Peroxisome proliferator-activated receptor γ ligands inhibit VEGF-mediated vasculogenic mimicry of prostate cancer through the AKT signaling pathway

  • Authors:
    • Liang Qin
    • Ye Ren
    • An-Min Chen
    • Feng-Jing Guo
    • Fei Xu
    • Chen Gong
    • Peng Cheng
    • Yu Du
    • Hui Liao
  • View Affiliations

  • Published online on: April 29, 2014     https://doi.org/10.3892/mmr.2014.2198
  • Pages: 276-282
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Vasculogenic mimicry (VM) describes functional vascular channels composed only of tumor cells and its presence predicts a poor prognosis for patients with prostate cancer. The present study demonstrated that prostate cancer PC-3 cells were able to form a patterned matrix or tubular VM in 3D cultures in vitro and rosiglitazone (RSG), the ligand of peroxisome proliferator-activated receptor γ (PPARγ) and effectively inhibited the formation of VM structures in a dose- and PPARγ‑dependent manner. In addition, RSG significantly inhibited prostate cancer cell migration and invasion. The inhibitory effect of RSG on VM formation could be at least partially explained by an RSG-driven downregulation of vascular endothelial growth factor (VEGF) levels and phosphorylation of AKT, which is known to be important in VM. Furthermore, the present study highlighted that VEGF and the phosphoinositide 3-kinase/AKT pathway exert a positive feedback regulation in the process of VM formation. These findings reveal new therapeutic potential for PPARγ ligands in anti‑cancer therapy.

Introduction

The development and growth of a tumor requires a sufficient blood supply. It is a widely accepted paradigm that tumor vasculature is mostly generated through angiogenesis, the process of new vessel growth from pre-existing vessels (1). However, tumors do not only rely on host blood vessels for nourishment; they are also able to form their own vasculature (2). In 1999, Maniotis et al (3) reported that highly aggressive uveal melanomas are able to form vessels using tumor cells instead of endothelial cells and this phenomenon of tumor vascularization has been named vasculogenic mimicry (VM). VM has been described in prostate cancer and certain other types of highly aggressive tumors, and is associated with tumor cell migration and invasion (47). Several key molecules, including vascular endothelial (VE)-cadherin, matrix metalloproteinases (MMPs), laminin-5γ2 chains and CD147 have been implicated in VM (8,9). Several studies have demonstrated that vascular endothelial growth factor (VEGF) is important in the formation of VM and suggested that VEGF→ephrin type-A receptor (EphA2)→MMPs→VM is the main pathway for VM formation (10). Although VM has been demonstrated to occur in prostate cancer, a degree of which correlates with a poor clinical outcome (11), the precise molecular events underlying the process of VM in prostate cancer have remained to be fully elucidated.

Peroxisome proliferator-activated receptor (PPAR) is a member of the family of nuclear receptor transcription factors. PPARγ is expressed in numerous different tissues and regulates lipid metabolism, glucose homoeostasis and inflammation (12). PPARγ is the molecular target of the thiazolidinedione (TZD) class of antidiabetic drugs. The activation of PPARγ by TZDs and other ligands have been demonstrated to inhibit proliferation and invasion, and to induce apoptosis and cell cycle arrest in prostate and other cancer cells through PPARγ-dependent and -independent pathways (1315). Certain studies demonstrated that TZDs are able to induce the inhibition of angiogenesis in cancer and human umbilical vein endothelial cells (16,17). By contrast, several studies suggested that PPARγ activation may enhance tumor angiogenesis (18). In summary, the molecular mechanisms underlying the anticancer effects of TZD remain elusive.

Previous studies have demonstrated that the activation of PPARγ affects angiogenesis and VEGF production by tumor cells. In addition, VEGF is crucial in tumor angiogenesis and VM formation (10). Thus, the present study hypothesized that VM formation may be a potential target of PPARγ ligands. The present study investigated the effect of rosiglitazone (RSG), a ligand of PPARγ, on the formation of VM in PC-3 cells and the underlying mechanisms in vitro. To the best of our knowledge, the present study was the first to provide evidence that exposure of PC-3 cells to RSG induced decreased migration, invasion and formation of VM. The inhibitory effect of RSG on VM formation may at least partially be explained by an RSG-driven downregulation of VEGF and the phosphorylation of AKT.

Materials and methods

Cell culture

The human prostate cancer cell line PC-3 was maintained in RPMI-1640 medium (HyClone, Logan, UT, USA) supplemented with 10% fetal bovine serum (FBS) (Gibco-BRL, Grand Island, NY, USA), 1×105 U/l of penicillin and streptomycin (HyClone, Logan, UT, USA) at 37°C in an incubator with 5% CO2.

Three-dimensional cultures

Matrigel® (BD Biosciences, San Jose, CA, USA) was dropped onto glass coverslips and allowed to polymerize for 1 h at 37°C. The cells were then seeded on top of the gels at a high density and allowed to incubate. The addition of conditioned media containing 10% FBS was performed by the pretreatment and the continuous treatment regimes during the 48 h incubation period in three-dimensional cultures (19). Fresh media containing drugs was added every 24 h. For quantization of VM networks, the number of tube connections per field (magnification, ×40) was counted. The Student’s t-test was performed on at least five fields for each variable collected from three different experiments.

Cell migration assay

Cell migration was evaluated using an in vitro wound-healing assay. PC-3 cells were plated in a six-well plate and allowed to grow to confluence. The monolayer culture was then scrape-wounded with a sterile micropipette tip to create a denuded zone (gap) of constant width. The cells were exposed to various concentrations of RSG, 24 h after removing the cellular debris with phosphate-buffered saline (PBS). The speed of wound closure was monitored by measuring the ratio of the distance to that at 0 h. Each experiment was performed in triplicate and the results are presented as the mean ± standard error.

Cell invasion assay

The invasive potential was quantified using a Matrigel-coated transwell system, as previously described (20). The chamber (Corning Inc., Corning, NY, USA), which contained an 8-μm pore size polycarbonate membrane filter, was coated with Matrigel® and inserted into a 24 well culture plate. RSG was pre-incubated with PC-3 cells at various concentrations, respectively, for 24 h prior to seeding. The cell suspension (200 μl) was added into the upper chamber in serum-free medium and 500 μl of medium, which was supplemented with 20% FBS, was added into the lower chamber. Following 48 h incubation, the cells on the upper surface of the filters were completely removed by wiping with cotton swabs. Then cells that invaded the lower surface of the membrane were fixed with methanol and stained with crystal violet. Each experiment was conducted in triplicate. The cells were counted in five fields in each well by light microscopy. The cells were assessed for their relative invasion ability as percentages of the untreated controls.

Western blot analysis

Western blot analysis was performed to assess the protein levels of VE-cadherin and the expression and phosphorylation of signal transduction molecules, including AKT. Following being treated with RSG (Sigma-Aldrich, St. Louis, MO, USA), recombinant human (rh) VEGF165 (R&D Systems, Minneapolis, MN, USA), LY294002 (Cell Signaling Technology, Danvers, MA, USA) or GW9662 (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA), PC-3 cells were washed twice with PBS and treated with extraction buffer (50 mM of Tris-Cl; pH 7.5; 150 mM of NaCl, 0.1% SDS, 1% NP-40 and 0.5% deoxycholic acid). The cell extractions were collected and centrifuged at 10,000 × g for 10 min at 4°C, and the supernatants were collected as cell lysates. The cell lysates were subjected to SDS-PAGE and transferred onto polyvinylidene fluoride membranes (Millipore, Bedford, MA, USA). The membranes were inhibited with 5% (w/v) bovine serum albumin in Tris-buffered saline containing 0.1% Tween-20 and then blotted with primary antibody. Subsequently, the membranes were incubated with an appropriate secondary antibody (horseradish peroxidase-conjugated goat anti-mouse or anti-rabbit immunoglobulin G; Wuhan Boster Biological Technology, Ltd, Wuhan, Hubei, China). The immune-detected proteins were then visualized by enhanced chemiluminescence. The untreated cells served as a negative controls and β-actin was used as a control for equal loading. The antibodies against VE-cadherin, AKT, p-AKT (Ser473) and β-actin were purchased from Cell Signaling Technology (Beverly, MA, USA).

ELISA assay

Conditioned medium was prepared as mentioned above and VEGF concentrations in the medium were determined using a commercial Human VEGF Quantikine ELISA kit (R&D Systems) according to the manufacturer’s instructions. The absorbance at 450 nm was measured and corrected using the 540 nm reading on an ELX-800 uv microplate reader (Bio-Tek Instruments, Inc, Winooski, VT, USA).

quantitative polymerase chain reaction (qPCR) assay

Total RNA was extracted using TRIzol reagent according to the manufacturer’s instructions (Invitrogen Life Technologies, Carlsbad, CA, USA). RNA (1 μg) was reverse-transcribed. qPCR was accomplished with TransStart Green qPCR SuperMix (Beijing TransGen Biotech Co., Ltd., Beijing, China) using a Bio-Rad iQ5 Sequence Detection System (Bio-Rad, Hercules, CA, USA). The PCR conditions were as follows: one cycle at 95°C for 10 min followed by 40 cycles at 95°C for 15 sec and 60°C for 1 min. The results were normalized to those obtained with GAPDH. Each sample was assayed in triplicate and the experiment was repeated twice. The CT value was measured and 2−ΔCT (ΔCT = CT - CTGAPDH) was defined as the quantity of the amplified fragment. The primer sequences are listed in Table I.

Table I

Primer sequences.

Table I

Primer sequences.

GeneForward (5′-3′)Reverse (5′-3′)
GAPDH CCATGAGAAGTATGACAACAGCC GGGTGCTAAGCAGTTGGTG
VE-cadherin GACGCCCGGCCTTCCCTCTA TCGTGGTCCGCCTCGTCCTT
VEGF TTACGGTCTGTGTCCAGTGTA TTCTCTGTTATGTTGCCAGCC

[i] VEGF, vascular endothelial growth factor; VE-cadherin, vascular endothelial cadherin.

Statistical analysis

Data are expressed as the mean ± standard deviation. Statistical analyses were performed using SPSS 11.0 statistical software (SPSS, Inc., Chicago, IL, USA). Statistical significance was analyzed by one-way analysis of variance. If significance was observed, the Dunnett’s post-hoc test was used to determine the difference between treatment groups and the untreated group. P<0.05 was considered to indicate a statistically significant difference.

Results

RSG inhibits the migration and invasion of PC-3 cells

VM is associated with cell migration and invasion. To examine the effect of RSG on PC-3 cells, an in vitro wound healing assay was performed. Since TZDs activate PPARα and PPARδ receptors at concentrations >10 μM (21), TZDs were used at concentrations of 10 μM or less. Following incubation with different concentrations of RSG for 24 h, the migration of PC-3 cells to the denuded zone was suppressed in a dose-dependent manner (Fig. 1Aa and 1B). These results revealed that RSG significantly inhibited the motility of PC-3 cells.

Using a chamber invasion assay, it was revealed that RSG suppressed the invasion of PC-3 cells across the Matrigel-coated filter in a dose-dependent manner. Treatment with 0.1, 1 and 10 μM of RSG inhibited 17.01, 27.62 and 35.74% of cell invasion, respectively (Fig. 1Ab and 1C). The results indicated that RSG markedly inhibited the invasion of PC-3 cells.

RSG prevents the development of VM in a PPARγ-dependent manner

In order to elucidate the VM capability of PC-3 cells, a well-established in vitro model of VM formation was utilized. PC-3 cells were able to form patterned matrix VM or typical pipe-like VM networks within the Matrigel medium within 48 h.

Next, the present study investigated the effect of RSG on VM formed by PC-3 cells. The in vitro tube formation assay demonstrated that RSG effectively inhibited the formation of capillary-like structures in a dose-dependent manner (Fig. 2A and B). Western blot analysis of whole-cell lysates from these experimental samples was conducted to assess the expression of VM-associated markers, including VE-cadherin. The results demonstrated that RSG significantly decreased the expression of VE-cadherin at the mRNA and protein levels (P<0.05; Fig. 2C and D).

Although RSG was able to affect the VM process and the expression of VE-cadherin, the mechanisms underlying these events remain to be elucidated. To confirm whether these effects were dependent upon the activation of PPARγ, GW9662, a PPARγ antagonist, was used to inhibit the function of PPARγ in prostate cancer cells. The results demonstrated that GW9662 inhibited RSG-induced reduction of VM formation. Furthermore, the RSG-induced downregulation of VE-cadherin in PC-3 cells was attenuated upon addition of GW9662 (Fig. 2). Taken together, these data suggested that RSG inhibited the VM formation of PC-3 cells in a PPARγ-dependent manner.

Effect of RSG on the expression of VEGF

Since prostate cancer cells induce angiogenesis through the expression of angiogenic factors, including VEGF, the present study evaluated whether VEGF stimulates the formation of VM in PC-3 cells, utilizing an in vitro tube formation assay. Quantification of the tubule number demonstrated that rhVEGF165 effectively promoted the formation of capillary-like structures (Fig. 5A and B). Furthermore, RSG inhibited the expression of VEGF in PC-3 cells. The secretion of VEGF protein was reduced to 59.38, 39.64 and 29.99% in PC-3 cells in the presence of RSG as compared with the control (Fig. 3A). Similar results were observed while detecting the VEGF mRNA levels by qPCR (Fig. 3B). Furthermore, the RSG-induced downregulation of VEGF in PC-3 cells was attenuated upon addition of GW9662 (Fig. 3). The present study also demonstrated that the addition of rhVEGF165 partly inhibited RSG-induced downregulation of VM formation and VE-cadherin expression (Fig. 5A and C). These data suggest that the inhibition VM of PC-3 cells by RSG may partly occur through suppressing VEGF expression.

RSG modulates the expression of VEGF and VM via the AKT pathway

Several studies have indicated that signaling proteins, including PI3K and AKT, are involved in the regulation of the expression of MMPs and the promotion of metastasis (22). To understand the signaling pathways responsible for VEGF production and VM formation, the activation of AKT was examined. As expected, RSG downregulated the phosphorylation of AKT, the active form of PI3K/AKT, in a dose- and time-dependent manner (Fig. 4), which indicated that AKT may also be involved in the inhibition of VM formation in PC-3 cells.

To further investigate whether the inhibition of VM formation and VE-cadherin expression proceeded through the inhibition of the PI3K-AKT signaling pathway, PC-3 cells were treated with a PI3K inhibitor (LY294002; 20 μM) for 48 h. This disturbance of the AKT pathway resulted in the downregulation of VEGF secretion in PC-3 cells (Fig. 5D), which is consistent with the results of other studies (23). In addition, treatment with LY294002 significantly reduced cell VM formation and VE-cadherin expression (Fig. 5A and C), suggesting that the inhibition of VM formation and VEGF expression by RSG partly occurred through suppressing the PI3K pathway. Of note, the addition of rhVEGF165 eradicated the inhibitory effect of RSG on the phosphorylation of AKT (Fig. 4A). These data suggest that the inactivation of the PI3K/AKT pathway by RSG may be mediated by the inhibition of VEGF in an autocrine manner.

Discussion

Globally, 903,500 new cases of prostate cancer were estimated to occur, second only to lung cancer in males, and 258,400 mortalities from prostate cancer were estimated for 2011 (24). The majority of patients with localized early stage disease do not develop metastases and do not succumb to prostate cancer. However, among patients with castration-resistant metastatic prostate cancer, >80% of patients develop bone metastasis, which is the most common site of metastases in this group (25). Therefore, understanding the molecular mechanisms of prostate cancer invasiveness and angiogenesis are of significant value to the prostate cancer field.

Targeting the blood supply by inhibiting the formation of blood vessels may lead to tumor growth arrest. Numerous angiogenesis inhibitors have been therapeutically used in preclinical and clinical settings (26). VEGF receptor tyrosine kinase inhibitors and a VEGF-neutralizing antibodies have been clinically validated to target VEGF or its receptors as an anticancer treatment. However, amongst other limitations of angiogenesis inhibitors, tubule formation was not inhibited. For instance, treatment with angiogenesis inhibitors did not inhibit tube formation of aggressive uveal and cutaneous melanoma cells in vitro (27). Of note, recent findings indicated that anti-angiogenesis treatment may elicit the malignant progression of various types of tumor (28). Furthermore, the inhibition of endothelial angiogenesis may unintentionally promote VM formation in tumors (29), as recently evidenced by a study demonstrating increased VM channels in tumor-bearing mice receiving short-term treatment of the anti-VEGF monoclonal antibody bevacizumab (30). It has been demonstrated that several drugs were able to inhibit VM (31,32). However, at present, data on the effect of anti-angiogenesis treatment on VM network formation remain inconsistent. The inhibition of endothelial cell migration by PPARγ ligands has been described, bolstering the anti-angiogenic activity of PPAR ligands (17,33). An earlier study has demonstrated that the activation of PPARγ by TZDs inhibits angiogenesis and neovascularization in vitro and in vivo and inhibits the release of VEGF from smooth muscle cells. The present study revealed that RSG was able to inhibit VM formation of the highly aggressive prostate cancer cell line PC-3 in vitro in a dose- and PPARγ-dependent manner. The results demonstrated that RSG significantly decreased the expression of VM-associated markers, including VE-cadherin, at the mRNA and protein levels, in line with the results of functional studies. RSG also reduced the migration and invasion capacity of PC-3 cells. Therefore, PPARγ agonists may be further used to unravel new biological mechanisms that drive VM, angiogenesis and tumor progression.

VEGF and its receptors appear to contribute to VM formation in certain types of cancer (10,34). However, VEGF did not demonstrate any effect on VM formation of Ewing sarcoma cells and hepatocellular carcinoma cell lines (35,36), and the effect of VEGF on the VM formation of prostate cancer cells remains to be elucidated. The present study demonstrated that rhVEGF165 increased the VM formation of PC-3 cells. Notably, it was demonstrated that VEGF was downregulated at the mRNA and protein secretion levels upon RSG treatment. These effects were dose dependent and the concentrations used in the present study were consistent with those reported by others (17). However, another study reported that TZDs enhanced VEGF expression in non-small cell lung cancer cells (37). The variation of outcome from those studies may be due to differences in culture and experimental conditions, the type and site of carcinoma or a combination of these factors.

Besides the functional study, elucidation of the mechanisms is crucial for understanding RSG as a potential anticancer agent. PI3K and AKT are important for the signaling of angiogenic growth factors that are closely associated with cancer cell migration, proliferation, angiogenesis and VM (38,39). Although LY294002 is useful for conducting preclinical studies on PI3K inhibition, its poor solubility and narrow therapeutic index preclude its use in humans. It has been reported that the PPARγ agonist was able to decrease the phosphorylation of AKT (33,40). The present study used the PPARγ agonist, RSG, to stimulate PC-3 cells. Our results demonstrated that RSG downregulated the phosphorylation of AKT in a dose- and time-dependent manner. In order to examine this possibility, the PI3K inhibitor LY294002 was used to inhibit these pathways to establish a link with VM formation. The data demonstrated that the disturbance of the AKT pathway resulted in the inhibition of the formation of VM in PC-3 cells and the downregulation of VE-cadherin and VEGF expression in PC-3 cells. Therefore, the present study suggested that RSG inhibited VM formation of PC-3 cells, which may be partly through suppressing PI3K-associated pathways. In addition, the stimulation of the cells with rhVEGF165 in the culture medium attenuated the inhibitory effect of RSG on the AKT pathway and cancer phenotypes. Previous studies demonstrated that the PI3K/AKT pathway may be activated by growth factors or hypoxia and are able to promote tumor angiogenesis through the enhanced expression of hypoxia-inducible factor 1 and VEGF (41). In addition, in the present study and in other studies (42,43), suppression of these pathways by a PI3K inhibitor markedly reduced VEGF protein levels. Thus, the present study concluded that the PI3K/AKT pathway and VEGF may generate a positive autocrine loop. These findings associated with the dual inhibition of the VEGF/PI3K/AKT cascade and the PI3K/AKT/VEGF cascade by RSG treatment in prostate cancer cells provided insights into the molecular mechanisms of the anti-tumor effects of RSG and support its potential clinical application.

In conclusion, the present study indicated that RSG effectively inhibited VM formation of PC-3 cells in vitro by inhibiting VEGF gene expression and phosphorylation of AKT. The present study may provide preliminary evidence for further elucidating the mechanisms underlying VM inhibition by TZDs and its mode of action. RSG was used as a proof-of-principle to determine the efficacy of PPARγ agonists in the inhibition of VM formation.

Acknowledgements

This study was supported by a grant from the Health Department of Hubei Province’s Program for Youth Science and Technology Talent (QJX2010-5) and the Key Laboratory of Cancer Invasion and Metastasis of the Ministry of Education of People’s Republic of China (no. 200804871-051).

References

1 

Hanahan D and Folkman J: Patterns and emerging mechanisms of the angiogenic switch during tumorigenesis. Cell. 86:353–364. 1996. View Article : Google Scholar : PubMed/NCBI

2 

Döme B, Hendrix MJ, Paku S, Tóvári J and Tímár J: Alternative vascularization mechanisms in cancer: Pathology and therapeutic implications. Am J Pathol. 170:1–15. 2007.PubMed/NCBI

3 

Maniotis AJ, Folberg R, Hess A, et al: Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol. 155:739–752. 1999. View Article : Google Scholar : PubMed/NCBI

4 

Basu GD, Pathangey LB, Tinder TL, Gendler SJ and Mukherjee P: Mechanisms underlying the growth inhibitory effects of the cyclo-oxygenase-2 inhibitor celecoxib in human breast cancer cells. Breast Cancer Res. 7:R422–R435. 2005. View Article : Google Scholar : PubMed/NCBI

5 

El Hallani S, Boisselier B, Peglion F, et al: A new alternative mechanism in glioblastoma vascularization: tubular vasculogenic mimicry. Brain. 133:973–982. 2010.PubMed/NCBI

6 

Sharma N, Seftor RE, Seftor EA, et al: Prostatic tumor cell plasticity involves cooperative interactions of distinct phenotypic subpopulations: role in vasculogenic mimicry. Prostate. 50:189–201. 2002. View Article : Google Scholar

7 

Sun B, Zhang D, Zhang S, Zhang W, Guo H and Zhao X: Hypoxia influences vasculogenic mimicry channel formation and tumor invasion-related protein expression in melanoma. Cancer Lett. 249:188–197. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Millimaggi D, Mari M, D’ Ascenzo S, Giusti I, Pavan A and Dolo V: Vasculogenic mimicry of human ovarian cancer cells: role of CD147. Int J Oncol. 35:1423–1428. 2009.PubMed/NCBI

9 

Zhang S, Zhang D and Sun B: Vasculogenic mimicry: current status and future prospects. Cancer Lett. 254:157–164. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Wang JY, Sun T, Zhao XL, et al: Functional significance of VEGF-a in human ovarian carcinoma: role in vasculogenic mimicry. Cancer Biol Ther. 7:758–766. 2008. View Article : Google Scholar : PubMed/NCBI

11 

Liu R, Yang K, Meng C, Zhang Z and Xu Y: Vasculogenic mimicry is a marker of poor prognosis in prostate cancer. Cancer Biol Ther. 13:527–533. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Kota BP, Huang TH and Roufogalis BD: An overview on biological mechanisms of PPARs. Pharmacol Res. 51:85–94. 2005. View Article : Google Scholar : PubMed/NCBI

13 

Kim S, Lee JJ and Heo DS: PPARgamma ligands induce growth inhibition and apoptosis through p63 and p73 in human ovarian cancer cells. Biochem Biophys Res Commun. 406:389–395. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Lyles BE, Akinyeke TO, Moss PE and Stewart LV: Thiazolidinediones regulate expression of cell cycle proteins in human prostate cancer cells via PPARgamma-dependent and PPARgamma-independent pathways. Cell Cycle. 8:268–277. 2009. View Article : Google Scholar

15 

Fujita M, Yagami T, Fujio M, et al: Cytotoxicity of troglitazone through PPARgamma-independent pathway and p38 MAPK pathway in renal cell carcinoma. Cancer Lett. 312:219–227. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Kim KY, Ahn JH and Cheon HG: Anti-angiogenic action of PPARgamma ligand in human umbilical vein endothelial cells is mediated by PTEN upregulation and VEGFR-2 downregulation. Mol Cell Biochem. 358:375–385. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Panigrahy D, Singer S, Shen LQ, et al: PPARgamma ligands inhibit primary tumor growth and metastasis by inhibiting angiogenesis. J Clin Invest. 110:923–932. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Tian L, Zhou J, Casimiro MC, et al: Activating peroxisome proliferator-activated receptor gamma mutant promotes tumor growth in vivo by enhancing angiogenesis. Cancer Res. 69:9236–9244. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Lissitzky JC, Parriaux D, Ristorcelli E, Verine A, Lombardo D and Verrando P: Cyclic AMP signaling as a mediator of vasculogenic mimicry in aggressive human melanoma cells in vitro. Cancer Res. 69:802–809. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Liu H, Chen A, Guo F and Yuan L: A short-hairpin RNA targeting osteopontin downregulates MMP-2 and MMP-9 expressions in prostate cancer PC-3 cells. Cancer Lett. 295:27–37. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Yamashita D, Shimizu M and Osumi T: Mechanism for the action of PPARs. Nihon Rinsho. 63:536–537. 2005.(In Japanese).

22 

Shukla S, Maclennan GT, Hartman DJ, Fu P, Resnick MI and Gupta S: Activation of PI3K-Akt signaling pathway promotes prostate cancer cell invasion. Int J Cancer. 121:1424–1432. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Wang J, Wang J, Sun Y, Song W, Nor JE, Wang CY and Taichman RS: Diverse signaling pathways through the SDF-1/CXCR4 chemokine axis in prostate cancer cell lines leads to altered patterns of cytokine secretion and angiogenesis. Cell Signal. 17:1578–1592. 2005. View Article : Google Scholar : PubMed/NCBI

24 

Jemal A, Bray F, Center MM, Ferlay J, Ward E and Forman D: Global cancer statistics. CA Cancer J Clin. 61:69–90. 2011. View Article : Google Scholar

25 

de Bono JS, Oudard S, Ozguroglu M, et al: Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet. 376:1147–1154. 2010.

26 

Samaranayake H, Määttä AM, Pikkarainen J and Ylä-Herttuala S: Future prospects and challenges of antiangiogenic cancer gene therapy. Hum Gene Ther. 21:381–396. 2010. View Article : Google Scholar : PubMed/NCBI

27 

van der Schaft DW, Seftor RE, Seftor EA, et al: Effects of angiogenesis inhibitors on vascular network formation by human endothelial and melanoma cells. J Natl Cancer Inst. 96:1473–1477. 2004.PubMed/NCBI

28 

Pàez-Ribes M, Allen E, Hudock J, et al: Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell. 15:220–231. 2009.PubMed/NCBI

29 

Qu B, Guo L, Ma J and Lv Y: Antiangiogenesis therapy might have the unintended effect of promoting tumor metastasis by increasing an alternative circulatory system. Med Hypotheses. 74:360–361. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Xu Y, Li Q, Li XY, Yang QY, Xu WW and Liu GL: Short-term anti-vascular endothelial growth factor treatment elicits vasculogenic mimicry formation of tumors to accelerate metastasis. J Exp Clin Cancer Res. 31:162012. View Article : Google Scholar : PubMed/NCBI

31 

Itzhaki O, Greenberg E, Shalmon B, et al: Nicotinamide inhibits vasculogenic mimicry, an alternative vascularization pathway observed in highly aggressive melanoma. PLoS One. 8:e571602013. View Article : Google Scholar

32 

Fan YZ and Sun W: Molecular regulation of vasculogenic mimicry in tumors and potential tumor-target therapy. World J Gastrointest Surg. 2:117–127. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Goetze S, Eilers F, Bungenstock A, et al: PPAR activators inhibit endothelial cell migration by targeting Akt. Biochem Biophys Res Commun. 293:1431–1437. 2002. View Article : Google Scholar : PubMed/NCBI

34 

Vartanian A, Stepanova E, Grigorieva I, Solomko E, Baryshnikov A and Lichinitser M: VEGFR1 and PKCalpha signaling control melanoma vasculogenic mimicry in a VEGFR2 kinase-independent manner. Melanoma Res. 21:91–98. 2011. View Article : Google Scholar : PubMed/NCBI

35 

van der Schaft DW, Hillen F, Pauwels P, et al: Tumor cell plasticity in Ewing sarcoma, an alternative circulatory system stimulated by hypoxia. Cancer Res. 65:11520–11528. 2005.PubMed/NCBI

36 

Lirdprapamongkol K, Chiablaem K, Sila-Asna M, Surarit R, Bunyaratvej A and Svasti J: Exploring stemness gene expression and vasculogenic mimicry capacity in well- and poorly-differentiated hepatocellular carcinoma cell lines. Biochem Biophys Res Commun. 422:429–435. 2012. View Article : Google Scholar

37 

Yoshizaki T, Motomura W, Tanno S, Kumei S, Yoshizaki Y and Okumura T: Thiazolidinediones enhance vascular endothelial growth factor expression and induce cell growth inhibition in non-small-cell lung cancer cells. J Exp Clin Cancer Res. 29:222010. View Article : Google Scholar

38 

Hess AR, Seftor EA, Seftor RE and Hendrix MJ: Phosphoinositide 3-kinase regulates membrane Type 1-matrix metalloproteinase (MMP) and MMP-2 activity during melanoma cell vasculogenic mimicry. Cancer Res. 63:4757–4762. 2003.PubMed/NCBI

39 

Chetty C, Lakka SS, Bhoopathi P and Rao JS: MMP-2 alters VEGF expression via alphaVbeta3 integrin-mediated PI3K/AKT signaling in A549 lung cancer cells. Int J Cancer. 127:1081–1095. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Chen WC, Lin MS and Bai X: Induction of apoptosis in colorectal cancer cells by peroxisome proliferators-activated receptor gamma activation up-regulating PTEN and inhibiting PI3K activity. Chin Med J (Engl). 118:1477–1481. 2005.PubMed/NCBI

41 

Trisciuoglio D, Iervolino A, Zupi G and Del Bufalo D: Involvement of PI3K and MAPK signaling in bcl-2-induced vascular endothelial growth factor expression in melanoma cells. Mol Biol Cell. 16:4153–4162. 2005. View Article : Google Scholar : PubMed/NCBI

42 

Pore N, Gupta AK, Cerniglia GJ and Maity A: HIV protease inhibitors decrease VEGF/HIF-1alpha expression and angiogenesis in glioblastoma cells. Neoplasia. 8:889–895. 2006. View Article : Google Scholar : PubMed/NCBI

43 

Zhong H, Chiles K, Feldser D, et al: Modulation of hypoxia-inducible factor 1alpha expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics. Cancer Res. 60:1541–1545. 2000.

Related Articles

Journal Cover

July-2014
Volume 10 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Qin L, Ren Y, Chen A, Guo F, Xu F, Gong C, Cheng P, Du Y and Liao H: Peroxisome proliferator-activated receptor γ ligands inhibit VEGF-mediated vasculogenic mimicry of prostate cancer through the AKT signaling pathway. Mol Med Rep 10: 276-282, 2014
APA
Qin, L., Ren, Y., Chen, A., Guo, F., Xu, F., Gong, C. ... Liao, H. (2014). Peroxisome proliferator-activated receptor γ ligands inhibit VEGF-mediated vasculogenic mimicry of prostate cancer through the AKT signaling pathway. Molecular Medicine Reports, 10, 276-282. https://doi.org/10.3892/mmr.2014.2198
MLA
Qin, L., Ren, Y., Chen, A., Guo, F., Xu, F., Gong, C., Cheng, P., Du, Y., Liao, H."Peroxisome proliferator-activated receptor γ ligands inhibit VEGF-mediated vasculogenic mimicry of prostate cancer through the AKT signaling pathway". Molecular Medicine Reports 10.1 (2014): 276-282.
Chicago
Qin, L., Ren, Y., Chen, A., Guo, F., Xu, F., Gong, C., Cheng, P., Du, Y., Liao, H."Peroxisome proliferator-activated receptor γ ligands inhibit VEGF-mediated vasculogenic mimicry of prostate cancer through the AKT signaling pathway". Molecular Medicine Reports 10, no. 1 (2014): 276-282. https://doi.org/10.3892/mmr.2014.2198