (200 mg/kg) treated group
(magnification ×200 μm). E) Photomicrographic image showing NG2 immunofluorescence staining in negative control group (magnification ×20 μm). F) Photomicrographic image of
tissue section from imatinib treated animal showing NG2 immunofluorescence staining (magnification ×20 μm). G) Photomicrographic image showing NG2 immunofluorescence
staining in scopoletin (100 mg/kg) treated group (magnification ×20 μm). H) Photomicrographic image showing NG2 immunofluorescence staining in scopoletin (200 mg/kg) treated
group (magnification ×20 μm).

Table 1
Summary of the docking scores and reported binding affinities of scopoletin and suramin with ERK1, VEGF, and FGF2 in the docking analysis using the LeadIT FlexX scoring functions (A =
total scores; B = match score; C = lipophilic area score; D = ambiguous area score, E = receptor-protein overlap score; F = ROT score; RMSD b 0).

Receptor–ligand Binding affinity energy, Ligand efficiency Total Match Lipophilic Ambiguous Receptor–ligand Rotational RMSD
complex (LeadIT) ΔG (Kcal/Mol) (Kcal/mol per heavy atom) score score area score area score interaction score bond score

ERK1-scopoletin −19 0.32 −13.14 −16.73 −5.88 −2.52 3.80 2.80 0.00
ERK1-suramin −10 0.03 −22.05 −21.21 −12.98 −12.69 11.04 8.40 0.00
FGF2-scopoletin −14 0.23 −8.9782 −11.5746 −−4.2712 −5.8630 4.5307 2.8000 0.00
FGF2- suramin No binding – – – – – – – –
VEGF-scopoletin −11 0.20 −3.29 −5.60 −4.0682 −4.90 3.08 2.80 0.00
VEGF-suramin −16 0.04 −28.12 −19.43 −16.55 −10.94 5.0047 8.4000 0.00

2000 mg/kg, thus the LD50 value of scopoletin for oral toxicity is consid- Scopoletin selectively inhibits proliferation of HUVECs
ered to be N 2000 mg/kg.
To characterize the mechanism of scopoletin's inhibitory action
on sprouting of aortic microvessels, a series of in vitro angiogenesis
Scopoletin inhibits sprouting of microvessels in rat aortic explants assays were conducted. Formation of blood vessels primarily de-
pends on proliferation of endothelial cells, thus scopoletin was test-
Results of the rat aortic ring assay demonstrated that scopoletin ed for its effect on proliferation of HUVECs. The results showed that
has significant anti-angiogenic activity. Enormous outgrowth of scopoletin possesses significant (p b 0.01) antiproliferative ability
microvessels was recorded in the vehicle-treated (negative control) toward endothelial cell proliferation with an IC50 of 0.73 μM. Howev-
aortic explants (Fig. 2A). In contrast, scopoletin inhibited sprouting of er, the antiproliferative effect of scopoletin was poor on other cell
microvessels (Fig. 2B–E) at the IC50 of 0.06 μM. The standard reference, types (Thani et al., 2010). A study of Pan et al. (2009) reported that
suramin, also demonstrated a significant inhibitory effect on scopoletin shows cytotoxicity against human endothelial cells only
microvessel formation from the rat aorta (Fig. 2F) at the IC50 of at higher concentration and therefore the study could not estimate
0.22 μM. All tested doses of scopoletin affected the length and density the IC50 value of scopoletin on the cells. However, contrary to the
of microvessels in aortic explants. Fig. 2G illustrates the dose- findings of Pan et al. (2009) the results of the present study revealed
dependent inhibitory effect of scopoletin on sprouting of microvessels that scopoletin possesses a significant cytotoxicity with low IC50
in rat aortic explants. value.

Fig. 6. Necrotic changes in tumor histology and evidence of the anti-angiogenic effect of scopoletin. At the termination of the study, H&E stained tumor sections were examined. A: Tumor
section of a representative animal in the negative control group stained with H&E (original magnification 20×). The photomicrographic image of the tumor section is fully composed of
viable tumor cells (indicated by “t”), a large number of blood vessels (arrows), and virtually no necrosis. B: Magnified (40×) tumor section of the negative control shows well-organized,
densely packed, hexagonal shaped viable tumor cells (t) and blood vessels (arrows). C: Significant changes in tumor histology were noted in immunocompromised animals treated with
scopoletin (100 mg/kg), such as loss of the compact arrangement of tumor cells, and moderate to severe necrotic regions (N) were visible all over the tumor section. Reduced tumor
vasculature was observed as a significantly number of reduced blood vessels (arrows) (original magnification 20×). D: Higher magnification (40×) of a section of the tumor harvested
from a scopoletin (100 mg/kg)-treated animal showing clear evidence of necrosis (N) caused by the treatment. E: A representative tumor from the scopoletin (200 mg/kg)-treated
group (original magnification 20×). The photomicrographic image of the section shows peripheral necrosis, which is evident from the patches of viable tumor cells (t) surrounded by
pink colored dead tissue (necrotic region, N). F: Magnified image (40 ×) of the tumor section show the antitumorigenic and anti-angiogenic effects of scopoletin (200 mg/kg)
treatment. In this image, the peripheral necrotic region can be seen clearly as a small area of live tumor tissue surrounded by dead tissue. A single blood vessel (arrow) can also be
seen at the center of the live tumor tissue area. G: Tumor section (20×) showing the effect of imatinib (100 mg/kg) treatment. Significant necrotic damage can be observed due to the
treatment. H: Magnified (40×) photomicrographic image of the tumor section shows the antitumorigenic effect of imatinib (100 mg/kg). I: Graphical representation of the inhibitory
effect of scopoletin on the mean blood vessel count in HCT-116 tumor sections (*p b 0.05, n = 10, values are mean ± SEM of 10 low power microscopic fields). (For interpretation of
the references to color in this figure legend, the reader is referred to the web version of this article.)
26 Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33

Table 2
Summary of the docking scores and reported binding affinities of scopoletin and suramin with ERK1, VEGF, and FGF2 in the docking analysis using the SYBYL-X Surflex scoring functions (A
= total scores; B = DScore; C = PMF score; D = GScore, E = ChemScore; F = CSCORE; RMSD b 0.5); (C) GOLD Genetic Algorithm Scoring functions (A = gold scores; B = chem score;
RMSD b 0.5).

Receptor–ligand complex Total Crash Polar DScore PMF GScore ChemScore CSCORE COMFA COMSIA COMSIA
(SYBYL) score score STERIC ELECTROSTATIC

ERK1-suramin −0.64 −3.54 6.03 −700.36 −120.47 268.78 −34.74 5 206 7.62 1.01
ERK1-scopoletin 2.02 −0.15 1.69 −417.18 −49.18 5.18 −13.37 4 70 9.59 1.45
FGF2-scopoletin −8.00 −8.10 0 −664.27 108.76 −9.12 −27.48 5 37 8.89 1.35
VEGF-scopoletin 4.17 −0.53 3.30 −53.37 −50.82 −26.75 −14.68 5 35 8.79 1.51
VEGF-suramin 1.90 −3.62 5.63 −158.30 −120.13 226.38 −33.81 5 208 8.33 1.084
FGF2-suramin – – – – – – – No binding – – –

Scopoletin inhibits matrigel induced vasculature in nude mice pellets was clearly evident in untreated mice (Fig. 3A and B). In contrast,
scopoletin (100 mg/kg) treatment significantly (p b 0.05) reduced the
Seven days after implantation of matrigel plugs supplemented with neovascularization process, which was evident from the H&E stained
HCT-116 cells, the formation of hemorrhagic lesions in the matrigel cross-sections of the excised matrigel implants (Fig. 3C and D).

Fig. 8. Visualization of ligands and protein interaction profile. Surface visualization of proteins and active site residue interactions of protein and hydrophobic interactions are shown in the
active pockets of the ligands: A: scopoletin 2D view. B: suramin 2D view. C: FGF2 (cartoon pattern)-scopoletin (ball-stick), SYBYL. D: ERK1 (cartoon pattern)-scopoletin (ball-stick), SYBYL.
E: VEGF (wareframe)-suramin (ball-stick), SYBYL. F: cartoon VEGF-suramin, SYBYL.
Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33 27

Microscopic examination of the blood vessels from the matrigel Scopoletin inhibits in vivo tumor vasculature in the xenograft model
plug sections showed that treatment with the high dose (200 mg/kg)
of scopoletin caused strong inhibition of vascularization in matrigel To evaluate in vivo inhibitory effect of scopoletin on tumor vascula-
plugs compared to the untreated control (Fig. 3E and F). The ture, xenografts of HCT-116 cells were implanted into immunocompro-
antineovascularization effect of scopoletin can be compared with that mised mice. Fig. 4A shows the representative animals from the negative
of the standard reference, imatinib (100 mg/kg). Fig. 3G and H shows control bearing enlarged tumors. Fig. 4B–D depicts the dose-dependent
the images of matrigel sections depicting the potent anti-angiogenic ef- suppression of growth of the tumor xenograft in scopoletin-treated
fect of imatinib. mice. All tested doses of scopoletin displayed a remarkable suppression
The mean blood vessel count of 10 low power fields in matrigel of HCT-116 tumor growth compared to that of the vehicle-treated con-
sections from representative groups is shown in a graphical representa- trol. Scopoletin displayed 33.01, 34.4, and 94.7% inhibition at 50, 100,
tion in Fig. 3I. The graph shows that the mean blood vessel count in the and 200 mg/kg doses, respectively. These results are comparable to
negative control group was 106 ± 8 and that treatment with scopoletin that of imatinib (Fig. 4E), which had a potent antitumor effect
significantly (p b 0.01) inhibited formation of new blood vessels in the with 84.3% inhibition at 100 mg/kg. Morphological investigation of the
matrigel implants, as the total mean count of blood vessels was 38 ± harvested tumors demonstrated a remarkable inhibitory effect of
4 and 10 ± 2 at 100 and 200 mg/kg, respectively. The value from scopoletin on tumor vasculature (Fig. 4F). The density of blood vessels
matrigel implants from mice treated with imatinib was 14 ± 3. in the tumors collected from the treated animals was drastically

Fig. 9. Visualization of ligands and protein interaction profile. Surface visualization of proteins and active site residue interactions of protein and hydrophobic interactions are shown in the
active pockets of the ligands: A: ERK1-scopoletin surface, LeadIT. B: ERK1-scopoletin active site residue interaction, LeadIT. C: VEGF-suramin surface, LeadIT. D: VEGF-suramin active site
residue interaction, LeadIT.
28 Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33

reduced compared to that of the tumors collected from the negative necrotic areas with reduced density of viable cells are visible as pink
control group. Fig. 5A shows the tumor growth profile in the respective colored regions in the tumor sections. Small regions of viable tissue
test groups. The average body weight for the respective groups is shown with cells can be seen in the treated tumor sections (Fig. 6D). Treat-
in Fig. 5B. ment with imatinib (Fig. 6G–H) resulted in significant (p b 0.01)
antitumorigenic activity and reduction in blood vessel density
Histological findings compared to the control. The mean blood vessel counts of 10 different
low power microscopic fields (Fig. 6I) in the tumor tissues from
Photomicrographs of all tumor sections excised from the mice were scopoletin-treated mice were 24 ± 6 and 11 ± 2 for the 100 and
stained with H&E. The sections were examined to quantify the blood 200 mg/kg doses, respectively. Negative control mice had a considerably
vessels and viable and necrotic tissue. Histological sections of the tu- higher number of blood vessels (84 ± 11).
mors from negative control mice (Fig. 6A) were full of growing tissue
with plenty of viable cells arranged compactly and resembling the Immunohistochemical findings
vigorous proliferation of cancerous cells. Higher magnification micro-
scopic evaluation revealed that the viable tissue was composed of highly Immunohistochemistry of blood-vessels is widely used in different
compact sheets of polygonal-shaped cells with prominent nuclei and areas of ongoing biomedical research, particularly within the scope of
numerous blood vessels (arrows in Fig. 6B). Either little or no necrotic tumor angiogenesis research. The techniques are based on immunohis-
areas were observed in the tissue sections of untreated mice (Fig. 6B). tochemical labeling of the endothelium by antibodies directed against
The tumors in the scopoletin treatments showed strong evidence of pan-endothelial markers such as von Willebrand factor, CD31, and
an anti-angiogenic effect (Fig. 6D–F). Due to reduced blood and oxygen CD34, followed by light microscopic evaluation. The qualitative assess-
supply, the tumors in scopoletin-treated animals exhibited loss of ment of the microvascular structures of the tissue leads to study the ex-
compactness of the viable cells with moderate to severe necrosis. The tent and nature of vasculature. CD34 is an antigen located in

Fig. 10. Visualization of ligands and protein interaction profile. Surface visualization of proteins and active site residue interactions of protein and hydrophobic interactions are shown in
the active pockets of the ligands: A: ERK1-suramin surface, LeadIT. B: ERK1-suramin active site residue interaction, LeadIT.
Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33 29

hematopoietic progenitor cells and endothelial cells. Anti-CD34 anti- Additionally, the antivascular property of scopoletin was further
body is a highly sensitive marker for endothelial cell differentiation confirmed by immunofluorescence staining with NG2-positive
and has also been used as a marker for vascular tumors (Vieira et al., areas in the histological sections. NG2 is a chondroitin sulfate proteo-
2005). In the present study, the tumor sections were subjected to im- glycan usually used to identify pericytes in tissue sections. It is
munohistochemical staining using CD34 antibody to locate the tumor expressed on the surface of nascent pericytes during the early stages
vasculature. The findings revealed that the tumor sections of negative of vasculogenic and angiogenic processes. Studies have reported the
control showed extensive vasculature with well-established networks role of tumor pericytes in promoting endothelial cells proliferation,
of blood vessels (arrows in Fig. 7A). Thus, the cellular architecture in differentiation and promoting the tumor vasculature through a vari-
negative control tumors found to be compact with live cells (Fig. 7A). ety of signaling networks (Raza et al., 2010). Findings of the present
Whereas, the positive control group treated with imatinib, showed sig- study showed that, in the negative control group (Fig. 7E), the area of
nificant inhibition of vasculature (Fig. 7B) and displayed necrosis (N). NG2-positive pericytes was abundant indicating the enriched vascu-
Similarly, scopoletin (100 mg/kg) exhibited enormous necrotic tissue lature in the tumor tissue. Whereas, treatment with the standard ref-
(N) and reduced vascularization (Fig. 7C), while at higher dose erence drug, imatinib demonstrated reduction in the fluorescent
(200 mg/kg), scopoletin demonstrated drastic inhibitory effect on signal (Fig. 7F). Similarly, the treatment with scopoletin revealed
tumor vascularization and necrosis (Fig. 7D). dose dependent antivascular effect in the immunohistological findings,

Fig. 11. Visualization of ligands and protein interaction profile. Surface visualization of proteins and active site residue interactions of protein and hydrophobic interactions are shown in
the active pockets of the ligands: A: FGF2-scopoletin surface, LeadIT. B: FGF2-scopoletin active site residue interaction, LeadIT.
30 Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33

as the area of NG2-positive pericytes was reduced with increasing the calculated upon docking of scopoletin with ERK1, FGF2, and VEGF
dose of scopoletin (Fig. 7G and H). were 0.32, 0.23, and 0.20 Kcal/mol per heavy atom (Table 1).
These results can be compared with those of the standard reference,
Scopoletin has potential binding efficiency toward ERK1, FGF2, and VEGF suramin. Table 1 also lists parameters such as match score, lipophil-
ic area score, ambiguous area score, and receptor–ligand interac-
The docking study results revealed that scopoletin displayed tion score used in the comparative evaluation of scopoletin and
more efficient activity than suramin. When the LeadIT FlexX meth- suramin.
od was used, scopoletin showed significant binding affinity toward Similar results were observed when the SYBYL Surflex dock method
ERK1, FGF2, and VEGF with docked energies (ΔG) of − 19, − 14, and was used. Table 2 illustrates the potent docking efficiency of scopoletin
− 11 Kcal/mol, respectively (Table 1). The ligand efficiencies with all tested pro-angiogenic ligands.

Fig. 12. Visualization of ligands and protein interaction profile. Surface visualization of proteins and active site residue interactions of protein and hydrophobic interactions are shown in
the active pockets of the ligands: A: VEGF-scopoletin surface, LeadIT. B: VEGF-scopoletin active site residue interaction, LeadIT.
Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33 31

Figs. 8–12 illustrate the amino acids (Leu124, Met125, Ala69, Val56, endothelial functions are highly controlled and regulated by the angio-
Leu173, and ll48) in the active pockets of the ligands (FGF2, ERK1 and genic mitogens. Therefore, anti-angiogenic compounds either directly
VEGF) displayed strong interactions with the phenyl group of interfere with endothelial cell functions or block the mitogen functions.
scopoletin, which indicates that the binding cavity of ERK1 might be a It has been reported that scopoletin possesses strong ability to inhibit
good fit for scopoletin. The analysis revealed that scopoletin was more the vital functions of endothelial cells, such as tube formation, differen-
significantly enfolded in the entire active pocket of the ligand compared tiation and migration (Pan et al., 2009). In the present study, an in vivo
to suramin (Figs. 8–10). The size of suramin might be the reason that it physiologic model of angiogenesis was employed using matrigel, an ex-
does not properly fit within any of the cavities of ERK1. tract composed of angiogenesis-inducing compounds. Assessment of
The overall results reveal that the docked energy and ligand binding angiogenesis in the matrigel implants was determined by scoring the
efficiency of scopoletin were more pronounced than those of the stan- histological sections for vascular density. The results revealed that
dard reference, suramin. The high molecular weight and bulky size of scopoletin showed clear evidence of inhibition of vascularization in
suramin may hamper effective binding and affinity with the ligands, matrigel plugs implanted in the treated mice.
particularly FGF2. This would explain the lack of interaction between To verify that the anti-angiogenic property of scopoletin can be use-
suramin and FGF2 in both docking analyses (Fig. 13). ful in suppression of tumor growth, a human tumor xenograft model
In the present study scopoletin demonstrated potent inhibitory ef- was employed. The in vivo tumor model demonstrated a clear inhibito-
fect on sprouting of microvessels in rat aortic explants. Rat aortic ring ry effect of scopoletin on the tumorigenesis process in tumor-bearing
assay is a 3-D quantitative ex vivo angiogenesis model, where develop- mice. In addition, the findings provided clear evidence of the anti-
ment of microvessels takes place via the main features of neovasculari- angiogenic property of scopoletin, as it significantly suppressed the vas-
zation similar to that observed in vivo. The microvessels that grow out cular supply to the growing tumor. It is well known that during tumor-
from aortic explants recruit chiefly the endothelial cells through prolif- igenesis the endothelial cells divide more rapidly and spread much
eration, migration and differentiation of the cells. General cytotoxic more quickly than they would during the normal course of blood vessel
compounds affect majority of the cells in aortic tissue and thereby kill formation (Maeshima et al., 2002). Several recent reports recommend-
the entire aortic tissue, whereas the antiangiogenic compounds selec- ed further studies of scopoletin to investigate its anticancer properties
tively target the endothelial functions. In the present study, in vitro pro- (Liu et al., 2012; Liu et al., 2001; Zhao et al., 2014). These preliminary
liferation test showed that scopoletin exhibited selective toxicity on studies reported the in vitro anticancer potential of scopoletin and dem-
endothelial cells which was evident through the highest selectivity onstrated that it could induce cell cycle arrest at the S and G2 phases and
index on other cell types (Kurdekar et al., 2014; Thani et al., 2010). activate apoptosis in tumor cells via the caspase pathway (Liu et al.,
The selective cytotoxicity of scopoletin on endothelial cells could be 2012; Liu et al., 2001; Zhao et al., 2014).
the principle reason for the anti-angiogenic effect on the rat aorta. The Malignant tumors require blood vessels for growth, and the main
aim of antiangiogenic therapy is to abolish the pathological vasculature
of tumor. However, recent data indicate that a suppression in tumor
vessels induced by antiangiogenic treatment using antiangiogenic
agents results in an inhibition of tumor growth but on the other hand
it permits tumor invasiveness (Pàez-Ribes et al., 2009). This increase
in tumor invasiveness is most probably due to increased hypoxia within
the tumor during antiangiogenic therapy (Rapisarda and Melillo, 2009).
Such tumor vessels are characterized by a leaky, disorganized and ab-
normal morphology (Mazzone et al., 2009). Subsequently, these patho-
logical tumor vessels impair blood flow into the tumor leading to a
hypoxic microenvironment rendering the tumor unresponsive to tradi-
tional chemotherapeutic agents. In addition, this abnormal phenotype
of tumor vasculature supports tumor progression and resistance to
treatment. However, Several studies have shown that antiangiogenic
agents prune the pathological vasculature and thus organize the con-
trolled and orderly arranged vasculature which helps to maintain a ho-
mogenous blood flow to the tumor tissue, and eventually leads to a
reduction in tumor hypoxia (Campbell et al., 2011; Russell et al.,
2015). Angiogenesis is an intricate process of signal transduction that
involves several mitogens (VEGF and FGF) and receptor protein kinases
(ERK1/2). Therefore, VEGF, FGF, and ERK1/2 are considered to be the
chief angiogenic factors that are essentially required for initiation and
prolongation of vascularization (Meadows et al., 2001). In the present
study, findings of the molecular docking analyses revealed that
scopoletin has the potential to interact with ERK1, VEGF-A, and FGF-2
and modulate their functions. These results support the findings of pre-
vious studies that reported the antagonistic effect of scopoletin toward
FGF-2 and ERK1/2 (Pan et al., 2009; Pan et al., 2011). However, Pan
et al. (2009) reported that scopoletin inhibits the phosphorylation of
ERK1/2, while the present study reported the binding and blocking effi-
ciencies of scopoletin toward ERK1/2.
In conclusion, the present work provides strong supporting evidence
that scopoletin possesses strong anti-angiogenesis activity in animal
models. Further, the inhibitory effect of scopoletin was exploited to
demonstrate its remarkable antitumorigenic activity in a human
Fig. 13. Graphical representation of the estimated binding activity (A) and ligand tumor xenograft model. Histological and immunohistochemical analy-
efficiency (B) of scopoletin and suramin with ERK1, FGF2, and VEGF in LeadIT. sis excised tumors revealed that scopoletin displayed drastic
32 Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33

suppression of tumor vasculature. In addition, the computational simu- Dahham, S.S., Tabana, Y.M., Hassan, L.E.A., Ahamed, M.B.K., Majid, A.S.A., Majid, A.M.S.A.,
2015b. In vitro antimetastatic activity of Agarwood (Aquilaria crassna) essential oils
lation models showed that scopoletin has strong binding efficiency with against pancreatic cancer cells. Alex. J. Med.
the angiogenic factors ERK1, VEGF-A, and FGF-2 and that it is configura- Eldridge, M.D., Murray, C.W., Auton, T.R., Paolini, G.V., Mee, R.P., 1997. Empirical scoring
tionally compatible with the active sites of the tested angiogenic li- functions: I. The development of a fast empirical scoring function to estimate the
binding affinity of ligands in receptor complexes. J. Comput. Aided Mol. Des. 11 (5),
gands. Thus, it can be concluded that the anti-angiogenic effect of 425–445.
scopoletin functions by regulating ERK1, VEGF-A, and FGF-2 signaling Florentine, S.K., Westbrooke, M.E., 2005. Invasion of the noxious weed Nicotiana glauca R.
pathways. Graham after an episodic flooding event in the arid zone of Australia. J. Arid Environ.
60 (4), 531–545.
Hassan, L.E., Ahamed, M.B., Majid, A.S., Baharetha, H., Muslim, N.S., Nassar, Z.D., Majid,
Conflict of interest A.M., 2014. Correlation of antiangiogenic, antioxidant and cytotoxic activities of
some Sudanese medicinal plants with phenolic and flavonoid contents. BMC Comple-
ment. Altern. Med. 14 (1), 406.
The authors declare no conflict of interest. Ibrahim, B.A., 2012. Phytochemistry of Nicotiana glauca & Some Biotransformation
Reactions.
Jones, G., Willett, P., Glen, R.C., Leach, A.R., Taylor, R., 1997. Development and validation of
Submission declaration a genetic algorithm for flexible docking. J. Mol. Biol. 267 (3), 727–748.
Kang, S.Y., Sung, S.H., Park, J.H., Kim, Y.C., 1998. Hepatoprotective activity of scopoletin, a
The authors declare that the work described has not been published constituent of Solanum lyratum. Arch. Pharm. Res. 21 (6), 718–722.
Khadeer Ahamed, M.B., Krishna, V., Dandin, C.J., 2010. In vitro antioxidant and in vivo pro-
previously. phylactic effects of two γ-lactones isolated from Grewia tiliaefolia against hepatotox-
icity in carbon tetrachloride intoxicated rats. Eur. J. Pharmacol. 631 (1–3), 42–52.
http://dx.doi.org/10.1016/j.ejphar.2009.12.034.
Author contributions
Kinoshita, K., Kono, H., Yura, K., 2008. Prediction of Molecular Interactions From 3D-
Structures: From Small Ligands to Large Protein Complexes. Prediction of Protein
Tabana, Amin, Hassan and Ahamed designed the experiments. Structures, Functions, and Interactions, pp. 159–186.
Kurdekar, R.R., Hegde, G.R., Kulkarni, M.V., Mulgund, G.S., 2014. Isolation and Characteri-
Tabana and Hassan carried out the isolation of the compound. Tabana
zation of Scopoletin—An Anticancerous Compound From the Bark of Hymenodictyon
and Dahham conducted in vitro biological experiments. Iqbal per- obovatum Wall.
formed the structural elucidation of the compound. Tabana and Saeed Liu, X.L., Zhang, L., Fu, X.L., Chen, K., Qian, B.C., 2001. Effect of scopoletin on PC3 cell pro-
carried out HPLC analysis. Khan conducted the molecular docking liferation and apoptosis. Acta Pharmacol. Sin. 22 (10), 929–933.
Liu, W., Hua, J., Zhou, J., Zhang, H., Zhu, H., Cheng, Y., Gust, R., 2012. Synthesis and in vitro
study. Tabana and Hassan carried out the in vivo study. Chern Ein Oon antitumor activity of novel scopoletin derivatives. Bioorg. Med. Chem. Lett. 22 (15),
conducted immunohistochemistry of tumor vessel analysis. Doblin 5008–5012.
Sandai help in the revision, Tabana, Ahamed and Dahham interpreted Maeshima, Y., Sudhakar, A., Lively, J.C., Ueki, K., Kharbanda, S., Kahn, C.R., ... Kalluri, R.,
2002. Tumstatin, an endothelial cell-specific inhibitor of protein synthesis. Science
the results. Tabana, Ahamed, Amin and Aman drafted the manuscript. 295 (5552), 140–143.
All the authors read and approved the final manuscript. Mandal, K., Kent, S.B., 2011. Total chemical synthesis of biologically active vascular endo-
thelial growth factor. Angew. Chem. 123 (35), 8179–8183.
Mazzone, M., Dettori, D., de Oliveira, R.L., Loges, S., Schmidt, T., Jonckx, B., ... de Almodovar,
Acknowledgments C.R., 2009. Heterozygous deficiency of PHD2 restores tumor oxygenation and inhibits
metastasis via endothelial normalization. Cell 136 (5), 839–851.
Mdee, L.K., Masoko, P., Ellof, J.N., 2009. The activity of extracts of seven common invasive
The first author would like to sincerely acknowledge University
plant species on fungal phytopathogens. S. Afr. J. Bot. 75 (2), 375–379.
Sains Malaysia (USM) for postgraduate fellowship. All authors acknowl- Meadows, K.N., Bryant, P., Pumiglia, K., 2001. Vascular endothelial growth factor induc-
edge the Ministry of Agriculture, Malaysia under NRGS grant no: 304/ tion of the angiogenic phenotype requires Ras activation. J. Biol. Chem. 276 (52),
PFARMASI/650583/K123. The authors also wish to acknowledge 49289–49298.
Meng, E.C., Shoichet, B.K., Kuntz, I.D., 1992. Automated docking with grid-based energy
Universiti Sains Malaysia (USM) for research university team grant no. evaluation. J. Comput. Chem. 13 (4), 505–524.
1001/PFARMASI/851001. The authors would like to acknowledge Muegge, I., Martin, Y.C., 1999. A general and fast scoring function for protein–ligand inter-
Cheng Wei Kang for help in optimization of antibody staining for CD34. actions: a simplified potential approach. J. Med. Chem. 42 (5), 791–804.
Nassar, Z.D., Aisha, A., Ahamed, M., Ismail, Z., Abu-Salah, K.M., Alrokayan, S.A., Majid, A.,
2011. Antiangiogenic properties of Koetjapic acid, a natural triterpene isolated from
Appendix A. Supplementary data Sandoricum koetjaoe Merr. Cancer Cell Int. 11 (1), 12.
Oliveira, E., Romero, M., Silva, M., Silva, B., Medeiros, I., 2001. Intracellular calcium mobiliza-
tion as a target for the spasmolytic action of scopoletin. Planta Med. 67 (07), 605–608.
Supplementary data to this article can be found online at http://dx. Pàez-Ribes, M., Allen, E., Hudock, J., Takeda, T., Okuyama, H., Viñals, F., ... Casanovas, O.,
doi.org/10.1016/j.mvr.2016.04.009. 2009. Antiangiogenic therapy elicits malignant progression of tumors to increased
local invasion and distant metastasis. Cancer Cell 15 (3), 220–231.
Pan, R., Dai, Y., Yang, J., Li, Y., Yao, X., Xia, Y., 2009. Anti-angiogenic potential of
References scopoletin is associated with the inhibition of ERK1/2 activation. Drug Dev.
Res. 70 (3), 214–219.
Ahamed, M.B.K., Aisha, A.F., Nassar, Z.D., Siddiqui, J.M., Ismail, Z., Omari, S., ... Majid, A.A., Pan, R., Gao, X., Lu, D., Xu, X., Xia, Y., Dai, Y., 2011. Prevention of FGF-2-induced angiogen-
2012. Cat's whiskers tea (Orthosiphon stamineus) extract inhibits growth of colon esis by scopoletin, a coumarin compound isolated from Erycibe obtusifolia Benth, and
tumor in nude mice and angiogenesis in endothelial cells via suppressing VEGFR its mechanism of action. Int. Immunopharmacol. 11 (12), 2007–2016.
phosphorylation. Nutr. Cancer 64 (1), 89–99. Passaniti, A., Taylor, R.M., Pili, R., Guo, Y., Long, P.V., Haney, J.A., ... Martin, G.R., 1992. A
Al-Salahi, O.S.A., Kit-Lam, C., Majid, A.M.S.A., Al-Suede, F.S.R., Saghir, S.A.M., Abdullah, simple, quantitative method for assessing angiogenesis and antiangiogenic agents
W.Z., ... Yusoff, N.M., 2013. Anti-angiogenic quassinoid-rich fraction from Eurycoma using reconstituted basement membrane, heparin, and fibroblast growth factor.
longifolia modulates endothelial cell function. Microvasc. Res. 90, 30–39. Lab. Investig. 67 (4), 519–528.
Beh, H.-K., Seow, L.-J., Asmawi, M.Z., Abdul Majid, A.M.S., Murugaiyah, V., Ismail, N., Ismail, Rapisarda, A., Melillo, G., 2009. Role of the hypoxic tumor microenvironment in the resis-
Z., 2012. Anti-angiogenic activity of Morinda citrifolia extracts and its chemical con- tance to anti-angiogenic therapies. Drug Resist. Updat. 12 (3), 74–80.
stituents. Nat. Prod. Res. 26 (16), 1492–1497. Rarey, M., Kramer, B., Lengauer, T., Klebe, G., 1996. A fast flexible docking method using an
Böhm, H.-J., 1998. Prediction of binding constants of protein ligands: a fast method for the incremental construction algorithm. J. Mol. Biol. 261 (3), 470–489.
prioritization of hits obtained from de novo design or 3D database search programs. Raza, A., Franklin, M.J., Dudek, A.Z., 2010. Pericytes and vessel maturation during tumor
J. Comput. Aided Mol. Des. 12 (4), 309-309. angiogenesis and metastasis. Am. J. Hematol. 85 (8), 593–598.
Campbell, N., Greenaway, J., Henkin, J., Petrik, J., 2011. ABT-898 induces tumor regression Russell, S., Duquette, M., Liu, J., Drapkin, R., Lawler, J., Petrik, J., 2015. Combined therapy
and prolongs survival in a mouse model of epithelial ovarian cancer. Mol. Cancer with thrombospondin-1 type I repeats (3TSR) and chemotherapy induces regression
Ther. 10 (10), 1876–1885. and significantly improves survival in a preclinical model of advanced stage epithelial
Cao, R., Ji, H., Feng, N., Zhang, Y., Yang, X., Andersson, P., ... Dissing, S., 2012. Collaborative ovarian cancer. FASEB J. 29 (2), 576–588.
interplay between FGF-2 and VEGF-C promotes lymphangiogenesis and metastasis. Shaw, C.Y., Chen, C.H., Hsu, C.C., Chen, C.C., Tsai, Y.C., 2003. Antioxidant properties of
Proc. Natl. Acad. Sci. 109 (39), 15894–15899. scopoletin isolated from Sinomonium acutum. Phytother. Res. 17 (7), 823–825.
Dahham, S.S., Tabana, Y.M., Iqbal, M.A., Ahamed, M.B., Ezzat, M.O., Majid, A.S., Majid, A.M., http://dx.doi.org/10.1002/ptr.1170.
2015a. The anticancer, antioxidant and antimicrobial properties of the sesquiterpene Tabana, Y.M., Dahham, S.S., Hassan, L.E.A., Al-Mansoub, M.A., Taleb-Agha, M., Majid,
β-caryophyllene from the essential oil of Aquilaria crassna. Molecules 20 (7), A.M.A., 2015. In vitro anti-metastatic and antioxidant activity of Nicotiana glauca frac-
11808–11829. tion against breast cancer cells. Adv. Biol. Res. 9 (2), 95–102.
Y.M. Tabana et al. / Microvascular Research 107 (2016) 17–33 33

Thani, W., Vallisuta, O., Siripong, P., Ruangwises, N., 2010. Anti-proliferative and antioxi- Wang, S., Zheng, Z., Weng, Y., Yu, Y., Zhang, D., Fan, W., ... Hu, Z., 2004. Angiogenesis and
dative activities of Thai noni/Yor (Morinda citrifolia Linn.) leaf extract. Southeast anti-angiogenesis activity of Chinese medicinal herbal extracts. Life Sci. 74 (20),
Asian J. Trop. Med. Public Health 41 (2), 482–489. 2467–2478.
Vieira, S.C., Silva, B.B., Pinto, G.A., Vassallo, J., Moraes, N.G., Santana, J.O., ... Zeferino, L.C., Zhao, P., Chen, L., Li, L.-H., Wei, Z.-F., Tong, B., Jia, Y.-G., ... Xia, Y.-F., 2014. SC-III3, a novel
2005. CD34 as a marker for evaluating angiogenesis in cervical cancer. Pathol. Res. scopoletin derivative, induces cytotoxicity in hepatocellular cancer cells through
Pract. 201 (4), 313–318. oxidative DNA damage and ataxia telangiectasia-mutated nuclear protein kinase
Wang, K., Zheng, J., 2012. Signaling regulation of fetoplacental angiogenesis. J. Endocrinol. activation. BMC Cancer 14 (1), 987.
212 (3), 243–255.

You might also like