62
Views
10
CrossRef citations to date
0
Altmetric
Original Research

Antiangiogenic effects of AA-PMe on HUVECs in vitro and zebrafish in vivo

, , , , &
Pages 1871-1884 | Published online: 04 Apr 2018

Abstract

Angiogenesis plays a vital role in many physiological and pathological processes and several diseases are connected with its dysregulation. Asiatic acid (AA) has demonstrated anticancer properties and we suspect this might be attributable to an effect on angio-genesis. A modified derivative of AA, N-(2α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester (AA-PMe), has improved efficacy over its parent compound, but its effect on blood vessel development remains unclear.

Methods

In this study, we investigated the antiangiogenic activity of AA and AA-PMe in zebrafish embryos and human umbilical vein endothelial cells (HUVECs). First of all, we treated HUVECs with increasing concentrations of AA-PMe or AA, with or without vascular endothelial growth factor (VEGF) present, and assessed cell viability, tube formation, and cell migration and invasion. Quantitative real-time polymerase chain reaction and Western blot analysis were later used to determine the role of vascular endothelial growth factor receptor 2 (VEGFR2)-mediated signaling in AA-PMe inhibition of angiogenesis. We extended these studies to follow angiogenesis using Tg(fli:EGFP) transgenic zebrafish embryos. For these experiments, embryos were treated with varying concentrations of AA-PMe or AA from 24 to 72 hours postfertilization prior to morphological observation, angiogenesis assessment, and endogenous alkaline phosphatase assay. VEGFR2 expression in whole embryos following AA-PMe treatment was also determined.

Results

We found AA-PMe decreased cell viability and inhibited migration and tube formation in a dose-dependent manner in HUVECs. Similarly, AA-PMe disrupted the formation of intersegmental vessels, the dorsal aorta, and the posterior cardinal vein in zebrafish embryos. Both in vitro and in vivo AA-PMe surpassed AA in its ability to block angiogenesis by suppressing VEGF-induced phosphorylation of VEGFR2 and disrupting downstream extracellular regulated protein kinase and AKT signaling.

Conclusion

For the first time, this study reveals that AA-PMe acts as a potent VEGFR2 kinase inhibitor and exerts powerful antiangiogenic activity, suggesting it to be a promising therapeutic candidate for further research.

Introduction

Angiogenesis, the process by which new blood vessels sprout from preexisting endothelium, is essential for various biological processes including embryonic vascular development and differentiation, wound healing, and organ regeneration.Citation1,Citation2 Concurrently, angiogenesis regulates multiple pathological processes including tumor progression,Citation1Citation6 infection,Citation2 ischemic and inflammatory diseases,Citation2 metastasis disorder,Citation1 age-related macular degeneration,Citation3,Citation5 and diabetic retinopathy.Citation1,Citation5,Citation7 Angiogenesis normally occurs under conditions in which pro- and antiangiogenic factors are in balance, maintaining homeostasis. In contrast, the tumor microenvironment promotes angiogenesis: hypoxic conditions within the tumor spur the expression and release of growth factors and cytokines that induce proliferation and migration of endothelial cellsCitation8,Citation9 and stimulate blood vessel growth to increase the local blood supply.Citation10

Vascular endothelial growth factor (VEGF) is a critical angiogenic factor well-documented for its mitogenic activity in endothelial cells and its role in promoting normal physiological as well as pathological neovascularization, such as that occurring in tumor angiogenesis.Citation3,Citation11 VEGF activates receptor tyrosine kinases in endothelial cells by binding one of two receptors: vascular endothelial growth factor receptor 1 (VEGFR1) or 2 (VEGFR2).Citation12 VEGFR2 serves as the primary receptor and activates downstream signaling molecules including focal adhesion kinase, extracellular signal-regulated kinase (ERK), AKT/protein kinase B (PKB), mammalian target of rapamycin, and ribosomal protein S6 kinase (p70S6K).Citation13 Under normal conditions, this results in the growth, migration, differentiation, and survival of endothelial cells in preexisting vasculature, but its activation can also be hijacked to drive pathologic angiogenesis.Citation14 The VEGFR2 signaling pathway is thus a promising target for inhibiting angiogenesis in disease states.

At present, several VEGFR inhibitors have been used in Phase I and II clinical trials, including drugs aimed at VEGF or VEGFRs:Citation15 bevacizumab, sunitinib malate, and sorafenib have all been approved by the US Food and Drug Administration for cancer treatment.Citation16,Citation17 However, clinical use of these treatments is restricted because they carry the risk of severe side effects including hypertension, bleeding, and gastrointestinal perforation.Citation18,Citation19 To identify safer and more efficient treatments for angiogenesis-related diseases attention is turning to small molecule therapeutic strategies targeting signal transduction pathways downstream of VEGFR2.

Consequently, there is increasing interest in natural products that contain a variety of antiangiogenic compounds and have a history of safe usage, including many Chinese herbs. Asiatic acid (2α,3β,23-trihydroxyurs-12-ene-28-oic acid; AA), a pentacyclic triterpenoid derived from the tropical medicinal plant Centella asiatica,Citation20 is one such promising candidate. This study examines the antiangiogenic efficacy of N-(2α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester (AA-PMe), which was semi-synthesized in our laboratory with the goal of improving its antiangiogenic efficacy. Unmodified AA has been reported to aid in wound healingCitation21,Citation22 as well as inhibit UV-induced photoaging,Citation23 glutamate- and β-amyloid-induced neurotoxicity,Citation24Citation27 and hepatofibrosis.Citation28 It has also demonstrated anticancerCitation29Citation33 and antiangiogenic efficacy.Citation34 Based on the antiangiogenic activity of AA,Citation34 we researched the effects of AA-PMe on in vitro and in vivo angiogenesis and sought to determine the underlying mechanism.

In human umbilical vein endothelial cells (HUVECs) treated with AA-PMe we observed apoptosis induction as well as inhibition of cell migration and tube formation, so we decided to further explore these effects in an in vivo system. Zebrafish embryos are an established and efficient model for investigating vascular development and represent a practical and efficient system for screening angiogenesis drugs.Citation35 The recent development of the transgenic zebrafish line Tg(fli1:EGFP), which carries a genetic modification that fluorescently labels blood vessels, represents a useful genetic model to accelerate angiogenesis research.Citation36,Citation37 We utilized this transgenic line to examine the impact of AA-PMe on angiogenesis and found a profound inhibitory effect. Our data further indicate that AA-PMe exerts its antiangiogenic activity through inhibition of VEGFR2-mediated signaling.

Materials and methods

Ethics statement

All animal experiments in this investigation were conducted in compliance with the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health (NIH publication no 85-23, revised 1996) and approved by the Animal Care and Use Committee of Nanjing Normal University, China (permit number 2090658).

Chemicals and reagents

AA was extracted from C. asiatica (L.) Urban and structurally modified in our laboratory to produce AA-PMe.Citation20 AA and AA-PMe were both dissolved in 1% dimethyl sulfoxide (DMSO; Sigma-Aldrich Co, St Louis, MO, USA) and their structures are presented in . Endogenous alkaline phosphatase (EAP) staining was tested by a phosphatase substrate kit (Pierce; Thermo Fisher Scientific, Waltham, MA, USA). Recombinant human VEGF was purchased from Thermo Fisher Scientific. Primary antibodies for total Akt, pAkt-ser473, total ERK1/2, p-pERK1/2, total VEGFR2, and pVEGFR2-Tyr1175 were brought from Cell Signaling Technology (Danvers, MA, USA). GAPDH and tubulin antibody were from Abcam (Cambridge, UK).

Figure 1 Chemical structure of AA and AA-PMe. (A) 2α,3β,23-Trihydroxyurs-12-ene-28-oic acid, AA; C30H48O5, MW 488.70 g/mol; (B) N-(2 α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester, AA-PMe; C42H63NO9, MW 725.95 g/mol.

Abbreviations: AA, asiatic acid; MW, molecular weight.
Figure 1 Chemical structure of AA and AA-PMe. (A) 2α,3β,23-Trihydroxyurs-12-ene-28-oic acid, AA; C30H48O5, MW 488.70 g/mol; (B) N-(2 α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester, AA-PMe; C42H63NO9, MW 725.95 g/mol.

Cell culture

HUVECs were purchased from American Type Culture Collection (Manassas, VA, USA) and maintained in complete growth medium (Dulbecco’s Modified Eagle’s Medium: F12) supplemented with 10% fetal bovine serum (FBS; Thermo Fisher Scientific), 0.1 mg/mL heparin, and 0.03%–0.05% mg/mL endothelial cell growth supplement.Citation38 Cells were incubated at 37°C in a humidified atmosphere with 5% CO2.

Zebrafish

Transgenic zebrafish Tg(fli:EGFP) with enhanced green fluorescent protein (EGFP) expressing endothelial cells and wild-type zebrafish (Tuebingen line) were obtained from Model Animal Research Center of Nanjing University. Adult zebrafish were maintained at 28.5°C, pH 7±0.2 H2O in a 14:10 hour light/dark photoperiod and fed with live brine shrimp once a day and dry food twice a day.Citation39 Embryos were produced by pairwise mating using a fish hatch box and were subsequently maintained at 28.5°C in E3 embryo medium: 5 mM NaCl, 0.17 mM KCl, 0.33 mM CaCl2, and 0.33 mM MgSO4 in dH2O with added antifungal solvent (0.01% methylene blue).Citation40 Embryos exhibiting normal development were dechorionated by forceps prior to drug treatment. All zebrafish studies were approved by the Institutional Animal Care and Use Committee at the Nanjing University of Technology.

In vitro viability assay

Proliferation assays were performed as previously described.Citation41 Briefly, 8000 HUVECs/well were seeded in 96-well plates and allowed to attach for 12 hours. Cells were then treated for 24 hours with various concentrations of AA and AA-PMe or 0.1% DMSO vehicle control. Cell proliferation was measured by cell counting kit (CCK)-8 colorimetric assay according to the manufacturer’s protocol (Vazyme Biotech, Nanjing, China).

In vitro migration assay

HUVEC migration assay was performed using the wound healing method as previously described.Citation42Citation44 Briefly, HUVECs were allowed to grow to full confluence in 6-well plates, manually disrupted by pipette tips, and washed twice with media to remove detached cells. Photomicrographs of initial wounds were taken at 100× magnification using a phase-contrast microscope and wound distance was measured using AxioVision Rel.4.7 software (Carl Zeiss Meditec AG, Jena, Germany). Cells were then treated with VEGF (10 ng/mL) and DMSO vehicle or the indicated concentrations of AA-PMe or AA and photomicrographs were taken at the indicated time points. After vehicle-treated HUVECs migrated to completely close the wound the experiment was terminated.

Migrationdistance=InitialdistanceFinaldistance2

In vitro Transwell invasion assay

To determine cell invasion, Transwell plates (Costar; Corning Incorporated, Corning, NY, USA) pre-coated with Matrigel (BD Biosciences, San Jose, CA, USA) were placed on 24-well plates and the lower chamber was filled with 600 μL culture medium supplemented with VEGF (10 ng/mL) as a chemoattractant and vehicle or the indicated concentrations of AA-PMe or AA were added. Then, 1×105 HUVECs in 200 μL RPMI containing 1% FBS were added to the upper chamber. After 24 hours, non-migrated cells were removed with cotton swabs while cells that traversed the membrane and spread to the lower surface of the filters were fixed with 90% alcohol for 30 minutes at room temperature and stained with 0.1% crystal violet for visualization. Then, 100× images were captured on a phase-contrast microscope, imaged using AxioVision Rel.4.7 software, and invasive cells were quantified by manual counting. Thereafter, invasive cells were decolored with 33% acetic acid for 10 minutes. Samples were transferred to 96-well plates and the absorbance at 570 nm, indicative of the number of cells present, was measured with a microplate reader (Thermo Fisher Scientific). The percentage of invasion inhibition by AA-PMe or AA was calculated by dividing the difference between the absorbance of treated and untreated groups and multiplying by 100:

Percentageofinvasioninhibition(%)=ODtreatedODblankODcontrolODblank×100%

In vitro tube formation assay

To examine the effect of AA-PMe and AA on in vitro angiogenesis tube formation assays were performed as previously described.Citation45 Briefly, 150 μL/well growth factor-reduced Matrigel was added to pre-chilled 24-well plates and polymerized for 45 minutes at 37°C. HUVECs (4×104) in complete media were then added to the Matrigel-coated plates. In the first part, varying concentrations of DMSO, AA-PMe, or AA were either added immediately or 6 hours after cells were seeded to allow initial capillary network formation. In the second part, varying concentrations of DMSO, AA-PMe, or AA were added 10 hours after HUVECs seeding. For further verifying the effect of AA-PMe on VEGF-stimulated tube formation, in the third experiment, HUVECs were washed twice with media containing 0.5% serum before seeding and plated with or without 10 ng/mL purified VEGF (Cell Signaling Tech nology) added in addition to AA-PMe or AA. For all experiments, tubular structures were photographed after 6 hours of treatment at 100× magnification using a phase-contrast microscope and were measured using AxioVision Rel.4.7 software.

Quantitative EAP assay

Quantitative EAP assay was performed as previously described using a Pierce phosphatase substrate kit (Thermo Fisher Scientific) according to the manufacturer’s protocol.Citation46 Briefly, one 24-hour postfertilization (hpf) zebrafish embryo was added to each well of a 96-well plate, in 100 μL E3 embryo medium. DMSO vehicle or various concentrations of AA and AA-PMe were added and embryos were incubated at 28.5°C for 48 hours. Embryos at 72 hpf were treated with increasing concentrations of ethanol for dehydration purpose.

Embryos were washed three times with diethanolamine buffer and stained according to the manufacturer’s protocol; the reaction was stopped by the addition of 50 μL 2M NaOH and optical density was measured at 405 nm using a microplate reader. Percent vessel formation was calculated as the difference in optical density compared to the untreated control.

Vesselformation(%)=ODtreatedday3ODcontrolday1ODcontrolday3ODcontrolday1×100%

Assessment of blood vessel changes in whole zebrafish embryos

Following drug treatment, zebrafish embryos were anesthetized with 0.016% tricaine (Sigma-Aldrich Co). Embryo intersegmental blood vessel (ISV) and subintestinal vessel (SIV) plexus development were observed and imaged at 48 and 72 hpf, respectively (100× magnification), using a fluorescence microscope (IX71; Olympus Corporation, Tokyo, Japan).

Western blot analysis

The effects of AA-PMe and AA on intracellular signaling cascades were determined by Western blot. Zebrafish embryos were treated with DMSO control or the indicated concentrations of AA-PMe or AA from 24 to 72 hpf at 28.5°C; HUVEC cells were treated with AA-PMe or AA for 24 hours at 37°C. Cells and whole embryos were lysed for 30 minutes on ice in RIPA buffer (0.01% phenylmethane sulfonyl fluoride [Sigma-Aldrich Co], 150 mM NaCl, 50 mM Tris [pH =8], 0.1% sodium dodecyl sulfate, 0.2% ethylenediaminetetraacetic acid, 1% Triton X-100, 1% sodium deoxycholate), supplemented with protease and phosphatase inhibitors (Hoffman-La Roche Ltd, Basel, Switzerland). Lysates were cleared by centrifugation at 12,000 rpm at 4°C for 15 minutes and protein concentration was determined by bicinchoninic acid assay.

For each sample, 30–50 μg protein was run on a 10% agarose gel, then transferred to polyvinylidene fluoride membranes (Merck Millipore, Billerica, MA, USA) using a semidry transfer system (Bio-Rad Laboratories Inc, Hercules, CA, USA). Membranes were blocked with 5% nonfat milk in Tris buffered saline (TBST) containing 0.05% Tween-20 for 1 hour at room temperature, washed, and incubated overnight with primary antibodies (R&D) at recommended dilutions in TBST milk. After washing, blots were probed with horseradish peroxidase-conjugated secondary antibody (Abcam) for 1 hour, washed, and protein expression was detected by chemiluminescence using a Tanon-3500R image system (Tanon, Shanghai, China).

Quantitative real-time polymerase chain reaction (qRT-PCR)

Transcriptional effects of AA and AA-PMe on VEGFR2 were determined by qRT-PCR. Embryos and HUVECs were treated as described for Western blots; then, total mRNA was extracted using Trizol reagent (Thermo Fisher Scientific) according to the manufacturer’s protocol. Extracted RNA integrity was assessed by agarose-gel electrophoresis and quantified by OD260/OD280 absorbance. cDNA was reverse-transcribed from RNA using a PrimeScript™ RT reagent kit (TaKaRa, Kyoto, Japan) and qRT-PCR was performed with the SYBR PrimeScript RT-PCR kit (TaKaRa) on a StepOne Plus™ system (Thermo Fisher Scientific) according to the manufacturer’s instructions. Primer sequences are listed in . mRNA expression was calculated by the ΔΔCt method and quantified relative to GAPDH mRNA.

Table 1 Primer sequences for real-time PCR

Statistical analysis

All experiments were performed at least three times and values are given as mean ± SEM. Data were analyzed using GraphPad Prism 6.0 software (GraphPad Software, Inc, La Jolla, CA, USA) and statistical significance was assessed by T-test. P-values <0.05 were considered significant.

Results

AA-PMe decreases endothelial cell viability

To determine the effect of AA-PMe on endothelial cells HUVECs were treated with various doses of AA-PMe or AA (5, 10, and 25 μM), and the degree of cell death was determined at various time points (24, 48, and 72 hours) by CCK-8 assay. Both compounds demonstrated dose-dependent effects on cell viability over time, but the inhibitory effect of AA-PMe was much stronger (). It was observed that 10 and 25 μM AA-PMe reduced viability by as much as 20% after 24 hours, and after 72 hours viability decreased in response to all concentrations, falling by 23.0%–39.7% in a dose-dependent manner. AA significantly decreased viability by 20.8% and 24.1% after 48 and 72 hours only at its highest dose of 25 μM.

Figure 2 AA-PMe inhibits HUVEC viability. HUVECs were treated with DMSO vehicle or varying concentrations (5, 10, or 25 μM) of (A) AA or (B) AA-PMe for 24, 48, and 72 hours. Both adherent and non-adherent cells were collected and processed for determination of viable cell number. Mean values ± SEM are shown, n=3. *P<0.05.

Abbreviations: AA, asiatic acid; AA-PMe, N-(2α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester; HUVEC, human umbilical vein endothelial cell; DMSO, dimethyl sulfoxide.
Figure 2 AA-PMe inhibits HUVEC viability. HUVECs were treated with DMSO vehicle or varying concentrations (5, 10, or 25 μM) of (A) AA or (B) AA-PMe for 24, 48, and 72 hours. Both adherent and non-adherent cells were collected and processed for determination of viable cell number. Mean values ± SEM are shown, n=3. *P<0.05.

AA-PMe inhibits VEGF-stimulated chemotactic motility and capillary structure formation in HUVECs

EC migration and invasion is essential for neo-angiogenesis and critical for tumor growth.Citation47 VEGF is the most important pro-angiogenic factor and is known to enhance proliferation, survival, and capillary tube formation by endothelial cells.Citation14 Accordingly, we investigated the effect of AA-PMe and AA treatment on VEGF-stimulated endothelial cell migration and invasion using wound healing and Transwell assays. Similar to our cell viability assays, both AA-PMe and AA inhibited wound healing and Matrigel invasion compared to VEGF-only controls, but AA-PMe showed a much stronger effect in both settings. After 6 hours treatment with 5, 10, and 25 μM doses, AA-PMe decreased migration distance by 44%, 62%, and 66% compared to 19%, 48%, and 57% migration inhibition by the same concentrations of AA (). The decrease in migration distance was even more pronounced after 12 hours, with AA-PMe treatment reducing the migrated distance by 34%, 56%, and 69%, compared to 25%, 38%, and 43% reductions by AA. Transwell experiments yielded even more striking results: after 24 hours, 5, 10, and 25 μM AA-PMe inhibited HUVEC invasion through Matrigel by 44%, 66%, and 83%, respectively, compared to 27%, 41%, and 54% inhibition by AA at the same concentrations ().

Figure 3 AA-PMe inhibits HUVEC migration. The effect of the indicated doses of AA-PMe or AA on the migratory potential of VEGF-treated HUVECs was analyzed by wound healing assay. (A) Representative photomicrographs of initial and final wounds are shown at 100× magnification. (B) Migration distance was calculated as described in the “Materials and methods” section. Mean values ± SEM are shown, n=3. *P<0.05. **P<0.01.

Abbreviations: AA, asiatic acid; AA-PMe, N-(2α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester; HUVEC, human umbilical vein endothelial cell; VEGF, vascular endothelial growth factor.
Figure 3 AA-PMe inhibits HUVEC migration. The effect of the indicated doses of AA-PMe or AA on the migratory potential of VEGF-treated HUVECs was analyzed by wound healing assay. (A) Representative photomicrographs of initial and final wounds are shown at 100× magnification. (B) Migration distance was calculated as described in the “Materials and methods” section. Mean values ± SEM are shown, n=3. *P<0.05. **P<0.01.

Figure 4 AA-PMe inhibits HUVEC invasion. The invasive potential of HUVECs treated with VEGF and the indicated doses of AA-PMe and AA was determined using invasion chambers as detailed in the “Materials and methods” section. (A) Representative images of invaded cells stained with crystal violet are shown at 100× magnification. (B) Inhibition of invasion in treated cells was determined by absorbance as described. (C) Inhibition of invasion by AA and AA-PMe was calculated by absorption of acetic acid elution. Mean ± SEM of three samples per treatment is depicted. *P<0.05. **P<0.01.

Abbreviations: AA, asiatic acid; AA-PMe, N-(2α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester; HUVEC, human umbilical vein endothelial cell; VEGF, vascular endothelial growth factor.
Figure 4 AA-PMe inhibits HUVEC invasion. The invasive potential of HUVECs treated with VEGF and the indicated doses of AA-PMe and AA was determined using invasion chambers as detailed in the “Materials and methods” section. (A) Representative images of invaded cells stained with crystal violet are shown at 100× magnification. (B) Inhibition of invasion in treated cells was determined by absorbance as described. (C) Inhibition of invasion by AA and AA-PMe was calculated by absorption of acetic acid elution. Mean ± SEM of three samples per treatment is depicted. *P<0.05. **P<0.01.

Another crucial step during neo-angiogenesis is the formation and merging of endothelial cell tubes to form a complex network of blood vessels. To understand the effects of AA-PMe and AA on early tube formation we cultured HUVECs in three-dimensional Matrigel and examined their structure after 6 hours. While untreated HUVECs formed a massive network of well-branched tubes after 6 hours, this was disrupted by treatment with 5–25 μM AA-PMe in a dose-dependent manner; however, AA had only minor effects on network formation even at 25 μM (). AA-PMe treatment also decreased the branch number by ~17%, 46%, and 65% at 5, 10, and 25 μM concentrations. Despite its weaker ability to disrupt tube formation, AA treatment did reduce branching by 10%, 30%, and 42% at the same doses ().

Figure 5 AA-PMe inhibits capillary structure formation in HUVECs. The effect of the indicated doses of AA-PMe and AA on tube formation was determined by observing HUVEC growth on Matrigel. Tubular structures were photographed at 100× magnification and branch point numbers were determined as described in the “Materials and methods” section. The effects of AA-PMe and AA on (A, D) early tube formation, (B, E) preformed HUVEC tubes, and (C, F) VEGF-treated cells were evaluated. Representative phase-contrast images at 100× magnification from two to three independent experiments are shown. Mean ± SEM of three samples per treatment is shown for percent inhibition of branching. *P<0.05, **P<0.01.

Abbreviations: AA, asiatic acid; AA-PMe, N-(2α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester; HUVEC, human umbilical vein endothelial cell; VEGF, vascular endothelial growth factor.
Figure 5 AA-PMe inhibits capillary structure formation in HUVECs. The effect of the indicated doses of AA-PMe and AA on tube formation was determined by observing HUVEC growth on Matrigel. Tubular structures were photographed at 100× magnification and branch point numbers were determined as described in the “Materials and methods” section. The effects of AA-PMe and AA on (A, D) early tube formation, (B, E) preformed HUVEC tubes, and (C, F) VEGF-treated cells were evaluated. Representative phase-contrast images at 100× magnification from two to three independent experiments are shown. Mean ± SEM of three samples per treatment is shown for percent inhibition of branching. *P<0.05, **P<0.01.

In a separate experiment, we examined the effect of AA-PMe and AA treatment on preformed tubes, beginning drug treatment after HUVECs had been allowed to form a tubular network for 6 hours. Again, the addition of AA-PMe significantly disrupted tube formation, reducing tube length by 30%, 52%, and 68% at 5, 10, and 25 μM, respectively, while AA reduced length by 17%, 32%, and 49% at the same doses ().

Finally, we tested whether AA-PMe and AA had similar effects on capillary development when HUVECs were treated with VEGF to stimulate growth. Although AA-PMe still decreased tube length and branch formation at 10 and 25 μM doses AA only had an effect at 25 μM ().

AA-PMe inhibits angiogenesis in transgenic zebrafish models

To extend these findings to the zebrafish model of whole organism angiogenesis Tg(fli:EGFP) embryos were treated with varying concentrations of AA and AA-PMe from 6 to 72 hpf, at which point all ISV had stretched to form dorsal longitudinal anastomotic vessels. In keeping with our in vitro findings, in vivo fluorescence imaging of EGFP expressing endothelial cells demonstrated potent inhibition of ISV, dorsal aorta (DA), and posterior cardinal vein (PCV) development.

At first, as shown in , the ISVs were the most easily observed angiogenic vessels in the embryos at 72 hpf. AA-PMe showed potent inhibition on ISV formation at 10 μM, but AA showed no obvious effect at 10 μM (, left panel and central panel). We also found that the mean ISV length was 34.63±0.84 μm after treatment with 10 μM AA-PMe. The mean ISV length of the AA group was 44.96±0.45 μm at 10 μM, and it is obvious that treatment with AA-PMe resulted in shorter ISV than AA ().

Figure 6 AA-PMe and AA treatment results in gross defects in zebrafish vascular development. (A) Live fluorescence microscopy images depicting gross morphology of 72 hpf Tg(fli:EGFP) zebrafish embryos treated with DMSO alone or the indicated concentrations of AA-PMe and AA. ISV and SIV are clearly defined by fluorescence, the latter appearing as a smooth basket-like structure with five to six arcades. Red and yellow bars indicate the lumina of the DA and PCV, respectively. Scale bar, 50 μm. (B) Length of the ISV; (C) the number of SIV branch points; (D) SIV length in 72 hpf embryos treated with DMSO or the indicated concentrations of AA-PMe and AA were determined as described. (E) DA lumina diameters. (F) PCV lumen diameter. Data are expressed as mean ± SEM from three independent experiments. *P<0.05, **P<0.01.

Abbreviations: AA, asiatic acid; AA-PMe, N-(2α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester; HUVEC, human umbilical vein endothelial cell; DMSO, dimethyl sulfoxide; ISV, intersegmental blood vessel; SIV, subintestinal vessel; DA, dorsal aorta; PCV, posterior cardinal vein.
Figure 6 AA-PMe and AA treatment results in gross defects in zebrafish vascular development. (A) Live fluorescence microscopy images depicting gross morphology of 72 hpf Tg(fli:EGFP) zebrafish embryos treated with DMSO alone or the indicated concentrations of AA-PMe and AA. ISV and SIV are clearly defined by fluorescence, the latter appearing as a smooth basket-like structure with five to six arcades. Red and yellow bars indicate the lumina of the DA and PCV, respectively. Scale bar, 50 μm. (B) Length of the ISV; (C) the number of SIV branch points; (D) SIV length in 72 hpf embryos treated with DMSO or the indicated concentrations of AA-PMe and AA were determined as described. (E) DA lumina diameters. (F) PCV lumen diameter. Data are expressed as mean ± SEM from three independent experiments. *P<0.05, **P<0.01.

While the SIVs develop first by 72 hpf, the smooth, basket-like structures of the SIVs become apparent and the DA and PCV diameters are measurable. In vehicle-treated embryos, 5–6 SIV arcades were visible, but with 5 μM AA-PMe only partial development occurred and at 10 μM AA-PMe formation failed. AA treatment was slightly less inhibitory, with severe disruption at 10 μM and nearly complete blockage of formation at 25 μM AA (, far right panel). SIV branch points and length were also affected by 25 μM AA-PMe and AA treatment: branch points were reduced by 85.7% and 51.4% () and SIV length decreased to 98.31±2.67 and 135.69±2.78 μm in response to 25 μM AA-PMe and AA, respectively ().

AA-PMe and AA treatment also decreased DA and PCV diameters (, central panel): DA diameters narrowed to 9.81±1.07 and 21.71±0.72 μM, and PCV diameters shrank to 16.9±0.39 and 21.39±0.39 μm in embryos treated with 10 μM AA-PMe and AA, respectively ().

In keeping with these morphometric findings, the quantitative EAP assay revealed a nearly 50% inhibition of blood vessel formation at 10 μM AA-PMe and a 20% inhibition at 10 μM AA (). Overall, while both compounds inhibit angiogenesis in zebrafish embryos, we found that AA-PMe has a dramatically more powerful effect than AA.

Figure 7 AA-PMe inhibits zebrafish embryo angiogenesis. Quantitative EAP assay was used to confirm our morphological observations of zebrafish embryos treated with the indicated concentrations of AA-PMe and AA. Data are presented as mean ± SEM, n=10. *P<0.05, **P<0.01.

Abbreviations: AA, asiatic acid; AA-PMe, N-(2α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester; EAP, endogenous alkaline phosphatase.
Figure 7 AA-PMe inhibits zebrafish embryo angiogenesis. Quantitative EAP assay was used to confirm our morphological observations of zebrafish embryos treated with the indicated concentrations of AA-PMe and AA. Data are presented as mean ± SEM, n=10. *P<0.05, **P<0.01.

AA-PMe changes VEGF-induced VEGFR2 mRNA expression in HUVECs

VEGF is a fundamental mediator of physiological and pathological angiogenesis and acts through two tyrosine kinase receptors, VEGFR1 and VEGFR2.Citation48 However, VEGFR2 has a higher affinity for VEGF and is the major transducer of VEGF signaling in endothelial cells during blood vessel formation, while VEGFR1 modulates VEGFR2 signaling.Citation49 In our study, 10 and 25 μM AA-PMe significantly decreased VEGFR2 mRNA expression, which might reduce the stimulatory effect of VEGF (). However, AA treatment had no significant effect at any concentration.

Figure 8 AA-PMe inhibits VEGF-induced upregulation of VEGFR2 mRNA in HUVECs. VEGFR2 mRNA in HUVECs treated with or without VEGF (10 ng/mL) and DMSO vehicle or the indicated concentrations of AA-PMe or AA for 24 hours. Expression values were normalized to GAPDH. Data are presented as the mean ± SEM from three independent experiments. *P<0.05, **P<0.01.

Abbreviations: AA, asiatic acid; AA-PMe, N-(2α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester; HUVEC, human umbilical vein endothelial cell; VEGF, vascular endothelial growth factor; VEGFR2, vascular endothelial growth factor receptor 2; DMSO, dimethyl sulfoxide.
Figure 8 AA-PMe inhibits VEGF-induced upregulation of VEGFR2 mRNA in HUVECs. VEGFR2 mRNA in HUVECs treated with or without VEGF (10 ng/mL) and DMSO vehicle or the indicated concentrations of AA-PMe or AA for 24 hours. Expression values were normalized to GAPDH. Data are presented as the mean ± SEM from three independent experiments. *P<0.05, **P<0.01.

AA-PMe inhibits both VEGFR2 phosphorylation and activation of VEGFR2-mediated signaling in HUVECs and zebrafish embryos

Three major tyrosine phosphorylation sites, Tyr951, Tyr1175, and Tyr1214, have been identified on VEGFR2. In developing vessels, pTyr1175 and pTyr1214 have been detected on all VEGFR2 expressing endothelial cells, and autophosphorylation of Tyr1175 on VEGFR2 is crucial for endothelial cell proliferation, making these sites promising targets for antiangiogenic therapeutics.Citation50,Citation51 To dissect the molecular basis underlying the antiangiogenic effects we noted in HUVECs and zebrafish embryos, we followed VEGFR2 protein expression and phosphorylation by Western blot in HUVECs treated with varying concentrations of AA-PMe and AA. While even 10 μM AA-PMe dramatically inhibited phosphorylation of VEGFR2 Tyr1175, AA had little effect on the total VEGFR2 protein expression () at any dose, but the same inhibitory effect on phosphorylation of VEGFR2 Tyr1175 at 25 μM as AA-PMe at 10 μM. Once again, our results demonstrate the far greater potency of AA-PMe compared to AA.

Figure 9 AA-PMe inhibits VEGF-induced phosphorylation of VEGFR2 and downstream kinases in HUVECs. Expression of (A) total and phospho-VEGFR2 and (B) total and phospho-ERK and AKT in response to the indicated doses of AA-PMe and AA.

Abbreviations: AA, asiatic acid; AA-PMe, N-(2α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester; HUVEC, human umbilical vein endothelial cell; VEGF, vascular endothelial growth factor; VEGFR2, vascular endothelial growth factor receptor 2; ERK, extracellular signal-related kinase.
Figure 9 AA-PMe inhibits VEGF-induced phosphorylation of VEGFR2 and downstream kinases in HUVECs. Expression of (A) total and phospho-VEGFR2 and (B) total and phospho-ERK and AKT in response to the indicated doses of AA-PMe and AA.

Multiple signaling pathways responsible for endothelial cell migration, proliferation, and survival are activated downstream of VEGF–VEGFR2 interaction;Citation52,Citation53 so, we examined phosphorylation and activation of several of these key kinases. We found that in addition to inhibiting VEGFR2 phosphorylation, AA-PMe significantly suppressed phos-phorylation and activation of ERK1/2 and AKT in HUVECs ().

To confirm these VEGFR2 findings in zebrafish embryos, we analyzed total and phospho-VEGFR2 protein and VEGFR2 mRNA expression following AA-PMe and AA treatment. Similar to our HUVEC findings, VEGFR2 phosphorylation was completely inhibited by 25 μM AA-PMe, with lower doses also decreasing p-VEGFR2. There was no effect on total VEGFR2 expression at any dose, resulting in a decreased ratio of phospho to total protein (). We noted that 25 μM AA had an effect on VEGFR2 phosphorylation similar to the lower AA-PMe doses. VEGFR2 mRNA expression followed a similar pattern, with all doses of AA-PMe significantly reducing and 25 μM AA partially reducing its expression ().

Figure 10 AA-PMe inhibits VEGFR2 activation in zebrafish embryos. (A) Total and phospho-VEGFR2 expression in 72 hpf whole embryo lysates following treatment with the indicated doses of AA-PMe or AA. A representative blot is shown. (B) Ratio of phospho to total VEGFR2 expression as determined by the densitometric analysis. Bars represent the mean ± SEM from three separate experiments. *P<0.05, **P<0.01.

Abbreviations: AA, asiatic acid; AA-PMe, N-(2α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester; HUVEC, human umbilical vein endothelial cell; VEGFR2, vascular endothelial growth factor receptor 2.
Figure 10 AA-PMe inhibits VEGFR2 activation in zebrafish embryos. (A) Total and phospho-VEGFR2 expression in 72 hpf whole embryo lysates following treatment with the indicated doses of AA-PMe or AA. A representative blot is shown. (B) Ratio of phospho to total VEGFR2 expression as determined by the densitometric analysis. Bars represent the mean ± SEM from three separate experiments. *P<0.05, **P<0.01.

Figure 11 AA-PMe inhibits VEGFR2 in zebrafish embryos. qRT-PCR analysis of VEGFR2 mRNA in 72 hpf whole embryos treated with DMSO control or the indicated concentrations of AA-PMe or AA. Data are presented as mean ± SEM from three independent experiments. *P<0.05, **P<0.01.

Abbreviations: AA, asiatic acid; AA-PMe, N-(2α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester; VEGFR2, vascular endothelial growth factor receptor 2; qRT-PCR, quantitative real-time polymerase chain reaction; DMSO, dimethyl sulfoxide.
Figure 11 AA-PMe inhibits VEGFR2 in zebrafish embryos. qRT-PCR analysis of VEGFR2 mRNA in 72 hpf whole embryos treated with DMSO control or the indicated concentrations of AA-PMe or AA. Data are presented as mean ± SEM from three independent experiments. *P<0.05, **P<0.01.

As a whole, our results suggest that AA-PMe and AA exert their antiangiogenic activity through inhibition of the VEGFR2-mediated signaling cascade in both endothelial cells and zebrafish embryos, with AA-PMe clearly surpassing AA in its activity.

Discussion and conclusion

Angiogenesis is considered to be a key process in many physiological and pathological states, including cancer growth and progression.Citation45,Citation54,Citation55 Complex cellular and extracellular components surround tumor cells at every stage of development to form the tumor microenvironment and this requires a steady blood supply.Citation55 Recent advances suggest that antiangiogenic strategies targeting endothelial cells in the tumor microenvironment could prove particularly valuable because these cells are generally non-transformed and are considered less prone to acquiring drug resistance.Citation45,Citation56 Nontoxic agents that effectively target endothelial cells and reduce pathological angiogenesis in the tumor microenvironment could therefore be important tools in cancer prevention and treatment. Consequently, there is growing interest in the careful analysis of natural compounds derived from traditional medicines like AA, which has been found to influence steps in cancer angiogenesis with very few side effects.Citation34 We have previously reported that AA-PMe, a modified form of AA synthesized in our laboratory, exerts antitumor activity by blocking cell cycling and cell mobility and inducing apoptosis.Citation57 We further demonstrated antiangiogenic effects of AA-PMe in zebrafish.Citation20 In the present study, we extend these findings to demonstrate powerful inhibition of endothelial cell growth, tube formation, and motility, all of which are crucial for blood vessel formation. This work highlighted the importance of AA-PMe as a novel antiangiogenic agent but the mechanism remained unclear.

Although angiogenesis is regulated by a large number of pro- and antiangiogenic cellular factors, the available evidence points to VEGF as a major paracrine mediator that directly contributes to angiogenesis.Citation58 Following VEGF–VEGFR2 binding, signaling pathways controlling endothelial cell survival, proliferation, migration, and tube formation are activated.Citation59,Citation60 The downregulation of VEGFR2 mRNA expression by AA-PMe may contribute to its effect as an angiogenesis inhibitor by decreasing VEGFR2 signaling over time. However, AA-PMe also appears to act immediately by inhibiting phosphorylation of VEGFR2 to block its activation.

In patients, successful antiangiogenic therapy may require the simultaneous blockade multiple pro-angiogenic factor receptors at downstream signaling pathway.Citation61 VEGF–VEGFR2 interaction alone activates numerous signaling pathways, inducing phosphorylation of ERK, c-Jun amino-terminal kinase, phosphatidylinositide 3-kinase, PKB (AKT), and p38 mitogen-activated protein kinase.Citation14,Citation62 Interestingly, AKT signaling has been found to regulate the expression of pro-angiogenic factors at the translational level, which makes this pathway a potential target for antiangiogenic therapy.Citation13 We discovered that AA-PMe blocked two signaling components downstream of VEGFR2, reducing AKT and ERK phosphorylation in endothelial cells. In zebrafish embryos, AA-PMe also decreased VEGFR2. Thus, we suggest that AA-PMe exerts its antiangiogenic activity at least in part by regulating VEGFR2-mediated signaling in HUVECs and zebrafish ().

Figure 12 Proposed antiangiogenic mechanism of AA-PMe by regulation of VEGF–VEGFR2 signaling.

Abbreviations: AA-PMe, N-(2α,3β,23-acetoxyurs-12-en-28-oyl)-L-proline methyl ester; VEGF, vascular endothelial growth factor; VEGFR2, vascular endothelial growth factor receptor 2; ERK, extracellular signal-related kinase.
Figure 12 Proposed antiangiogenic mechanism of AA-PMe by regulation of VEGF–VEGFR2 signaling.

Our findings begin to elucidate the mechanism underlying the antiangiogenic activity of AA-PMe. We demonstrate here that AA-PMe inhibits phosphorylation of VEGFR2, suppressing the VEGFR2-mediated signaling pathway that plays crucial roles in regulating neovascularization and inducing apoptosis. These data support further development of AA-PMe as a novel VEGFR2 kinase inhibitor for the treatment of angiogenesis-related diseases.

Author contributions

All authors made substantial contributions to conception and design, acquisition of data, or analysis and interpretation of data; took part in drafting the article or revising it critically for important intellectual content; gave final approval of the version to be published; and agree to be accountable for all aspects of the work.

Acknowledgments

This work was supported by grants from the Natural Science Foundation of China (grant no 81573833).

Disclosure

The authors report no conflicts of interest in this work.

References

  • QuesadaARMunoz-ChapuliRMedinaMAAnti-angiogenic drugs: from bench to clinical trialsMed Res Rev20062648353016652370
  • CarmelietPAngiogenesis in life, disease and medicineNature200543893293616355210
  • FerraraNKerbelRSAngiogenesis as a therapeutic targetNature200543896797416355214
  • ZhongHBowenJPMolecular design and clinical development of VEGFR kinase inhibitorsCurr Top Med Chem200771379139317692027
  • FerraraNVascular endothelial growth factor: basic science and clinical progressEndocr Rev20042558161115294883
  • DongDFLiEXWangJBAnti-angiogenesis and anti-tumor effects of AdNT4-anginexCancer Lett200928521822419540664
  • SimoRCarrascoEGarcia-RamirezMHernandezCAngiogenic and antiangiogenic factors in proliferative diabetic retinopathyCurr Diabetes Rev20062719818220619
  • PratheeshkumarPBudhrajaASonYOQuercetin inhibits angiogenesis mediated human prostate tumor growth by targeting VEGFR-2 regulated AKT/mTOR/P70S6K signaling pathwaysPLoS One20127e4751623094058
  • PotenteMGerhardtHCarmelietPBasic and therapeutic aspects of angiogenesisCell201114687388721925313
  • LiaoDJohnsonRSHypoxia: a key regulator of angiogenesis in cancerCancer Metastasis Rev20072628129017603752
  • FerraraNVEGF and the quest for tumour angiogenesis factorsNat Rev Cancer2002279580312360282
  • LinCWuMHDongJYQuercetin-42-O-beta-D-glucopyranoside (QODG) inhibits angiogenesis by suppressing VEGFR2-mediated signaling in zebrafish and endothelial cellsPLoS One20127e3170822348123
  • LiuBFaiaLHuMNussenblattRBPro-angiogenic effect of IFNgamma is dependent on the PI3K/mTOR/translational pathway in human retinal pigmented epithelial cellsMol Vis20101618419320157617
  • FerraraNGerberHPLeCouterJThe biology of VEGF and its receptorsNat Med2003966967612778165
  • FerraraNHillanKJGerberHPNovotnyWDiscovery and development of bevacizumab, an anti-VEGF antibody for treating cancerNat Rev Drug Discov2004339140015136787
  • Randrup HansenCGrimmDBauerJWehlandMMagnussonNEEffects and side effects of using sorafenib and sunitinib in the treatment of metastatic renal cell carcinomaInt J Mol Sci201718E46128230776
  • ShantakumarSNordstromBLDjousseLOccurrence of hepa-totoxicity with pazopanib and other anti-VEGF treatments for renal cell carcinoma: an observational study utilizing a distributed database networkCancer Chemother Pharmacol20167855956627438066
  • NobleMEEndicottJAJohnsonLNProtein kinase inhibitors: insights into drug design from structureScience20043031800180515031492
  • DongDKoBBaumeisterPVascular targeting and antiangio-genesis agents induce drug resistance effector GRP78 within the tumor microenvironmentCancer Res2005655785579115994954
  • JingYWangGGeYXuMGongZSynthesis, anti-tumor and anti-angiogenic activity evaluations of asiatic acid amino acid derivativesMolecules2015207309732425905607
  • SomboonwongJKankaisreMTantisiraBTantisiraMHWound healing activities of different extracts of Centella asiatica in incision and burn wound models: an experimental animal studyBMC Complement Altern Med20121210322817824
  • ShuklaARasikAMJainGKShankarRKulshresthaDKDhawanBNIn vitro and in vivo wound healing activity of asiaticoside isolated from Centella asiaticaJ Ethnopharmacol19996511110350364
  • Soo LeeYJinD-QBeakS-MLeeE-SKimJ-AInhibition of ultraviolet-A-modulated signaling pathways by asiatic acid and ursolic acid in HaCaT human keratinocytesEur J Pharmacol200347617317812969763
  • Mook-JungIShinJEYunSHProtective effects of asiaticoside derivatives against beta-amyloid neurotoxicityJ Neurosci Res19995841742510518115
  • JewSSYooCHLimDYStructure–activity relationship study of asiatic acid derivatives against beta amyloid (Aβ)-induced neurotoxicityBioorg Med Chem Lett20001011912110673093
  • LeeMKKimSRSungSHAsiatic acid derivatives protect cultured cortical neurons from glutamate-induced excitotoxicityRes Commun Mol Pathol Pharmacol2000108758611758977
  • KrishnamurthyRGSenutMCZemkeDAsiatic acid, a pentacyclic triterpene from Centella asiatica, is neuroprotective in a mouse model of focal cerebral ischemiaJ Neurosci Res2009872541255019382233
  • TangLXHeRHYangGAsiatic acid inhibits liver fibrosis by blocking TGF-beta/Smad signaling in vivo and in vitroPLoS One20127e3135022363627
  • HsuYLKuoPLLinLTLinCCAsiatic acid, a triterpene, induces apoptosis and cell cycle arrest through activation of extracellular signal-regulated kinase and p38 mitogen-activated protein kinase pathways in human breast cancer cellsJ Pharmacol Exp Ther200531333334415626723
  • ParkBCBosireKOLeeESLeeYSKimJAAsiatic acid induces apoptosis in SK-MEL-2 human melanoma cellsCancer Lett2005218819015639343
  • TangX-LYangX-YJungH-JAsiatic acid induces colon cancer cell growth inhibition and apoptosis through mitochondrial death cascadeBiol Pharm Bull2009321399140519652380
  • ChoCWChoiDSCardoneMHKimCWSinskeyAJRhaCGlio-blastoma cell death induced by asiatic acidCell Biol Toxicol20062239340816897440
  • LeeYSJinD-QKwonEJAsiatic acid, a triterpene, induces apoptosis through intracellular Ca2+ release and enhanced expression of p53 in HepG2 human hepatoma cellsCancer Lett2002186839112183079
  • KavithaCVAgarwalCAgarwalRDeepGAsiatic acid inhibits pro-angiogenic effects of VEGF and human gliomas in endothelial cell culture modelsPLoS One20116e2274521826202
  • ZhaoDXQinCJFanXHLiYCGuBHInhibitory effects of quercetin on angiogenesis in larval zebrafish and human umbilical vein endothelial cellsEur J Pharmacol201472336036724239714
  • XuZLiYXiangQDesign and synthesis of novel xyloketal derivatives and their vasorelaxing activities in rat thoracic aorta and angiogenic activities in zebrafish angiogenesis screenJ Med Chem2010534642465320481602
  • LawsonNDWeinsteinBMIn vivo imaging of embryonic vascular development using transgenic zebrafishDev Biol200224830731812167406
  • YuXBTongYHanXQAnti-angiogenic activity of Herba Epimedii on zebrafish embryos in vivo and HUVECs in vitroPhytother Res2013271368137523147754
  • TruongLHarperSLTanguayRLEvaluation of embryotoxicity using the zebrafish modelMethods Mol Biol201169127127920972759
  • MuXYPangSSunXZEvaluation of acute and developmental effects of difenoconazole via multiple stage zebrafish assaysEnviron Pollut201317514715723391686
  • HeMFHuangYHWuLWGeWShawPCButPPTriptolide functions as a potent angiogenesis inhibitorInt J Cancer201012626627819569053
  • LamHWLinHCLaoSCThe angiogenic effects of Angelica sinensis extract on HUVEC in vitro and zebrafish in vivoJ Cell Biochem200810319521117497682
  • LuXLXuZLYaoXLMarine cyclotripeptide X-13 promotes angiogenesis in zebrafish and human endothelial cells via PI3K/Akt/eNOS signaling pathwaysMar Drugs2012101307132022822374
  • LuXLLuoDYaoXLdl-3n-Butylphthalide promotes angio-genesis via the extracellular signal-regulated kinase 1/2 and phosphati-dylinositol 3-kinase/Akt-endothelial nitric oxide synthase signaling pathwaysJ Cardiovasc Pharmacol20125935236222286127
  • SinghRPDhanalakshmiSAgarwalCAgarwalRSilibinin strongly inhibits growth and survival of human endothelial cells via cell cycle arrest and downregulation of survivin, Akt and NF-kappaB: implications for angioprevention and antiangiogenic therapyOncogene2005241188120215558015
  • ParngCSengWLSeminoCMcGrathPZebrafish: a preclinical model for drug screeningAssay Drug Dev Technol20021414815090155
  • van MoorselaarRJVoestEEAngiogenesis in prostate cancer: its role in disease progression and possible therapeutic approachesMol Cell Endocrinol200219723925012431818
  • ZacharyISignaling mechanisms mediating vascular protective actions of vascular endothelial growth factorAm J Physiol Cell Physiol2001280C1375C138611350732
  • RobertsDMKearneyJBJohnsonJHRosenbergMPKumarRBautchVLThe vascular endothelial growth factor (VEGF) receptor Flt-1 (VEGFR-1) modulates Flk-1 (VEGFR-2) signaling during blood vessel formationAm J Pathol20041641531153515111299
  • MatsumotoTBohmanSDixeliusJVEGF receptor-2 Y951 signaling and a role for the adapter molecule TSAd in tumor angiogenesisEMBO J2005242342235315962004
  • TakahashiTYamaguchiSChidaKShibuyaMA single autophos-phorylation site on KDR/Flk-1 is essential for VEGF-A-dependent activation of PLC-gamma and DNA synthesis in vascular endothelial cellsEMBO J2001202768277811387210
  • PangXYiZZhangJCelastrol suppresses angiogenesis-mediated tumor growth through inhibition of AKT/mammalian target of rapamycin pathwayCancer Res2010701951195920160026
  • EdirisingheIYangSRYaoHVEGFR-2 inhibition augments cigarette smoke-induced oxidative stress and inflammatory responses leading to endothelial dysfunctionFASEB J2008222297231018263699
  • CrawfordTNAlfaroDV3rdKerrisonJBJablonEPDiabetic retinopathy and angiogenesisCurr Diabetes Rev2009581319199892
  • AlbiniASpornMBThe tumour microenvironment as a target for chemopreventionNat Rev Cancer2007713914717218951
  • CarmelietPJainRKAngiogenesis in cancer and other diseasesNature200040724925711001068
  • JingYWangGGeYXuMTangSGongZAA-PMe, a novel asiatic acid derivative, induces apoptosis and suppresses proliferation, migration, and invasion of gastric cancer cellsOnco Targets Ther201691605162127073325
  • JainRKdi TomasoEDudaDGLoefflerJSSorensenAGBatchelorTTAngiogenesis in brain tumoursNat Rev Neurosci2007861062217643088
  • GuQWangDWangXBasic fibroblast growth factor inhibits radiation-induced apoptosis of HUVECs. I. The PI3K/AKT pathway and induction of phosphorylation of BADRadiat Res200416169270215161350
  • Gingis-VelitskiSZetserAFlugelmanMYVlodavskyIIlanNHeparanase induces endothelial cell migration via protein kinase B/Akt activationJ Biol Chem2004279235362354115044433
  • KandaSMiyataYKanetakeHSmithgallTENon-receptor proteintyrosine kinases as molecular targets for antiangiogenic therapy (review)Int J Mol Med20072011312117549397
  • AmorosoFCapeceMRotondoAThe P2X7 receptor is a key modulator of the PI3K/GSK3beta/VEGF signaling network: evidence in experimental neuroblastomaOncogene2015345240525125619831