Inhibition of lymphangiogenic factor VEGF-C expression and production by the histone deacetylase inhibitor suberoylanilide hydroxamic acid in breast cancer cells

  • Authors:
    • Hsueh-Tsen Cheng
    • Wen-Chun Hung
  • View Affiliations

  • Published online on: December 14, 2012     https://doi.org/10.3892/or.2012.2188
  • Pages: 1238-1244
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Suberoylanilide hydroxamic acid (SAHA), a potent histone deacetylase (HDAC) inhibitor, has been shown to exert anticancer effects in various types of human cancer and is now used in the clinic for cancer treatment. In addition to cytostatic and cytotoxic activities, SAHA also represses angiogenesis to inhibit tumor growth. However, the effect of SAHA on tumor lymphangiogenesis, a step in which cancer cells produce pro-lymphangiogenic factors such as vascular endothelial growth factor-C (VEGF-C) to stimulate proliferation and migration of lymphatic endothelial cells, remains largely unclear. In this study, we investigated the expression of VEGF-C in breast cancer cell lines and found that VEGF-C was highly expressed in MDA-MB-231, MCF-7, MDA-MB-453 and BT-474 cells. SAHA inhibited VEGF-C expression in a dose-dependent manner in these cell lines. The secretion of VEGF-C into conditioned medium was also suppressed. We cloned human VEGF-C gene promoter and demonstrated that SAHA directly repressed promoter activity in MDA-MB-231 cells. Promoter deletion assay suggested that SAHA repressed VEGF-C via the -185/+38 region which contained several transcription factor binding sites. Notably, we found that SAHA reduced Sp1, but not Sp3 and NF-κB protein levels. Treatment with Sp1 inhibitor mithramycin A also inhibited VEGF-C expression in breast cancer cells. In addition, enforced expression of Sp1 partially rescued the inhibition of VEGF-C by SAHA. Collectively, our results suggest that SAHA inhibits VEGF-C expression in breast cancer cells via transcriptional repression and this drug may exert anti-lymphangiogenic activity in cancer treatment.

Introduction

Epigenetic alteration is a general phenomenon during carcinogenesis (1). Both DNA methylation and histone modification play critical roles in the control of expression of oncogenes and tumor suppressor genes (2,3). One of the most important histone modifications is acetylation which is controlled by histone acetyltransferases (HATs) and histone deacetylases (HDACs). These enzymes are considered to be crucial targets for the development of anticancer drugs. Previous studies demonstrated that HDAC inhibitors show potent anticancer activities against different types of human cancer (46).

HDAC inhibitors are classified into different groups based on their chemical structures and their ability to inhibit individual HDACs (7,8). The first class is short chain fatty acids including butyrate and valproate. The second class is hydroxamic acid derivatives such as trichostatin A, suberoylanilide hydroxamic acid (SAHA, also known as vorinostat). The third class is benzamide and the fourth class is cyclic tetrapeptide. Among them, SAHA is one of the pioneer HDAC inhibitors that has entered clinical therapy and is now approved for the treatment of cutaneous T cell lymphoma (9,10). This drug is also undergoing clinical trials for the treatment of solid tumors, including non-small cell lung cancer and breast cancer. The anticancer effects of SAHA are mediated by different mechanisms. First, SAHA directly causes apoptosis in cancer cells (11,12). Second, SAHA upregulates anti-proliferative genes, such as p21, to inhibits proliferation (13,14). Third, SAHA acts as an immune modulating drug to improve anticancer immune surveillance (15,16). Fourth, SAHA suppresses angiogenesis to attenuates tumor growth in vivo(17).

Lymphangiogenesis is the process by which cancer cells promote the proliferation of lymphatic endothelial cells and enhance the migration of these cells toward tumor part (18). Previous studies demonstrated that induction of lymphangiogenesis is strongly associated with tumor metastasis and poor prognosis in cancer patients (1921). The key step in inducing lymphangiogenesis is the production of lymphangiogenic factors by cancer cells. Among the lymphangiogenic factors studied, vascular endothelial growth factor-C (VEGF-C) has received considerable attention since lymphatic endothelial cells express high levels of its cognate receptor VEGFR3 and their proliferation is potently stimulated by VEGF-C.

Although SAHA was able to inhibit angiogenesis in different types of cancer, its effect on lymphangiogenesis has not been demonstrated. In this study, we investigated the expression of VEGF-C in breast cancer cell lines and its regulation by SAHA. In addition, we tried to elucidate the underlying mechanism by which SAHA modulated VEGF-C transcription.

Materials and methods

Cell culture

Human breast cancer cell lines MDA-MB-231, MCF-7, MDA-MB-453 and the normal breast cell line M10 were purchased from the cell bank of the National Health Research Institute (Maoli, Taiwan). MDA-MB-231 and MDA-MB-453 cells were grown in the L15 medium (Invitrogen, Carlsbad, CA, USA) containing 10% FCS, L-glutamate and antibiotics. MCF-7 and M10 cells were cultured in the MEM medium (Invitrogen) containing 10% FCS and antibiotics. BT-474 breast cancer cells were kindly provided by Dr Jin-Yuh Shew (National Taiwan University, Taiwan) and were maintained in the DMEM/F12 medium (Invitrogen) supplemented with 10% FCS, L-glutamate, nonessential amino acids, sodium pyruvate and antibiotics. All cells were cultured at 37°C in a humanized incubator with 5% CO2.

Reagents and antibodies

SAHA was purchased from LC Laboratories (Woburn, MA, USA). SAHA was dissolved in DMSO at a concentration of 20 mM and stored at −70°C. It was diluted in the culture medium to different working concentrations and used for cell treatment. Mithramycin A was purchased from Sigma-Aldrich (St. Louis, MO, USA). Antibody against VEGF-C was purchased from R&D Systems (Minneapolis, MN, USA). Antibody against NF-κB (p65) and Sp3 were obtained from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Antibodies against actin and Sp1 were purchased from Millipore (Billerica, MA, USA). Antibody against acetyl lysine-histone 3 was purchased from Cell Signaling Technology (Beverly, MA, USA).

Construction of VEGF-C promoter plasmids and the luciferase assay

Genomic DNA was extracted from MCF-7 cells using an extraction kit (Qiagen, Hilden, Germany) and the −1046/+38 region from translational start site of VEGF-C gene (NM_005429) was amplified by PCR using two specific primers (VEGF-C-1046-forward 5′-ATACTCGAGCTTTTACAACCCCCAGG ACA-3′ and VEGF-C-1046-reverse 5′-AAAAAGCTTAGAGA ACACGCCACAGAGAAG-3′). A 1.1-kb DNA fragment was subcloned into the luciferase reporter gene vector pGL3 (Promega, Madison, WI, USA) by XhoI and HindIII to yield the luciferase reporter construct pGL3-1.1kb-VEGF-C (-1046/+38). Using this construct as a template, two 5′-deletion constructs were generated by the following primers: VEGF-C-439-forward 5′-ATACTCGAGCTCTCACTTCGGGGAAGG-3′ and VEGF-C-185-forward 5′-ATACTCGAGGCCCTGCAAAGTT GGGAAC-3′. MDA-MB-231 cells were seeded into 6-well plates and transfected with 1 μg of serial VEGF-C promoter-luciferase plasmids. After 24 h, cells were incubated without or with SAHA (10 μM) for an additional 24 h. The luciferase activity was detected by reporter assay system (Promega), according to the manufacturer’s instructions, and normalized by protein concentration in cell lysates.

Transient cell transfection

GFP-Sp1 expression vector (kindly provided by Dr Jan-Jong Hung, National Cheng Kung University, Taiwan) and control vector were transfected into MDA-MB-231 cells using Lipofectamine 2000 (Invitrogen). After 48 h, cells were treated with SAHA for an additional 24 h. Conditioned medium and cell lysates were harvested for further analysis.

Reverse transcription-polymerase chain reaction (RT-PCR)

MDA-MB-231, MCF-7, BT-474 and M10 breast cells were seeded into the 6-well plates. Following overnight incubation, cells were treated with different concentrations of SAHA for 24 h. Total RNA was isolated by RNeasy mini kit (Qiagen) and mRNAs were reverse-transcribed to cDNA by MMLV reverse transcriptase (Promega) using oligo-dT primers according to the manufacturer’s instructions. PCR reaction was performed under the following conditions: initialization step for 5 min at 95°C, 30 cycles of amplification, with 40 sec at 95°C for denaturation and 40 sec at 60°C for annealing and 40 sec at 72°C for elongation, and 7 min at 72°C for further extension. The PCR primers used are shown in Table I.

Table I

PCR primers used in this study.

Table I

PCR primers used in this study.

VEGF-CF: 5′-CAGTTACGGTCTGTGTCCAGTGTAG-3′
VEGF-CR: 5′-GGACACACATGGAGGTTTAAAGAAG-3′
Sp1F: 5′-TATAGCAAATGCCCCAGGT-3′
Sp1R: 5′-TTGCCATACACTTTCCCACA-3′
Sp3F: 5′-CCTGCAGATATTAGGATCAAGG-3′
Sp3R: 5′-GCCTCTGTAATTCATCACTTCG-3′
RelAF: 5′-TCAATGGCTACACAGGACCA-3′
RelAR: 5′-ATCTTGAGCTCGGCAGTGTT-3′
ActinF: 5′-TGTTACCAACTGGGACGACA-3′
ActinR: 5′-GGGGTGTTGAAGGTCTCAAA-3′

[i] F, forward; R, reverse.

Enzyme-linked immunosorbent assay (ELISA) for VEGF-C

MDA-MB-231 cells were incubated under various concentrations of SAHA in L15 medium for 24 h. The conditioned medium was collected and centrifuged at 1500 rpm for 5 min to remove cell debris. VEGF-C concentration was measured using Quantikines Human VEGF-C Immunoassay kit from R&D.

Western blotting

Cells were seeded into the 6-well plates. Following overnight incubation, cells were treated with different concentrations of SAHA for 6, 12 or 24 h. Cells were washed with cold PBS and lysed in RIPA buffer (50 mM Tris-HCl, pH 7.4, 50 mM NaCl, 1 mM EDTA, 0.5 M sucrose, 0.25% sodium deoxycholate, 10% glycerol, 1% NP-40; protease inhibitors were added prior to use) on ice for 10 min. Cell debris was removed by centrifugation and cellular proteins were further fractionated by SDS-PAGE and transferred onto polyvinylidene difluoride membranes (Millipore). After incubation with 5% non-fat milk in TBST, the membranes were probed with different primary antibodies followed by horseradish peroxidase-conjugated anti-mouse or anti-rabbit antibodies. Enhanced chemiluminescence (ECL) reagent (Millipore) was used to detect the blots according to the manufacturer’s instructions.

Statistical analysis

Data were expressed as the means ± SE. Student’s t-test was used to evaluate the differences between various experimental groups. p<0.05 was considered to indicate a statistically significant difference.

Results

VEGF-C expression is inhibited by SAHA in breast cancer cell lines

As shown in Fig. 1A, we found that all the cell lines, except SKBR3, investigated in this study expressed higher levels of VEGF-C than the normal breast epithelial cell line M10. SAHA dose-dependently downregulated the expression of VEGF-C in MDA-MB-231, MCF-7, MDA-MB-453 and BT-474 cells (Fig. 1B).

Reduction of VEGF-C production by SAHA

We next investigated whether production of the VEGF-C protein was indeed inhibited. Since MDA-MB-231 cells expressed the highest amount of VEGF-C, we used this cell line as a model in the subsequent experiments. Two approaches were performed. First, cellular VEGF-C protein level was determined by western blotting. As shown in Fig. 2A, three isoforms of VEGF-C with a molecular weight of 58, 34 and 21 kDa were reduced by SAHA in a dose-dependent manner. We also found that histone H3 acetylation was significantly increased indicating SAHA at these concentrations indeed exerted potent inhibitory effects on HDACs. In addition, VEGF-C released into the conditioned medium detected by ELISA assay was also reduced accordingly. Treatment of 10 μM SAHA reduced VEGF-C concentration by 50% (Fig. 2B).

SAHA represses VEGF-C promoter activity via the −185/+38 of the promoter region

Since SAHA reduces VEGF-C mRNA levels, this drug might directly inhibit VEGF-C transcription. We cloned human VEGF-C promoter region between −1046/+38 bp region from translational start site and generated different deletion promoter-luciferase constructs (Fig. 3A). These constructs were transfected into MDA-MB-231 and the effect of SAHA was examined. Our data demonstrated that SAHA at the concentration of 10 μM inhibited the full-length (−1046/+38) promoter by 30–40% (Fig. 3B). Deletion of promoter to −185 region did not affect SAHA-induced inhibition indicating the responsive elements were located between −185/+38 region.

SAHA reduces Sp1-mediated VEGF-C expression

Bioinformatics search revealed several potential transcription factor binding sites including Sp1, AP-2 and NF-κB within this region. Notably, we found that SAHA caused reduction of Sp1 but not Sp3 and NF-κB protein levels in MDA-MB-231 cells (Fig. 4A). Our data demonstrated that the Sp1 protein level was significantly decreased at 12 h after SAHA addition. However, no significant change at the mRNA level of Sp1, Sp3 and NF-κB was found until a 24-h treatment suggesting SAHA inhibited Sp1 via a post-translational regulation (Fig. 4B). To confirm the importance of Sp1, we treated cells with an Sp1 inhibitor mithramycin A and found that this inhibitor reduced Sp1 protein and attenuated VEGF-C mRNA expression dose-dependently (Fig. 5A). The amount of VEGF-C in the conditioned medium was also significantly reduced (Fig. 5B).

Enforced expression of Sp1 partially reverses the inhibition of VEGF-C by SAHA

The aforementioned data suggest that SAHA inhibited VEGF-C expression partly via Sp1. Consistent with this hypothesis, enforced expression of Sp1 in MDA-MB-231 cells partly reversed the inhibition of VEGF-C by SAHA (Fig. 6A). The VEGF-C released into the medium was also restored by Sp1 overexpression (Fig. 6B, lanes 2 and 4). Ectopic expression of Sp1 alone increased VEGF-C mRNA expression and the amount of VEGF-C in the conditioned medium. However, this upregulation was still attenuated by SAHA as this drug caused significant reduction of endogenous Sp1 protein as shown in Fig. 6A. The effect of Sp1 is specific since expression of KLF10, another member of the Kruppel-like/Sp1 gene family, could not antagonize the inhibition of VEGF-C by SAHA (Fig. 6C). Collectively, our results suggest that SAHA induced downregulation of Sp1 protein via a post-translational mechanism which led to reduction of Sp1-driven VEGF-C expression in breast cancer cells.

Discussion

Pathological studies have demonstrated that expression of VEGF-C and its receptor VEGFR3 is associated with angiogenesis, lymphangiogenesis and lymph node metastasis in breast cancer (22,23). By using an orthotopic transplantation model, Skobe et al clearly showed that induction of lymphangiogenesis by VEGF-C promotes breast cancer metastasis (24). Therefore, targeting VEGF-C/VEGFR3 signaling axis is important for breast cancer therapy. Since VEGFR3 is a tyrosine kinase, tyrosine kinase inhibitors are potential candidates to inhibit VEGFR3 activity. Sorafenib was initially identified as a potent inhibitor of c-RAF. However, it also inhibits VEGFR2 and VEGFR3 at concentrations of approximately 6–10 nM (25). This drug is now used in the clinic for the treatment of solid tumors. Other kinase inhibitors including sunitinib, AMG706, axitinib, and AZD2171 have also shown inhibitory activity against VEGFR3 and are now under different phases of clinical trials (26). Another strategy to suppress VEGFR3 activity is by blocking antibodies. A recombinant bispecific antibody has been developed to neutralize the biological activities of VEGFR2 and VEGFR3 (27). Roberts et al also demonstrated that inhibition of VEGFR3 activation with the antagonistic antibody potently suppresses lymph node and distant metastasis in an orthotopic spontaneous breast cancer model (28). An antibody that inhibits homodimerization of VEGFR3 and its heterodimerization with VEGFR2 has recently been shown to suppress both angiogenesis and lymphangiogenesis in vivo(29).

The targeting of VEGF-C has not received significant attention in the past decade. By using antibody phage-display, Rinderknecht et al identified a VEGF-C-blocking antibody which could effectively inhibit the interaction between VEGF-C and VEGFR3 and suppress its downstream signaling (30). The underlying mechanism mediating the upregulation of VEGF-C in cancer cells is largely unclear. A pioneer study of the genomic organization of human and mouse VEGF-C genes revealed that the upstream sequences contain conserved putative binding sites for Sp1, AP-2, and NF-κB transcription factors but not TATA box (31). In the present study, we demonstrated for the first time that a clinically used histone deacetylase inhibitor SAHA was able to inhibit Sp1-mediated expression of VEGF-C in human breast cancer cells and reduced VEGF-C concentration in the conditioned medium. Results of this study suggest that multiple transcription factors may be simultaneously involved in the regulation of VEGF-C. In addition, we demonstrated that the Sp1 specific inhibitor mithramycin A also attenuated VEGF-C expression suggesting a critical role of Sp1 in VEGF-C transcription. Our results provide a new strategy to suppress the expression of a lymphangiogenic factor in cancer cells by using HDAC inhibitors. Reduction of the production of VEGF-C will attenuate lyphangiogenesis and lymphatic metastasis in vivo. Since SAHA has already been approved for cancer treatment in the clinic, it will be of benefit to test whether this drug, in addition to its cytotoxic effect, also shows anti-lymphangiogenic activity in animals and patients.

Sp1 is a ubiquitous transcription factor expressed in mammalian cells and was initially recognized as a constitutive activator of housekeeping genes. However, recent studies demonstrated that Sp1 is involved in the control of tissue-specific and inducible genes and these target genes play critical roles in proliferation, differentiation, apoptosis and oncogenesis (32). In addition, increased Sp1 protein was found in various types of human cancer suggesting a possible oncogenic role (33,34). Our finding that SAHA causes downregulation of Sp1 protein is noteworthy. Although the underlying mechanism remains unclear, two potential mechanisms may be involved. First, a previous study identified lysine-703 as a major acetylation site of Sp1. Inhibition of HDAC activity by SAHA may directly affect the acetylation status of Sp1 and subsequently change protein stability (35). Second, heat shock protein 90 (hsp90) is an important regulator of Sp1 stability (36). SAHA has been demonstrated to inhibit the HDAC6-hsp90 chaperone complex to induce the degradation of client proteins such as mutant p53 (37). Therefore, it seems possible that SAHA may inhibit hsp90 and then affect Sp1 degradation.

In the present study, we demonstrated that SAHA is a potent inhibitor of VEGF-C expression in breast cancer cells and we showed that the inhibition is mediated by repression of Sp1. Our results also suggest that SAHA may exert anti-lymphangiogenic activity in cancer treatment.

Acknowledgements

This study was supported by grant CA-101-PP-39 from the National Health Research Institutes and DOH 101-TD-C-111-002 and DOH 101-TD-C-111-004 from the Department of Health, Taiwan, R.O.C.

References

1 

Sarkies P and Sale JE: Cellular epigenetic stability and cancer. Trends Genet. 28:118–127. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Esteller M and Herman JG: Cancer as an epigenetic disease: DNA methylation and chromatin alterations in human tumours. J Pathol. 196:1–7. 2002. View Article : Google Scholar : PubMed/NCBI

3 

Kouzarides T: Chromatin modifications and their function. Cell. 128:693–705. 2007. View Article : Google Scholar : PubMed/NCBI

4 

Huang L and Pardee AB: Suberoylanilide hydroxamic acid as a potential therapeutic agent for human breast cancer treatment. Mol Med. 6:849–866. 2000.PubMed/NCBI

5 

Kim MS, Kwon HJ, Lee YM, et al: Histone deacetylases induce angiogenesis by negative regulation of tumor suppressor genes. Nat Med. 7:437–443. 2001. View Article : Google Scholar : PubMed/NCBI

6 

Butler LM, Agus DB, Scher HI, et al: Suberoylanilide hydroxamic acid, an inhibitor of histone deacetylase, suppresses the growth of prostate cancer cells in vitro and in vivo. Cancer Res. 60:5165–5170. 2000.PubMed/NCBI

7 

Marks P, Rifkind RA, Richon VM, Breslow R, Miller T and Kelly WK: Histone deacetylases and cancer: causes and therapies. Nat Rev Cancer. 1:194–202. 2001. View Article : Google Scholar : PubMed/NCBI

8 

Minucci S and Pelicci PG: Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer. 6:38–51. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Marks PA and Breslow R: Dimethyl sulfoxide to vorinostat: Development of this histone deacetylase inhibitor as an anticancer drug. Nat Biotechnol. 25:84–90. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Duvic M and Vu J: Vorinostat: a new oral histone deacetylase inhibitor approved for cutaneous T-cell lymphoma. Expert Opin Investig Drugs. 16:1111–1120. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Peart MJ, Tainton KM, Ruefli AA, et al: Novel mechanisms of apoptosis induced by histone deacetylase inhibitors. Cancer Res. 63:4460–4471. 2003.PubMed/NCBI

12 

Shao Y, Gao Z, Marks PA and Jiang X: Apoptotic and autophagic cell death induced by histone deacetylase inhibitors. Proc Natl Acad Sci USA. 101:18030–18035. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Gui CY, Ngo L, Xu WS, Richon VM and Marks PA: Histone deacetylase (HDAC) inhibitor activation of p21WAF1 involves changes in promoter-associated proteins, including HDAC1. Proc Natl Acad Sci USA. 101:1241–1246. 2004. View Article : Google Scholar : PubMed/NCBI

14 

Mitsiades CS, Mitsiades NS, McMullan CJ, et al: Transcriptional signature of histone deacetylase inhibition in multiple myeloma: Biological and clinical implications. Proc Natl Acad Sci USA. 101:540–545. 2004. View Article : Google Scholar : PubMed/NCBI

15 

Khan AN and Tomasi TB: Histone deacetylase regulation of immune gene expression in tumor cells. Immunol Res. 40:164–178. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Ogbomo H, Michaelis M, Kreuter J, Doerr HW and Cinatl J Jr: Histone deacetylase inhibitors suppress natural killer cell cytolytic activity. FEBS Lett. 581:1317–1322. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Deroanne CF, Bonjean K, Servotte S, et al: Histone deacetylases inhibitors as anti-angiogenic agents altering vascular endothelial growth factor signaling. Oncogene. 21:427–436. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Alitalo K, Tammela T and Petrova TV: Lymphangiogenesis in development and human disease. Nature. 438:946–953. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Mandriota SJ, Jussila L, Jeltsch M, et al: Vascular endothelial growth factor-C- mediated lymphangiogenesis promotes tumour metastasis. EMBO J. 20:672–682. 2001. View Article : Google Scholar : PubMed/NCBI

20 

Maula SM, Luukkaa M, Grenman R, Jackson D, Jalkanen S and Ristamaki R: Intratumoral lymphatics are essential for the metastatic spread and prognosis in squamous cell carcinomas of the head and neck region. Cancer Res. 63:1920–1926. 2003.

21 

Dadras SS, Paul T, Bertoncini J, et al: Tumor lymphangiogenesis: a novel prognostic indaicator for cutaneous melanoma metastasis and survival. Am J Pathol. 162:1951–1960. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Valtola R, Salven P, Heikkila P, et al: VEGFR-3 and its ligand VEGF-C are associated with angiogenesis in breast cancer. Am J Pathol. 154:1381–1390. 1999. View Article : Google Scholar : PubMed/NCBI

23 

Wang CA, Jedlicka P, Patrick AN, et al: Six1 induces lymphangiogenesis and metastasis via upregulation of VEGF-C in mouse models of breast cancer. J Clin Invest. 122:1895–1906. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Skobe M, Hawighorst T, Jackson DG, et al: Induction of tumor lymphangiogenesis by VEGF-C promotes breast cancer metastasis. Nat Med. 7:192–198. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Flaherty KT: Sorafenib in renal cell carcinoma. Clin Cancer Res. 13:S747–S752. 2007. View Article : Google Scholar

26 

Hanrahan EO and Heymach JV: Vascular endothelial growth factor receptor tyrosine kinase inhibitors Vandetanib (ZD6474) and AZD2171 in lung cancer. Clin Cancer Res. 13:S4617–S4622. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Jimenez X, Lu D, Brennan L, et al: A recombinant, fully human, bispecific antibody neutralizes the biological activities mediated by both vascular endothelial growth factor receptors 2 and 3. Mol Cancer Ther. 4:427–434. 2005.

28 

Roberts N, Kloos B, Cassella M, et al: Inhibition of VEGFR-3 activation with the antagonistic antibody more potently suppresses lymph node and distant metastases than inactivation of VEGFR-2. Cancer Res. 66:2650–2657. 2006. View Article : Google Scholar : PubMed/NCBI

29 

McDonald DM: New antibody to stop tumor angiogenesis and lymphatic spread by blocking receptor partnering. Cancer Cell. 18:541–543. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Rinderknecht M, Villa A, Ballmer-Hofer K, Neri D and Detmar M: Phage-derived fully human monoclonal antibody fragments to human vascular endothelial growth factor-C block its interaction with VEGF receptor-2 and 3. PLoS One. 5:e119412010. View Article : Google Scholar

31 

Chilov D, Kukk E, Taira S, et al: Genomic organization of human and mouse genes for vascular endothelial growth factor C. J Biol Chem. 272:25176–25183. 1997. View Article : Google Scholar : PubMed/NCBI

32 

Li L and Davie JR: The role of Sp1 and Sp3 in normal and cancer cell biology. Ann Anat. 192:275–283. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Wang L, Wei D, Huang S, et al: Transcription factor Sp1 expression is a significant predictor of survival in human gastric cancer. Clin Cancer Res. 9:6371–6380. 2003.PubMed/NCBI

34 

Safe S and Abdelrahim M: Sp transcription factor family and its role in cancer. Eur J Cancer. 41:2438–2448. 2005. View Article : Google Scholar : PubMed/NCBI

35 

Hung JJ, Wang YT and Chang WC: Sp1 deacetylation induced by phorbol ester recruits p300 to activate 12(S)-lipoxygenase gene transcription. Mol Cell Biol. 26:1770–1785. 2006. View Article : Google Scholar : PubMed/NCBI

36 

Wang SA, Chuang JY, Yeh SH, et al: Heat shock protein 90 is important for Sp1 stability during mitosis. J Mol Biol. 387:1106–1119. 2009. View Article : Google Scholar : PubMed/NCBI

37 

Li D, Marchenko ND and Moll UM: SAHA shows preferential cytotoxicity in mutant p53 cancer cells by destabilizing mutant p53 through inhibition of the HDAC6- Hsp90 chaperone axis. Cell Death Differ. 19:1268–1276. 2012.PubMed/NCBI

Related Articles

Journal Cover

March 2013
Volume 29 Issue 3

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
Cheng H and Cheng H: Inhibition of lymphangiogenic factor VEGF-C expression and production by the histone deacetylase inhibitor suberoylanilide hydroxamic acid in breast cancer cells. Oncol Rep 29: 1238-1244, 2013
APA
Cheng, H., & Cheng, H. (2013). Inhibition of lymphangiogenic factor VEGF-C expression and production by the histone deacetylase inhibitor suberoylanilide hydroxamic acid in breast cancer cells. Oncology Reports, 29, 1238-1244. https://doi.org/10.3892/or.2012.2188
MLA
Cheng, H., Hung, W."Inhibition of lymphangiogenic factor VEGF-C expression and production by the histone deacetylase inhibitor suberoylanilide hydroxamic acid in breast cancer cells". Oncology Reports 29.3 (2013): 1238-1244.
Chicago
Cheng, H., Hung, W."Inhibition of lymphangiogenic factor VEGF-C expression and production by the histone deacetylase inhibitor suberoylanilide hydroxamic acid in breast cancer cells". Oncology Reports 29, no. 3 (2013): 1238-1244. https://doi.org/10.3892/or.2012.2188