Fasudil inhibits prostate cancer-induced angiogenesis in vitro

  • Authors:
    • Weihua Chen
    • Kaili Mao
    • Thong Hua-Huy
    • Yihua Bei
    • Zhongmin Liu
    • Anh-Tuan Dinh-Xuan
  • View Affiliations

  • Published online on: September 17, 2014     https://doi.org/10.3892/or.2014.3491
  • Pages: 2795-2802
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Inhibition of angiogenesis is an important therapeutic strategy for advanced stage prostate cancer (PCa). RhoA/Rho-associated protein kinases (ROCK) are key regulators of the cytoskeleton and have been implicated in PCa angiogenesis. We investigated the anti-angiogenic effects of fasudil, a ROCK inhibitor, on PCa-induced angiogenesis in vitro. Proliferation of PCa-conditioned human umbilical vein endothelial cells (HUVECs) was assessed using a bromodeoxyuridine (BrdU) assay, and migration was assessed with a wound healing assay. In vitro angiogenesis of PCa-conditioned HUVECs was evaluated by tube formation and a spheroid sprouting assay. Fasudil inhibited PCa-induced endothelial cell proliferation at a concentration of 100 µM, and also decreased PCa-induced endothelial cell migration at a concentration of 30 µM. In the in vitro angiogenesis assay, fasudil exerted a more significant effect. Tube formation was significantly inhibited at fasudil concentrations exceeding 3 µM, and spheroid sprouts were significantly thinner and shorter (at fasudil concentrations of 10 and 30 µM, respectively). Western blotting results showed that expression of phosphorylated myosin phosphatase target subunit 1 (MYPT-1) was significantly lower after fasudil treatment, confirming that fasudil inhibited ROCK activity in these model systems. These data suggest that fasudil may be a useful anti-angiogenic agent for PCa.

Introduction

Prostate cancer (PCa) is the most common malignancy and the second leading cause of cancer-related death in men in Western countries. Advanced and metastatic stages of the disease are found in 35% of patients with PCa diagnosed at autopsy (1). Among patients with localized cancer who are eligible for radical prostatectomy, ~35% will develop recurrence (metastatic disease) within 10 years of surgery (2,3).

Androgen deprivation therapy (ADT) can be effective in patients who present with or progress to advanced or meta-static disease. Unfortunately, the median duration of response to ADT is limited to between 8 months and 3 years (4), and these patients will eventually become castration resistant. Chemotherapy is an effective treatment for castration-resistant PCa, but the median duration of response is only 10.3 months (5). There is clearly an urgent need to develop additional systemic interventions for patients with progressive PCa. Angiogenesis plays a crucial role in PCa progression and metastasis. Microvessel density (MVD) has been found to be more prominent in PCa than in benign prostatic hyperplasia (BPH) and normal tissue (6,7). It has been reported that MVD increases with increased Gleason’s score, particularly in poorly differentiated PCa (8). MVD was also significantly correlated with cancer-specific survival in 221 patients with PCa followed up for a median of 15 years (9).

Vascular endothelial growth factor (VEGF) is the most prominent regulator of physiological angiogenesis and has been correlated with increased levels of angiogenesis in clinical studies comparing PCa with BPH (7). Higher VEGF expression and serum levels have also been found in patients with metastasis or poorly differentiated tumors, as well as in those with a poor prognosis (1013). However, it has become increasingly apparent that current anti-angiogenic therapy targeting VEGF has only a modest effect in the clinical setting.

RhoA and its downstream effector, Rho-associated protein kinase (ROCK), serve as key regulators of extracellular stimulus-mediated signaling networks that are involved in various cellular processes, including motility, mitosis, proliferation and apoptosis (14). Suppression of the RhoA/ROCK signaling pathway with the ROCK inhibitor, Y-27632, was found to inhibit VEGF-induced angiogenesis in vitro (15). Another ROCK inhibitor, fasudil, has been shown to inhibit VEGF-induced angiogenesis in vitro and in vivo (16). A study carried out on endothelial cells from transgenic adenocarcinoma of the mouse prostate (TRAMP) mice revealed that their behavior correlated with a constitutively high level of baseline activity of Rho GTPase and ROCK (17). This suggests that the RhoA/ROCK pathway has an important role in PCa angiogenesis. However, the anti-angiogenic effects of ROCK inhibitors in PCa are unclear. We investigated the role of fadusil, a ROCK inhibitor, that has been approved for clinical use for pulmonary arterial hypertension, on PCa-induced angiogenesis in vitro.

Materials and methods

Cell culture

Human umbilical vein endothelial cells (HUVECs) were purchased from PromoCell (C-12200; Heidelberg, Germany) and cultured in endothelial cell growth medium (C-22010; PromoCell). Cultures were maintained at 37°C in a humidified atmosphere containing 5% CO2. Subcultures were obtained by trypsinization and were used for experiments at passages 3 to 9. Before performing the experiments, the cells were made quiescent by incubating overnight in endothelial cell basal medium (C-22210) containing 0.5% (w/v) fetal bovine serum (FBS). The PCa cell line, PC-3, was purchased from The European Collection of Cell Cultures and grown in F-12K (Gibco-Invitrogen, Carlsbad, CA, USA) containing 10% (w/v) FBS. PC-3 cells were seeded at a concentration of 6×106 cells/T75 flask. On the following day, the medium was replaced with basal medium without FBS, and the supernatants were harvested after a 24-h incubation to serve as conditioned medium (PC3CM). Recombinant human VEGF 165 was purchased from R&D Systems (293-VE; Minneapolis, MN, USA). HUVECs were cultured in endothelial cell basal medium plus 2% (w/v) FBS (control group), in PC3CM plus 2% (w/v) FBS (PC3CM group), or in basal medium plus 2% (w/v) FBS and 30 ng/ml VEGF (VEGF group).

Cell proliferation assay

HUVEC proliferation was evaluated using a BrdU incorporation assay kit (Amersham; Cell Proliferation Biotrak ELISA System; RPN250; GE Healthcare, Little Chalfont, UK), according to the manufacturers’ instructions. In brief, HUVECs were plated in 96-well microculture plates (3×103 cells/well). After a 48-h incubation at 37°C in a 5% CO2 atmosphere, with or without fasudil (1–100 μM), 10 μl BrdU labeling reagent was added, and the cells were cultured for a further 2 h. Cells were washed twice with Dulbecco’s PBS (D8537; Sigma-Aldrich, St. Louis, MO, USA), fixed with fixative solution and then blocked with blocking buffer. BrdU incorporation was revealed by incubation with 100 μl/well horseradish peroxidase (HRP)-labeled anti-BrdU working solution for ~ 90 min. Tetramethylbenzidine (TMB) substrate at room temperature was added at 100 μl/well for 20 min. Absorbance was measured at 450 nm using a microplate reader. All determinations were performed in octuplicate, and each experiment was repeated three times.

Cell migration assay

Cell motility was assessed using a wound-healing migration assay. HUVECs were seeded to full confluency in 6-well plates. The following day, a uniform scratch was made down the centre of the well using a 100-μl micropipette tip, and the cells were washed twice with PBS. After incubation for 24 h with or without 30 μM fasudil in the control, PC3CM and VEGF groups, the cells were fixed and photographed. Photographic imaging was performed using a Leica inverted microscope. Cell migration was quantified by measuring the ratio of the migration area to the total area of the wound gap. Each experiment was repeated three times.

Tube formation assay

Ninety-six-well plates were chilled to 4°C and coated with 50 μl of Matrigel (354234; BD Biosciences, Oxford, UK) per well. Freshly passaged HUVECs were seeded onto the gel. Endothelial tube morphogenesis was carried out in the presence or absence of fasudil (3–30 μM). Endothelial tube formation was observed after 16 h and photographed under phase contrast microscopy using a Leica inverted microscope. Quantification of the digital images was performed by counting the total number of tubes in five 40× fields, and total tube length was quantified using ImageJ™ software (NIH, Bethesda, MD, USA). Tube formation was expressed as fold change or percentage, compared to the controls. All determinations were performed three times, and each experiment was repeated three times.

Spheroid sprouting assay

HUVECs were suspended in culture medium containing 0.2% (w/v) methylcellulose (Sigma-Aldrich) and seeded in non-adherent round-bottom 96-well plates (Greiner, Frickenhausen, Germany). All suspended cells formed a single spheroid in each well of defined size and cell number (~400 cells/spheroid). Spheroids were left to form for 24 h and then embedded in 1.5 mg/ml collagen gel. The spheroid-containing gel was rapidly transferred to pre-warmed 24-well plates and allowed to polymerize for 30 min. Endothelial basal medium or PC3CM with or without fasudil (1–100 μM) was then added to the surface of the gel (500 μl/well). After 16 h, images were captured using a Leica inverted microscope. Sprouting was quantified using NIH ImageJ software by measuring the cumulative sprout length, which consisted of every sprout from 10 spheroids in each group.

Western blot assay

Protein was extracted on ice from the cultured HUVECs with cold RIPA lysis buffer (9806; Cell Signaling Technology, Boston, MA, USA) containing Pierce™ Protease and Phosphatase Inhibitor (88669; Thermo Scientific, Rockford, IL, USA). Lysates were centrifuged at 12,000 × g for 20 min at 4°C, and the supernatant was collected. Total protein concentrations were determined using a bicinchoninic acid assay (BCA) protein assay kit (23250; Thermo Scientific). Equal amounts of protein were subjected to sodium dodecyl sulfate polyacrylamide gel electrophoresis and then electrically transferred onto nitrocellulose membranes. The membranes were blocked for 1 h with 5% (w/v) non-fat milk in PBS-0.1% (v/v) Tween-20 (PBST) and incubated with primary antibodies against MYPT-1 (1:1,000; sc-25618; Santa Cruz Biotechnology, Dallas, TX, USA), phospho-MYPT-1 (1:500; ABS45; Millipore, Billerica, MA, USA), anti-ROCK1 (1:500; sc-6055), anti-ROCK2 (1:1,000; sc-1851; both from Santa Cruz Biotechnology) and β-actin (1:500; ab8229; Abcam, Cambridge, UK) overnight at 4°C. Finally, the membrane was incubated with HRP-conjugated secondary antibodies as follows: goat anti-mouse IgG-HRP (1:5,000; sc-2005; Santa Cruz Biotechnology), rabbit anti-goat IgG-HRP (1:10,000; sc-2768; Santa Cruz Biotechnology), goat anti-rabbit IgG-HRP (1:10,000; sc-2004; Santa Cruz Biotechnology) for 1 h at room temperature. After washing three times with PBST, proteins were visualized using an ECL Prime Western blotting detection kit (GE Healthcare). Photographs of the protein bands were captured using a digital imaging system (ImageQuant LAS; GE Healthcare), and densitometric measurements of band intensity in the western blotting were performed using NIH ImageJ software. The results shown are representative of three or more independent experiments.

Statistical analysis

Data are expressed as means ± standard deviation. Significance of differences was determined by the two-tailed Student’s t-test or the analysis of variance least significant difference (ANOVA LSD) test. A P-value <0.05 was considered to indicate a statistically significant difference.

Results

Fasudil inhibits PC3CM-induced HUVEC proliferation

Endothelial cell proliferation is crucial for angiogenesis. PC3CM-treated HUVECs were exposed to fasudil concentrations ranging from 1 to 100 μM, and HUVEC proliferation was examined using a BrdU assay. Fasudil concentrations of ≥ 30 μM had a significant inhibitory effect on PC3CM-induced cell proliferation, while proliferation in the control group was unchanged (Fig. 1).

Fasudil inhibits PC3CM-induced HUVEC migration

The inhibitory effects of fasudil on endothelial cell motility were assessed using a wound-healing migration assay. Fasudil (30 μM) significantly decreased the number of cells migrating into the scratched gap in the control, PC3CM and VEGF groups, indicating the potent inhibitory effect of fasudil on HUVEC movement and migration. VEGF increased HUVEC migration significantly more than PC3CM-induced HUVEC migration. After treatment with 30 μM fasudil, all migrations decreased to similar levels (Fig. 2).

Fasudil inhibits PC3CM-induced HUVEC tube formation

The effect of fasudil on capillary-like structure formation in vitro was examined using a 3-dimensional (3D) Matrigel assay. When seeded onto Matrigel, HUVECs form tube structures and connect with each other, mimicking the in vivo process of angiogenesis. Sixteen hours after seeding, untreated HUVECs exhibited a clear capillary-like network formation. However, fasudil treatment dramatically decreased the capillary-like network formation in a dose-dependent manner. As fasudil concentration increased, total tube length gradually decreased (Fig. 3).

Fasudil inhibits PC3CM-induced HUVEC spheroid sprouting

In the sprout formation assay, HUVECs seeded in non-adhesive conditions in round bottom 96-well plates contributed to the formation of a single spheroid with a quiescent, non-proliferating surface monolayer within 24 h. The spheroids were then embedded in a 3D collagen matrix. In the untreated control group, baseline sprouting was low (Fig. 4Aa). When cultured with PC3CM (Fig. 4Ab), baseline sprouting increased dramatically, although it was still less than that in the cells cultured with basal medium containing 30 ng/ml VEGF (Fig. 4Ac). Sprouting was almost completely inhibited by treatment with 100 μM fasudil (Fig. 4Ad–f). We then examined the dose-dependent response of fasudil on PCa-induced HUVEC sprouting. As shown in Fig. 4B, fasudil decreased HUVEC sprouting in a dose-dependent manner and 100 μM fasudil again inhibited sprouting almost completely.

Furthermore, when treated with increasing concentrations of fasudil, the sprouts became thinner and the HUVEC nucleus seldom emerged from the spheroids. These sprouts resembled cell protrusions, were markedly thinner compared with the untreated HUVEC sprouts, and were more abundant compared with the ordered architecture of the single HUVEC spheroid sprouts (Fig. 4B).

Fasudil inhibits PC3CM-induced HUVEC ROCK activation

MYPT-1 is one of the most crucial downstream effectors of ROCK. As fasudil is a ROCK inhibitor, we examined the inhibitory effects of fasudil on ROCK by measuring phospho-MYPT-1 (pMYPT-1), the active form of MYPT-1.

As shown in Fig. 5, when cultured with PC3CM or basal medium containing VEGF, expression of both MYPT-1 and pMYPT-1 was increased in the HUVECs, resulting in a moderate increase in the pMYPT-1/MYPT-1 ratio, indicating ROCK activation. Fasudil treatment lead to a significant decrease in pMYPT-1 and a slight decrease in MYPT-1, resulting in a significant decrease in the pMYPT-1/MYPT-1 ratio (Fig. 5A–D). A moderate increase in ROCK1 and ROCK2 expression was also detected, but ROCK expression was not altered significantly by fasudil treatment (Fig. 5E and F).

Discussion

To our knowledge, there have been no previous reports on the effects of fasudil on PCa-induced angiogenesis. In this study, HUVECs were cultured with the PCa cell line PC3CM to mimic endothelial cells in PCa tissue. Fasudil was then added to examine its effects on PC3CM-induced HUVECs using in vitro angiogenesis assays.

When cultured with PC3CM, ROCK1 and ROCK2 expression increased in the HUVECs, as did pMYPT-1 and total MYPT-1 expression. The pMYPT-1/MYPT-1 ratio was also increased. This indicates activation of the RhoA/ROCK pathway in PC3CM-stimulated HUVECs. It has been reported that endothelial cells in PCa tissue from TRAMP mice, a spontaneous PCa mouse model, have a constitutively high baseline level of activity of Rho GTPase and its downstream effector ROCK (17). This suggests that the RhoA/ROCK pathway plays a crucial role in PCa angiogenesis. HUVECs cultured in PC3CM share some of the characteristics of PCa endothelium and can therefore be used to represent it.

Angiogenesis involves a complex series of events that take place in a multi-step process. Endothelial cells migrate through the basement membrane toward an angiogenic stimulus. The leading front of migrating cells is driven by enhanced proliferation of endothelial cells, followed by the formation of capillary tubes via endothelial cell organization. The RhoA/ROCK pathway plays a role in each of these steps.

We evaluated the effects of fasudil on each of these steps in PCa-induced HUVECs. Fasudil was found to inhibit PC3CM-induced HUVEC proliferation, migration, tube formation and spheroid sprouting. This is in accordance with previous studies on VEGF-induced endothelial cell proliferation, migration and tube formation after treatment with the RhoA inhibitor, C3, or ROCK inhibitors, Y-27632 and fasudil (15,16,18).

It is interesting to note the morphological changes that occurred in the spheroid sprouting assay after treatment with fasudil. After treatment with 10 μM fasudil, the sprouts were much thinner than those on untreated cells. However, the HUVEC nucleus was observed less frequently moving out of the spheroids than in the controls. The movement of the nucleus decreased as the fasudil concentration increased, whereas sprouting was not affected until the concentration of fasudil exceeded 30 μM. These sprouts were more akin to cell protrusions, were markedly thinner compared with PC3CM-induced HUVEC sprouts, and were more abundant and disorganized compared with the ordered architecture of single HUVEC spheroid sprouting.

In conclusion, fasudil significantly inhibits the key steps of endothelial cell angiogenesis, including proliferation, migration and capillary tube formation, in a dose-dependent manner. These effects may be due to inhibition of ROCK activity induced by PCa cell secretions. Fasudil may be a useful anti-angiogenic agent and should be investigated further for its potential role in the anti-angiogenic treatment of PCa.

Acknowledgements

This study was supported by a grant from The Program of International Science and Technology Cooperation (grant no. 2012DFG31440), awarded by The Ministry of Science and Technology, P.R. China. The authors are grateful to NewMed Publishing Services for providing final editing services.

References

1 

Bubendorf L, Schöpfer A, Wagner U, et al: Metastatic patterns of prostate cancer: An autopsy study of 1,589 patients. Hum Pathol. 31:578–583. 2000. View Article : Google Scholar : PubMed/NCBI

2 

Roehl KA, Han M, Ramos CG, Antenor JA and Catalona WJ: Cancer progression and survival rates following anatomical radical retropubic prostatectomy in 3,478 consecutive patients: long-term results. J Urol. 172:910–914. 2004. View Article : Google Scholar

3 

Hull GW, Rabbani F, Abbas F, Wheeler TM, Kattan MW and Scardino PT: Cancer control with radical prostatectomy alone in 1,000 consecutive patients. J Urol. 167:528–534. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Daneshgari F and Crawford ED: Endocrine therapy of advanced carcinoma of the prostate. Cancer. 71:1089–1097. 1993. View Article : Google Scholar : PubMed/NCBI

5 

Eymard JC, Oudard S, Gravis G, et al: Docetaxel reintroduction in patients with metastatic castration-resistant docetaxel-sensitive prostate cancer: a retrospective multicentre study. BJU Int. 106:974–978. 2010. View Article : Google Scholar

6 

Bigler SA, Deering RE and Brawer MK: Comparison of microscopic vascularity in benign and malignant prostate tissue. Hum Pathol. 24:220–226. 1993. View Article : Google Scholar : PubMed/NCBI

7 

Stefanou D, Batistatou A, Kamina S, Arkoumani E, Papachristou DJ and Agnantis NJ: Expression of vascular endothelial growth factor (VEGF) and association with microvessel density in benign prostatic hyperplasia and prostate cancer. In Vivo. 18:155–160. 2004.PubMed/NCBI

8 

Weidner N, Carroll PR, Flax J, Blumenfeld W and Folkman J: Tumor angiogenesis correlates with metastasis in invasive prostate carcinoma. Am J Pathol. 143:401–409. 1993.PubMed/NCBI

9 

Borre M, Offersen BV, Nerstrom B and Overgaard J: Microvessel density predicts survival in prostate cancer patients subjected to watchful waiting. Br J Cancer. 78:940–944. 1998. View Article : Google Scholar : PubMed/NCBI

10 

Green MM, Hiley CT, Shanks JH, et al: Expression of vascular endothelial growth factor (VEGF) in locally invasive prostate cancer is prognostic for radiotherapy outcome. Int J Radiat Oncol Biol Phys. 67:84–90. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Bok RA, Halabi S, Fei DT, et al: Vascular endothelial growth factor and basic fibroblast growth factor urine levels as predictors of outcome in hormone-refractory prostate cancer patients: a cancer and leukemia group B study. Cancer Res. 61:2533–2536. 2001.

12 

Peyromaure M, Camparo P, Badoual C, Descazeaud A and Dinh-Xuan AT: The expression of vascular endothelial growth factor is associated with the risk of cancer progression after radical prostatectomy. BJU Int. 99:1150–1153. 2007. View Article : Google Scholar : PubMed/NCBI

13 

George DJ, Halabi S, Shepard TF, et al: Prognostic significance of plasma vascular endothelial growth factor levels in patients with hormone-refractory prostate cancer treated on Cancer and Leukemia Group B 9480. Clin Cancer Res. 7:1932–1936. 2001.

14 

Etienne-Manneville S and Hall A: Rho GTPases in cell biology. Nature. 420:629–635. 2002. View Article : Google Scholar

15 

van Nieuw Amerongen GP, Koolwijk P, Versteilen A and van Hinsbergh VW: Involvement of RhoA/Rho kinase signaling in VEGF-induced endothelial cell migration and angiogenesis in vitro. Arterioscler Thromb Vasc Biol. 23:211–217. 2002.PubMed/NCBI

16 

Yin L, Morishige K, Takahashi T, et al: Fasudil inhibits vascular endothelial growth factor-induced angiogenesis in vitro and in vivo. Mol Cancer Ther. 6:1517–1525. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Ghosh K, Thodeti CK, Dudley AC, Mammoto A, Klagsbrun M and Ingber DE: Tumor-derived endothelial cells exhibit aberrant Rho-mediated mechanosensing and abnormal angiogenesis in vitro. Proc Natl Acad Sci USA. 105:11305–11310. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Bryan BA, Dennstedt E, Mitchell DC, et al: RhoA/ROCK signaling is essential for multiple aspects of VEGF-mediated angiogenesis. FASEB J. 24:3186–3195. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2014
Volume 32 Issue 6

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen W, Mao K, Hua-Huy T, Bei Y, Liu Z and Dinh-Xuan A: Fasudil inhibits prostate cancer-induced angiogenesis in vitro. Oncol Rep 32: 2795-2802, 2014
APA
Chen, W., Mao, K., Hua-Huy, T., Bei, Y., Liu, Z., & Dinh-Xuan, A. (2014). Fasudil inhibits prostate cancer-induced angiogenesis in vitro. Oncology Reports, 32, 2795-2802. https://doi.org/10.3892/or.2014.3491
MLA
Chen, W., Mao, K., Hua-Huy, T., Bei, Y., Liu, Z., Dinh-Xuan, A."Fasudil inhibits prostate cancer-induced angiogenesis in vitro". Oncology Reports 32.6 (2014): 2795-2802.
Chicago
Chen, W., Mao, K., Hua-Huy, T., Bei, Y., Liu, Z., Dinh-Xuan, A."Fasudil inhibits prostate cancer-induced angiogenesis in vitro". Oncology Reports 32, no. 6 (2014): 2795-2802. https://doi.org/10.3892/or.2014.3491