Lenalidomide induces apoptosis and inhibits angiogenesis via caspase‑3 and VEGF in hepatocellular carcinoma cells

  • Authors:
    • Zhen Qu
    • Chunping Jiang
    • Junhua Wu
    • Yitao Ding
  • View Affiliations

  • Published online on: October 5, 2016     https://doi.org/10.3892/mmr.2016.5797
  • Pages: 4781-4786
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Hepatocellular carcinoma (HCC) remains a disease with a high mortality rate, and the treatment for HCC remains limited. It is necessary to develop efficient and low toxicity drugs for the clinical treatment of HCC. Lenalidomide is a novel analogue of thalidomide and has anti‑inflammatory, immunomodulatory and anti‑angiogenic effects. The current study investigated the inhibitory effect against HCC cells of lenalidomide and thalidomide. The MTT assay was used to determine the cytotoxicity of lenalidomide and thalidomide, and morphological changes were observed by fluorescence microscopy. Caspase and VEGF protein expression were measured by ELISA analysis and western blotting. It was identified that treatment of cells with lenalidomide and thalidomide led to a dose‑dependent inhibition of cell proliferation, and the two drugs were able to induce cells apoptosis and inhibit VEGF expression in HCC cells. In addition, lenalidomide was identified to exhibit greater effects than thalidomide at the same concentration. In conclusion, the results indicated that lenalidomide induces apoptosis and inhibits angiogenesis in HCC cells via caspase‑3 and VEGF pathway, and these may provide a potential perspective for lenalidomide's application in clinical.

Introduction

There is a high incidence of hepatocellular carcinoma (HCC) in China, HCC accounts for more than 80% of cases of primary liver cancer (1,2). The majority of patients with either HCC or hepatic metastasis carcinoma are not eligible candidates for surgical resection (3). Transarterial chemoembolization and the orally available targeted drug sorafenib have been demonstrated to increase survival in selected candidates (4). It is necessary to investigate and develop efficient and low toxicity drugs for the clinical treatment of HCC.

The use of thalidomide was terminated due to its teratogenicity (5). Lenalidomide is a new analogue of thalidomide and has been demonstrated to be more potent than thalidomide in the stimulation of T-cells, interleukin (IL)-2, and interferon (IFN)-γ production (6,7). Unlike thalidomide, lenalidomide exhibits almost no sedative or constipation-causing properties, and induces only minimal neurotoxicity in the initial clinical application (8). Previous studies have demonstrated that the anti-inflammatory, immunomodulatory and anti-angiogenic of lenalidomide served important roles in its anticancer activity (9,10). Lenalidomide induces apoptosis of myeloma cells and exhibits an immunomodulatory effect on cytokine secretion, enhancing T cell proliferation and IL-2 and IFN-γ production in patients with multiple myeloma (MM), and it additionally increases lysis of autologous MM cells through cytotoxicity mediated by natural killer cells (11,12). However, it remains unclear whether it may be efficacious in solid tumors.

In the current study, SMMC-7721 hepatoma cells were treated with lenalidomide or thalidomide at different concentrations, and it was identified that lenalidomide significantly inhibits proliferation of SMMC-7721 hepatoma cells in vitro. The two drugs tested can promote cell apoptosis and inhibit the expression of vascular endothelial growth factor (VEGF). In addition, lenalidomide was identified to be more potent than thalidomide, with observations of cell morphology by microscopy confirming these results. It was suggested that lenalidomide may induce apoptosis through the pathway of caspase-3 activation.

Materials and methods

Cells and reagents

The human HCC cell line SMMC-7721 was purchased from the Soochow University Cell Banks (Suzhou, China). Lenalidomide (Natco Pharma Limited, Hyderabad, India) and thalidomide (Sigma-Aldrich; Merck Millipore, Darmstadt, Germany) was dissolved in dimethyl sulfoxide (DMSO; Sigma-Aldrich; Merck Millipore) to prepare 10 and 40 mM stock solutions. Cells were stained with Annexin V-Fluorescein Isothiocyanate (FITC) following the manufacturer's instructions (Annexin V-FITC Apoptosis kit; Beijing BLKW Biotechnology Co., Ltd., Beijing, China) and analyzed for apoptosis by FACS using CellQuest software version 7.0 (BD Bioscience, Franklin Lakes, NJ, USA). Flag-tagged caspase-3 was purchased Bio-Box Biotech (Beijing, China). VEGF enzyme-linked immunosorbent assay (ELISA) analysis was performed with a commercial VEGF ELISA kit (cat. no. EH010-56; BLKW Biotechnology, Co., Ltd.) following the manufacturer's protocol. The antibodies for caspase-3 (cat. no. 9664P) and VEGF (cat. no. 2478S) were obtained from Cell Signaling Technology, Inc. (Danvers, MA, USA). GAPDH antibody (cat. no. AB22131) was obtained from Bioworld Technology, Inc. (St. Louis Park, MN, USA) The Cell Counting Kit 8 (CCK-8) was purchased from Dojindo Molecular Technologies, Inc. (Kumamoto, Japan).

Cell culture

SMMC-7721 cells were cultured in Dulbecco's modified Eagle's medium (Gibco; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (10%; Gibco; Thermo Fisher Scientific, Inc.). The SMMC-7721 human HCC cell line was maintained at 37°C in a humidified atmosphere with 5% CO2.

Cell proliferation assay

CCK-8 assay was used to evaluate the relative cell viability. Briefly, cells were plated in 96-well plates at a density of 5,000 cells/well in the media. The cells were pretreated with compounds at 6.25, 12.5, 25, 50, 100 and 200 µg/ml in a final concentration of 0.25% DMSO in triplicate at 37°C in a humidfied incubator at 5% CO2 for 48 h. Subsequently, CCK-8 reagent (100 µl/ml medium) was applied and incubated with cells at 37°C, 5% CO2 for 1 h. Cells were then incubated for an additional 4 h and the optical density (OD) was measured at 450 nm using a VersaMax Microtiter Plate Reader (Molecular Devices, LLC, Sunnyvale, CA, USA). Relative cell viability was calculated with the following formula: Relative cell viability (%) = OD(treatment group)/OD(control group) × 100%. The experiment was performed in triplicate.

Apoptosis assay

Cells (5×105) were treated with 100 µg/ml or 200 µg/ml drugs (lenalidomide and thalidomide) for 48 h then washed once with Annexin-V wash buffer (BD Biosciences). Cells were incubated with Annexin-V binding protein (5 µl) and propidium iodide (PI; 10 µl) (BD Biosciences) for 10 min. Cells were diluted with 500 µl wash buffer and analyzed by a FACSCalibur flow cytometer using CellQuest software. Furthermore, cells (5×105) were treated with the indicated treatments for 48 h, the supernatant was removed, 1 ml 70% cold ethanol was added along the six hole plate edge, then cells were fixed for 15 min. The cells were then washed with cold phosphate-buffered saline and PI (0.5 ml) was added prior to observation under the microscope.

Caspase-3 assay

Cells (5×105) were treated with 100 µg/ml or 200 µg/ml of the drugs (lenalidomide and thalidomide) for 48 h. Lysates (50 µl/2×106 cells) were added, then the cells were reprecipitated, placed in an ice bath for 30 min, during which they were oscillated 3–4 times (10 sec each time), then the crude cytosol was obtained as the supernatant as a result of centrifugation at 6,140 × g for 20 min at 4°C. Cell lysates (50 µl) were then extracted and mixed with Ac-DEVD-pNA (Sigma-Aldrich; Merck Millipore), incubated for 4 h at 37°C, and then the OD was measured using a microplate reader. Caspase-3 activation was determined by the rate of OD induced and OD control. All experiments were performed in triplicates.

ELISA assay

Cells (5×105) were treated with lenalidomide and thalidomide at the indicated doses for 48 h. ELISA analysis was performed as previously described (13). VEGF ELISA was performed using 200 µl culture supernatant in duplicates using the Quantikine VEGF ELISA kit (BLKW Biotechnology, Co., Ltd.) according to the manufacturer's instructions. Briefly, 200 µl culture supernatants was added to the wells and they were incubated for 2 h at 37°C. The plate was washed with 400 µl wash buffer twice and 200 µl VEGF conjugate was added followed by incubation for 2 h. Subsequent to the addition of substrate and stop solution, the optical density was determined using a microplate reader (RT-21000; Rayto Life and Analytical Sciences Co., Ltd., Shenzhen, China) at 450 nm with wavelength correction of 570 nm.

Western blotting

To determine the level of VEGF proteins, lenalidomide and thalidomide-treated cell lysates were prepared as described. A total of 20 µg proteins was analyzed by western blot analysis. The PVDF membranes with the transferred proteins were incubated with primary antibodies (cleaved caspase-3, 1:500; VEGF, 1:1,000; GAPDH, 1:10,000) at 4°C overnight and horseradish peroxidase-conjugated secondary antibodies (1:10,000; cat. no. 7074P2; Cell Signaling Technology, Inc.) at room temperature for 2 h. The signal was developed by the enhanced chemiluminescence reagent (EMD Millipore, Billerica, MA, USA) and visualized by FluorChem FC2 Imaging System (Alpha Innotech, San Leandro, CA, USA).

Statistical analysis

Data are expressed as the mean ± standard deviation. Data analysis was performed using the SPSS software, version 18.0 (SPSS, Inc., Chicago, IL, USA). Difference between groups were assessed with Student's t-test. P<0.05 was considered to indicate a statistically significant difference.

Results

Lenalidomide inhibits SMMC-7721 cell proliferation

The anti-proliferative rate was detected by CCK-8, results are expressed as the mean ± standard deviation. Treatment of cells with lenalidomide and thalidomide in different concentrations for 48 h led to a dose-dependent induction of the inhibition of cell proliferation. The anti-proliferative effects of lenalidomide were identified to be more potent than that of thalidomide in the 25, 50, 100 and 200 µg/ml groups (P<0.01; Fig. 1A and Table I).

Table I.

Cell growth inhibition rates in each drug concentration group.

Table I.

Cell growth inhibition rates in each drug concentration group.

Inhibition rate (%)

Drug concentration (µg/ml)ThalidomideLenalidomide
6.25 1.57±0.291 4.55±0.852
12.5 3.01±0.447 11.05±0.859
25 7.89±0.349 17.35±2.366a
50 17.28±1.223 28.44±2.331a
100 26.03±1.897 35.51±0.383a
200 32.40±1.296 41.87±0.949a

a P<0.05, lenalidomide vs. thalidomide.

Lenalidomide promotes apoptosis in SMMC-7721 cells

Cells (5×105) were treated with lenalidomide and thalidomide at the indicated doses for 48 h. Typical apoptotic morphological alterations were observed using fluorescence microscopy, and included cell nucleus shrinkage, chromatin condensation and the appearance of apoptotic bodies when treated with different doses of the drugs tested (Fig. 2). Lenalidomide was observed to exhibit an increased effect of inducing cell apoptosis than thalidomide at the same concentration (Fig. 2). Caspase-3 activity of samples was detected using a microplate reader, and the OD was analyzed at 405 nm. Activity of caspase-3 was upregulated with increased lenalidomide and thalidomide concentrations. Caspase-3 activity in the lenalidomide groups was greater than in the thalidomide groups at the same concentrations, and this difference was significant (16.69±1.54 vs. 13.37±1.59; 24.31±2.24 vs. 20.75±1.75; P<0.01, P<0.05; Fig. 1B, Table II). Hepatoma SMMC-7721 cell apoptosis was examined using the Annexin-V staining-based FACS assay following lenalidomide and thalidomide treatment for 48 h. The rate of apoptosis is presented in Fig. 3. Lenalidomide has a higher rate of induced cell apoptosis than thalidomide of the same concentration (P<0.01; Fig. 3), and the effect was observed to be greater with the increase of the lenalidomide concentration (Fig. 4).

Table II.

Expression of activated caspase-3 in each drug concentration group.

Table II.

Expression of activated caspase-3 in each drug concentration group.

Drug100 µg/ml200 µg/ml
Thalidomide 13.37±1.95 20.75±1.75
Lenalidomide 16.90±1.54a 24.31±2.24a

a P<0.05, lenalidomide vs. thalidomide.

Lenalidomide inhibits VEGF expression in SMMC-7721 cells

The data were presented as the mean ± standard deviation. In the current study, it was observed that lenalidomide can significantly inhibit VEGF expression of SMMC-7721 cells in vitro (Fig. 4) and is more potent than thalidomide in the 100 µg/ml groups (0.3760±0.01813; 0.4985±0.01097; P<0.05). No significant differences were observed between the 200 µg/ml groups (0.2255±0.01921; 0.2460±0.01192; P>0.05; Fig. 1C, Table III).

Table III.

Expression of vascular endothelial growth factor in each drug concentration group.

Table III.

Expression of vascular endothelial growth factor in each drug concentration group.

Drug100 µg/ml200 µg/ml
Thalidomide 0.4985±0.01097 0.2460±0.01192
Lenalidomide 0.3760±0.01813a 0.2255±0.01921

a P<0.05, lenalidomide vs. thalidomide.

Discussion

Lenalidomide is a novel analogue of thalidomide and previous studies have demonstrated its anticancer effects (8,9,14). The results of the current study demonstrated that lenalidomide and thalidomide can significantly inhibit the proliferation of the human SMMC-7721 HCC cell line. These suggested that lenalidomide has anti-proliferative activity for HCC cell lines in vitro, and the same effect of lenalidomide inhibiting cell proliferation has been observed in multiple myeloma cell lines (15). In addition, in the present study typical apoptotic morphological alterations were identified by fluorescence microscopy, including cell nucleus shrinkage, chromatin condensation and the presence of apoptotic bodies with treatment with different doses of the drugs.

Caspase-3 is an intracellular protease activated early during apoptosis of cells and serves an important role in cell apoptosis. This protease activity can be measured spectrophotometrically by detection of the chromophore (p-nitroanilide) subsequent to cleavage from the labeled substrate (DEVD-pNA). In previous studies, Dmoszynska et al (11) reported that the mixture of lovastatin and thalidomidemay increases the rate of multiple myeloma cell apoptosis. Ezell et al (16) observed that low dose thalidomide treatment of human T leukemic cells exhibited rapid increases in caspase-3 activity, in addition, thalidomide and its immunomodulatory analogs trigger activation of caspase-8, enhancing MM cell sensitivity to Fas-induced apoptosis (17). The current study identified that activity of caspase-3 is upregulated with increases in lenalidomide and thalidomide concentration, and caspase-3 activity in lenalidomide groups is significantly higher than that of the thalidomide groups with the same concentrations (P<0.05; Figs. 1B and 4 and Table II). The present study indicated that lenalidomide inhibits proliferation of SMMC-7721 cells via the induction of cell apoptosis and suggested that caspase-3 may serve an important role in this process.

Angiogenesis serves an important role during tumor growth, invasion and metastasis (18), and VEGF is an endothelial-specific growth factor that stimulates endothelial function and angiogenesis (15). A previous study identified that lenalidomide possesses anti-angiogenic activity, and enhances T cell proliferation in MM patients (8). Tan et al (7) demonstrated that thalidomide can suppress VEGF and hypoxia-inducible factor 1α in a dose-dependent manner (P<0.05). In the present study, it was demonstrated that lenalidomide can significantly inhibit VEGF expression of SMMC-7721 cells in vitro, and is more potent than that of thalidomide in the 100 µg/ml groups (P<0.05; Figs. 1C and 4 and Table III). No significant difference was observed between the 200 µg/ml groups (P>0.05), These results indicated that the anti-angiogenesis activity of lenalidomide was induced through suppression of VEGF, and this may be a critical factor in the inhibition of cell proliferation.

Taken together, the results suggest that the inhibition of SMMC-7721 cell proliferation by lenalidomide in vitro is more potent than that of thalidomide and in addition, induction of apoptosis and inhibition of angiogenesis may be two potential mechanisms for its anti-HCC activity.

Acknowledgements

The present study was supported by the Science and Technology Projects (grant no. CY20119002) from the Changzhou Science and Technology Bureau of China. The authors would like to thank Dr Peng Jiang for the technical assistance and Professor Yong Jiang for the helpful discussion.

References

1 

El-Serag HB and Mason AC: Rising incidence of hepatocellular carcinoma in the United States. N Engl J Med. 340:745–750. 1999. View Article : Google Scholar : PubMed/NCBI

2 

Sangiovanni A, Del Ninno E, Fasani P, De Fazio C, Ronchi G, Romeo R, Morabito A, De Franchis R and Colombo M: Increased survival of cirrhotic patients with a hepatocellular carcinoma detected during surveillance. Gastroenterology. 126:1005–1014. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Merchant N, David CS and Cunningham SC: Early hepatocellular carcinoma: Transplantation versus resection: The case for liver resection. Int J Hepatol. 2011:1420852011. View Article : Google Scholar : PubMed/NCBI

4 

Burrel M, Reig M, Forner A, Barrufet M, de Lope CR, Tremosini S, Ayuso C, Llovet JM, Real MI and Bruix J: Survival of patients with hepatocellular carcinoma treated by transarterial chemoembolisation (TACE) using drug eluting beads. Implications for clinical practice and trial design. J Hepatol. 56:1330–1335. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Settles B, Stevenson A, Wilson K, Mack C, Ezell T, Davis MF and Taylor LD: Down-regulation of cell adhesion molecules LFA-1 and ICAM-1 after in vitro treatment with the anti-TNF-alpha agent thalidomide. Cell Mol Biol (Noisy-le-grand). 47:1105–1114. 2001.PubMed/NCBI

6 

Sun P, Zhang LM, Sun DJ and Dong LL: Inhibitory effect of thalidomide on growth of human hepatoma cell line SMMC 7721 cells. Zhonghua Zhong Liu Za Zhi. 31:582–586. 2009.(In Chinese). PubMed/NCBI

7 

Tan H, Chen H, Xu C, Ge Z, Gao Y, Fang J, Liu W and Xiao S: Role of vascular endothelial growth factor in angiodysplasia: An interventional study with thalidomide. J Gastroenterol Hepatol. 27:1094–1101. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Galustian C, Meyer B, Labarthe MC, Dredge K, Klaschka D, Henry J, Todryk S, Chen R, Muller G, Stirling D, et al: The anti-cancer agents lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory cells. Cancer Immunol Immunother. 58:1033–1045. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Agliano A, Martin-Padura I, Marighetti P, Gregato G, Calleri A, Prior C, Redrado M, Calvo A and Bertolini F: Therapeutic effect of lenalidomide in a novel xenograft mouse model of human blastic NK cell lymphoma/blastic plasmacytoid dendritic cell neoplasm. Clin Cancer Res. 17:6163–6173. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Park E, Levis WR, Greig N, Jung E and Schuller-Levis G: Effect of thalidomide on nitric oxide production in lipopolysaccharide-activated RAW 264.7 cells. J Drugs Dermatol. 9:330–333. 2010.PubMed/NCBI

11 

Dmoszynska A, Podhorecka M, Klimek P and Grzasko N: Lovastatin and thalidomide have a combined effect on the rate of multiple myeloma cell apoptosis in short term cell cultures. Eur J Clin Pharmacol. 62:325–329. 2006. View Article : Google Scholar : PubMed/NCBI

12 

Dmoszynska A, Podhorecka M, Manko J, Bojarska-Junak A, Rolinski J and Skomra D: The influence of thalidomide therapy on cytokine secretion, immunophenotype, BCL-2 expression and microvessel density in patients with resistant or relapsed multiple myeloma. Neoplasma. 52:175–181. 2005.PubMed/NCBI

13 

Vaithilingam V, Oberholzer J, Guillemin GJ and Tuch BE: Beneficial effects of desferrioxamine on encapsulated human islets-in vitro and in vivo study. Am J Transplant. 10:1961–1969. 2010. View Article : Google Scholar : PubMed/NCBI

14 

van de Donk NW, Wittebol S, Minnema MC and Lokhorst HM: Lenalidomide (Revlimid) combined with continuous oral cyclophosphamide (endoxan) and prednisone (REP) is effective in lenalidomide/dexamethasone-refractory myeloma. Br J Haematol. 148:335–337. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Rao KV: Lenalidomide in the treatment of multiple myeloma. Am J Health Syst Pharm. 64:1799–1807. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Ezell TN, Maloney N, Githua JW and Taylor LD: Exposure to the anti-TNF-alpha drug thalidomide induces apoptotic cell death in human T leukemic cells. Cell Mol Biol (Noisy-le-grand). 49:1117–1124. 2003.PubMed/NCBI

17 

Mitsiades N, Mitsiades CS, Poulaki V, Chauhan D, Richardson PG, Hideshima T, Munshi NC, Treon SP and Anderson KC: Apoptotic signaling induced by immunomodulatory thalidomide analogs in human multiple myeloma cells: Therapeutic implications. Blood. 99:4525–4530. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Folkman J: Tumor angiogenesis: Therapeutic implications. N Engl J Med. 285:1182–1186. 1971. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 14 Issue 5

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
Qu Z, Jiang C, Wu J and Ding Y: Lenalidomide induces apoptosis and inhibits angiogenesis via caspase‑3 and VEGF in hepatocellular carcinoma cells. Mol Med Rep 14: 4781-4786, 2016
APA
Qu, Z., Jiang, C., Wu, J., & Ding, Y. (2016). Lenalidomide induces apoptosis and inhibits angiogenesis via caspase‑3 and VEGF in hepatocellular carcinoma cells. Molecular Medicine Reports, 14, 4781-4786. https://doi.org/10.3892/mmr.2016.5797
MLA
Qu, Z., Jiang, C., Wu, J., Ding, Y."Lenalidomide induces apoptosis and inhibits angiogenesis via caspase‑3 and VEGF in hepatocellular carcinoma cells". Molecular Medicine Reports 14.5 (2016): 4781-4786.
Chicago
Qu, Z., Jiang, C., Wu, J., Ding, Y."Lenalidomide induces apoptosis and inhibits angiogenesis via caspase‑3 and VEGF in hepatocellular carcinoma cells". Molecular Medicine Reports 14, no. 5 (2016): 4781-4786. https://doi.org/10.3892/mmr.2016.5797