Livistona chinensis seeds inhibit hepatocellular carcinoma angiogenesis in vivo via suppression of the Notch pathway

  • Authors:
    • Wei Lin
    • Jinyan Zhao
    • Zhiyun Cao
    • Qunchuan Zhuang
    • Liangpu Zheng
    • Jianwei Zeng
    • Zhenfeng Hong
    • Jun Peng
  • View Affiliations

  • Published online on: February 24, 2014     https://doi.org/10.3892/or.2014.3051
  • Pages: 1723-1728
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Livistona chinensis seeds have been used for centuries to clinically treat various types of cancer. Our published data suggest that Livistona chinensis seeds are able to inhibit hepatocellular carcinoma (HCC) growth in vitro and in vivo via promotion of mitochondrial-dependent apoptosis. To further elucidate the molecular mechanisms of its antitumor activity, in the present study, we used an HCC xenograft mouse model to evaluate the effect of an ethanol extract of Livistona chinensis seeds (EELC) on tumor angiogenesis and on the activation of the Notch pathway. Intratumoral microvessel density (MVD) in HCC xenograft mouse tumors was evaluated via immunohistochemical (IHC) staining for CD31. The mRNA and protein expression of vascular endothelial growth factor A (VEGF-A), VEGFR-2, Notch, Dll4 and Jagged1 was evaluated using RT-PCR and IHC, respectively. We found that EELC profoundly reduced MVD in the HCC mouse tumors, demonstrating the in vivo inhibitory effect of EELC on tumor angiogenesis. In addition, EELC treatment reduced the expression of VEGF-A and VEGFR-2 in tumor tissues. Furthermore, EELC treatment inhibited the expression of Notch, Dll4 and Jagged1. Our findings suggest that Livistona chinensis seeds inhibit tumor angiogenesis through suppression of the Notch pathway.

Introduction

Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths worldwide (1). Although surgical treatment and non-surgical therapeutic modalities such as radiotherapy, chemotherapy and interventional therapy have been employed, the prognosis of HCC patients remains discouraging due to the high recurrence rate and frequent incidence of intrahepatic metastasis (2,3). Angiogenesis is essential for continuous tumor growth and provides an avenue for hematogenous metastasis (49). Tumor angiogenesis is strongly regulated by multiple intracellular signaling transduction cascades such as the Notch pathway (10,11). The activation of Notch signaling is initiated by the binding of transmembrane Notch proteins (Notch1, Notch2, Notch3 and Notch4) to their specific membrane-bound ligands (Jagged1, Jagged2 and Delta-likes Dll1, Dll3 and Dll4). Mounting evidence shows that perturbation of the Notch pathway often leads to tumorigenesis (1216), and the potential role of Notch signaling in the development of HCC designates Notch and its ligands as promising targets for HCC therapy (1720).

Natural products, including traditional Chinese medicines (TCMs), have been used clinically for thousands of years as important alternative remedies for a variety of diseases including cancer (2123). Therefore, interest in the use of natural products as therapeutic agents for cancer has recently increased. Livistona chinensis, belonging to the monocotyledonous Palmaceae family, is a medicinal herb widely distributed in Eastern Asia. The seeds of Livistona chinensis have long been used in China to clinically treat various types of cancer (2427). Our previous findings suggest that Livistona chinensis seeds may be effective in cancer treatment via promoting mitochondrial-dependent apoptosis in vivo and in vitro (28). To further elucidate the molecular mechanisms of its antitumor activity, in the present study, we used a HCC xenograft mouse model to evaluate the effect of an ethanol extract of Livistonae chinensis seeds (EELC) on tumor angiogenesis and on the activation of the Notch pathway.

Materials and methods

Materials and reagents

Dulbecco’s modified Eagle’s medium (DMEM), fetal bovine serum (FBS), penicillin-streptomycin, trypsin-EDTA and TRIzol reagent were purchased from Invitrogen (Carlsbad, CA, USA). SuperScript II reverse transcriptase was obtained from Promega (Madison, WI, USA). CD31, vascular endothelial growth factor A (VEGF-A), VEGFR-2, Notch, Dll4 and Jagged1 antibodies and secondary antibodies were obtained from Cell Signaling (Beverly, MA, USA). All other chemicals, unless otherwise stated, were obtained from Sigma-Aldrich (St. Louis, MO, USA).

Preparation of ethanol extract from Livistona chinensis seeds

Livistona chinensis (500 g) seeds were extracted with 5,000 ml of 85% ethanol using a refluxing method and were filtered. The resultant solution was concentrated to a relative density of 1.05, and the dried powder of EELC was obtained by spraying desiccation method using a spray dryer (Model B-290; Buchi, Switzerland). For animal experiments, the powder of EELC was dissolved in saline to a working concentration of 300 mg/ml.

Cell culture

Human HCC HepG2 cells were obtained from the Cell Bank of the Chinese Academy of Sciences (Shanghai, China). The cells were cultured in DMEM containing 10% (v/v) FBS, and 100 U/ml penicillin and 100 μg/ml streptomycin in a 37°C humidified incubator with 5% CO2. The cells were subcultured at 80–90% confluency.

Animals

Male BALB/c athymic (nude) mice (with an initial body weight of 20–22 g) were obtained from Shanghai SLAC Laboratory Animal Co., Ltd. (Shanghai, China) and housed under pathogen-free conditions with controlled temperature (22°C), humidity, and a 12 h light/dark cycle. Food and water were given ad libitum throughout the experiment. All animal treatments were strictly performed in accordance with international ethical guidelines and the National Institutes of Health Guide Concerning the Care and Use of Laboratory Animals. The experiments were approved by the Institutional Animal Care and Use Committee of Fujian University of Traditional Chinese Medicine.

In vivo nude mouse xenograft study

HepG2 (4×106) cells mixed with Matrigel (1:1) were subcutaneously injected into the right flank of mice to initiate tumor growth. After 7 days of xenograft implantation when tumor size reached ~3 mm in diameter, mice were randomized into two groups (n=10) and given an intragastric administration of 3 g/kg of EELC or saline daily, 5 days a week for 21 days. At the end of the experiment, the animals were anaesthetized with pelltobarbitalum natricum, and the tumor tissue was removed. A portion of each tumor was fixed in 10% buffered formalin, and the remaining tissue was snap-frozen in liquid nitrogen and stored at −80°C.

Immunohistochemical analysis

After being fixed with 10% formaldehyde for 12 h, tumor samples were processed conventionally for paraffin-embedded tumor slides. The slides were subjected to antigen retrieval and the endogenous peroxidase activity was quenched with hydrogen peroxide. After blocking non-specific proteins with normal serum in phosphate-buffered saline (PBS) (0.1% Tween-20), slides were incubated with rabbit polyclonal antibodies against CD31, VEGF-A, VEGFR-2, Notch, Dll4 and Jagged1 (all in a 1:100 dilution). After washing with PBS, slides were incubated with the biotinylated secondary antibody followed by conjugated horseradish peroxidase (HRP)-labeled streptavidin (Dako), and then washed with PBS. The slides were then incubated with diaminobenzidine (DAB) as the chromogen, followed by counterstaining with diluted Harris’ hematoxylin (both from Sigma). After staining, five high-power fields (x400) were randomly selected in each slide, and the average proportion of positive cells in each field was counted using the true color multi-functional cell image analysis management system (Image-Pro Plus; Media Cybernetics, Bethesda, MD, USA). To rule out any non-specific staining, PBS was used to replace the primary antibody as a negative control.

RNA extraction and RT-PCR analysis

Briefly, total RNA from tumor tissues was isolated with TRIzol reagent. Oligo(dT)-primed RNA (1 μg) was reverse-transcribed with SuperScript II reverse transcriptase (Promega) according to the manufacturer’s instructions. The obtained cDNA was used to determine the mRNA amount of VEGF-A, VEGFR-2, Notch, Dll4 and Jagged1 by PCR. GAPDH was used as an internal control. The sequences of the primers used for amplification of VEGF-A, VEGFR-2, Notch, Dll4, Jagged1 and GAPDH transcripts are as follows: VEGF forward, 5′-GCC TTG CCT TGC TGC TCT A-3′ and reverse, 5′-GAT GTC CAC CAG GGT CTC G-3′; VEGFR-2 forward, 5′-ACG CCG ATT ATG TGA GA-3′ and reverse, 5′-AGG CAG GAG TTG AGT ATG-3′; Notch forward, 5′-CCG TCA TCT CCG ACT TCA TC-3′ and reverse, 5′-GGA CTT GCC CAG GTC ATC TAC-3′; Dll4 forward, 5′-ACA GTG CCT GAA CCG A-3′ and reverse, 5′-GCC CAC AAA GCC ATA A-3′; Jagged1 forward, 5′-TCG CTG TAT CTG TCC ACC T-3′ and reverse, 5′-TCC TTT CCA CCC ATT TTT A; GAPDH forward, 5′-GT CAT CCA TGA CAA CTT TGG-3′ and reverse, 5′-GA GCT TGA CAA AGT GGT CGT-3′.

Statistical analysis

Data are presented as means ± SD for the indicated number of independently performed experiments, and the data were analyzed using the SPSS Package for Windows (version 11.5). Statistical analysis of the data was performed with the Student’s t-test. Differences with p<0.05 were considered to be statistically significant.

Results

EELC inhibits tumor angiogenesis in the HCC xenograft mouse tumors

To determine the effect of EELC on tumor angiogenesis, we performed immunohistochemical (IHC) staining for the endothelial cell-specific marker CD31 to examine the intratumoral microvessel density (MVD) of HCC xenograft mouse tumors following EELC treatment. As shown in Fig. 1, the percentage of CD31-positive cells in the control and EELC-treated mice was 36.3±5.21 and 20.41±3.87%, respectively (p<0.05), demonstrating the in vivo inhibitory effect of EELC on tumor angiogenesis.

EELC inhibits the expression of VEGF-A and VEGFR-2 in the HCC xenograft mouse tumors

We next determined the effect of EELC on the expression of VEGF-A and VEGFR-2 in the HCC mouse tumors. Data from RT-PCR indicated that EELC reduced the mRNA expression of VEGF-A and VEGFR-2 in tumors (Fig. 2A). Results of the IHC assay showed that protein expression patterns of VEGF-A and VEGFR-2 were similar to their respective mRNA levels. The percentage of VEGF-A- and VEGFR-2-positive cells in the control group was 47.14±6.14 and 39.63±5.11%, while that in the EELC-treated mice was 10.78±3.11 and 15.34±3.04% (Fig. 2B).

EELC suppresses the Notch pathway in the HCC xenograft mouse tumors

To further investigate the mechanism of anti-angiogenic activity of EELC, we evaluated its effect on the Notch pathway by examining the expression of several key mediators of this signaling. As shown in Fig. 3A, EELC treatment profoundly reduced the mRNA expression of Notch, Dll4 and Jagged1 in the tumor tissues. Consistently, their protein expression was also significantly inhibited following EELC treatment. The percentage of Notch-, Dll4- and Jagged1-positive cells in the control group was 28.86±5.35, 32.39±4.27 and 27.33±3.58%, whereas that in the EELC-treated mouse tumors was 15.73±4.42, 13.8±2.24 and 12.15±2.11% (Fig. 3B).

Discussion

Deregulated angiogenesis plays a crucial role in the development of various diseases including cancer (2931). Induction of angiogenesis is mediated by a variety of molecules released by tumor cells (32). Vascular endothelial growth factor A (VEGF-A) is one of the most effective biologic inducers of angiogenesis (3335). After secretion VEGF-A, primarily interacts with specific receptors (VEGFR-2) present on the surface of vascular endothelial cells, which in turn triggers a tyrosine kinase signaling cascade, inducing endothelial cell proliferation, migration, survival, sprouting and consequent tube formation (35,38). VEGF-A was found to be highly expressed in a wide variety of human cancers and is associated with cancer progression, invasion and metastasis and poor patient prognosis (37,38). Therefore, it is not surprising that VEGF is considered as an attractive strategic target for inhibiting tumor angiogenesis.

A role for Notch signaling is well documented in both hematological malignancies and in solid tumors. Notch has been demonstrated to act either as a tumor suppressor or as an oncogene in a context-dependent manner (39). In addition, several lines of evidence suggest that Notch also plays an important role in tumor angiogenesis. Tumor vasculature and tumor cells have also been shown to overexpress the Notch ligand Dll4. Genetic studies have shown that reduction in Dll4 results in hypersprouting phenotypes and disruption of the vasculature (40,41). Another Notch ligand Jagged1 expressed in tumor cells has been shown to stimulate Notch-dependent angiogenesis (42,43). These findings suggest that Notch-specific blockade may inhibit tumor growth by disrupting angiogenesis. Therefore, Notch has become a focus for the development of targeted therapeutics in cancer.

Similar to the majority of other medicinal herbs, Livistona chinensis is composed of numerous chemical components such as flavonoids, diterpenoids, alkaloids, steroides and polysaccharides. Herbal medicines including Livistona chinensis are thus considered to be multi-target agents that exert their therapeutic function in a more holistic manner. Our previous findings suggest that Livistona chinensis seeds may be effective in inhibiting HCC xenograft mouse tumor growth, and in promoting cancer cell apoptosis (28). In the present study, we evaluated the effect of EELC on tumor angiogenesis using a xenograft mouse model. Our current data revealed that EELC reduced tumor MVD in HCC mice. In addition, EELC treatment suppressed the expression of VEGF-A and VEGFR-2 in tumor tissues, which in turn resulted in the inhibition of tumor angiogenesis. Furthermore, EELC treatment inhibited the expression of Notch, Dll4 and Jagged1.

In conclusion, our findings suggest that inhibition of tumor angiogenesis via suppression of the Notch pathway may be one of the mechanisms by which Livistona chinensis seeds play an important role in the treatment of cancers.

Acknowledgements

This study was sponsored by the National Natural Science Foundation of China (81073097 and 81202790).

Abbreviations:

EELC

ethanol extract of Livistona chinensis seeds

MVD

microvessel density

VEGF

vascular endothelial growth factor

VEGFR

vascular endothelial growth factor receptor

TCM

traditional Chinese medicine

References

1 

Parkin DM, Bray F, Ferlay J and Pisani P: Global cancer statistics, 2002. CA Cancer J Clin. 55:74–108. 2005. View Article : Google Scholar

2 

Lang H, Sotiropoulos GC, Brokalaki EI, Schmitz KJ, Bertona C, Meyer G, Frilling A, Paul A, Malagó M and Broelsch CE: Survival and recurrence rates after resection for hepatocellular carcinoma in noncirrhotic livers. J Am Coll Surg. 205:27–36. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Gish RG and Baron A: Hepatocellular carcinoma (HCC): current and evolving therapies. IDrugs. 11:198–203. 2008.PubMed/NCBI

4 

Gordaliza M: Natural products as leads to anticancer drugs. Clin Transl Oncol. 9:767–776. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Jia L: Cancer complementary and alternative medicine research at the US National Cancer Institute. Chin J Integr Med. 18:325–332. 2012. View Article : Google Scholar

6 

Carmady B and Smith CA: Use of Chinese medicine by cancer patients: a review of surveys. Chin Med. 9:222011. View Article : Google Scholar

7 

Cheueng S and Tai J: In vitro studies of the dry fruit of Chinese fan palm Livistona chinensis. Oncol Rep. 5:1331–1336. 2005.

8 

Sartippour MR, Liu C and Shao ZM: Livistona extract inhibits angiogenesis and cancer growth. Oncol Rep. 6:1355–1357. 2001.

9 

Huang WC, Hsu RM, Chi LM, et al: Selective downregulation of EGF receptor and downstream MAPK pathway in human cancer cell lines by active components partially purified from the seeds of Livistona chinensis R. Brown. Cancer Lett. 248:137–146. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Wang H, Li A, Dong XP and Xu XY: Screening of anti-tumor parts from the seeds of Livistona chinensis and its anti-angiogenesis effect. Zhong Yao Cai. 31:718–722. 2008.(In Chinese).

11 

Lin W, Zhao J, Cao Z, Zhuang Q, Zheng L, Cai Q, Chen D, Wang L, Hong Z and Peng J: Livistona chinensis seed suppresses hepatocellular carcinoma growth through promotion of mitochondrial-dependent apoptosis. Oncol Rep. 29:1859–1866. 2013.

12 

Folkman J and Klagsbrun M: Angiogenic factors. Science. 235:442–447. 1987. View Article : Google Scholar

13 

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

14 

Schneider BP and Miller KD: Angiogenesis of breast cancer. J Clin Oncol. 23:1782–1790. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Uzzan B, Nicolas P, Cucherat M and Perret GY: Microvessel density as a prognostic factor in women with breast cancer: a systematic review of the literature and meta-analysis. Cancer Res. 64:2941–2955. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Weidner N: The importance of tumor angiogenesis: the evidence continues to grow. Am J Clin Pathol. 122:675–677. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Kut C, Mac Gabhann F and Popel AS: Where is VEGF in the body? A meta-analysis of VEGF distribution in cancer. Br J Cancer. 97:978–985. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Takeshita K, Satoh M, Ii M, Silver M, Limbourg FP, Mukai Y, Rikitake Y, Radtke F, Gridley T, Losordo DW and Liao JK: Critical role of endothelial Notch1 signaling in postnatal angiogenesis. Circ Res. 100:70–78. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Garcia A and Kandel JJ: Notch: a key regulator of tumor angiogenesis and metastasis. Histol Histopathol. 27:151–156. 2012.PubMed/NCBI

20 

Dufraine J, Funahashi Y and Kitajewski J: Notch signaling regulates tumor angiogenesis by diverse mechanisms. Oncogene. 27:5132–5137. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Ridgway J, Zhang G, Wu Y, Stawicki S, Liang WC, Chanthery Y, Kowalski J, Watts RJ, Callahan C, Kasman I, Singh M, Chien M, Tan C, Hongo JA, de Sauvage F, Plowman G and Yan M: Inhibition of Dll4 signalling inhibits tumour growth by deregulating angiogenesis. Nature. 444:1083–1087. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Miele L, Golde T and Osborne B: Notch signaling in cancer. Curr Mol Med. 6:905–918. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Yanagawa S, Lee JS, Kakimi K, Matsuda Y, Honjo T and Ishimoto A: Identification of Notch1 as a frequent target for provirus insertional mutagenesis in T-cell lymphomas induced by leukemogenic mutants of mouse mammary tumor virus. J Virol. 74:9786–9791. 2000.

24 

Ellisen LW, Bird J, West DC, Soreng AL, Reynolds TC, Smith SD and Sklar J: TAN-1, the human homolog of the Drosophila Notch gene, is broken by chromosomal translocations in T lymphoblastic neoplasms. Cell. 66:649–661. 1991. View Article : Google Scholar

25 

Sjölund J, Manetopoulos C, Stockhausen MT and Axelson H: The Notch pathway in cancer: differentiation gone awry. Eur J Cancer. 41:2620–2629. 2005.PubMed/NCBI

26 

Nijjar SS, Crosby HA, Wallace L, Hubscher SG and Strain AJ: Notch receptor expression in adult human liver: a possible role in bile duct formation and hepatic neovascularization. Hepatology. 34:1184–1192. 2001. View Article : Google Scholar

27 

Nijjar SS, Wallace L, Crosby HA, Hubscher SG and Strain AJ: Altered Notch ligand expression in human liver disease: further evidence for a role of the Notch signaling pathway in hepatic neovascularization and biliary ductular defects. Am J Pathol. 160:1695–1703. 2002. View Article : Google Scholar

28 

Dorrell MI, Aguilar E, Scheppke L, Barnett FH and Friedlander M: Combination angiostatic therapy completely inhibits ocular and tumor angiogenesis. Proc Natl Acad Sci USA. 104:967–972. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Folkman J and Shing Y: Angiogenesis. J Biol Chem. 267:10931–10934. 1992.

30 

Folkman J: Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. 1:27–31. 1995. View Article : Google Scholar : PubMed/NCBI

31 

Folkman J: Angiogenesis. Annu Rev Med. 57:1–18. 2006. View Article : Google Scholar

32 

Weidner N, Semple JP, Welch WR and Folkman J: Tumor angiogenesis and metastasis - correlation in invasive breast carcinoma. N Engl J Med. 324:1–8. 1991. View Article : Google Scholar : PubMed/NCBI

33 

Risau W: Mechanisms of angiogenesis. Nature. 386:671–674. 1997. View Article : Google Scholar : PubMed/NCBI

34 

Jain RK: Tumor angiogenesis and accessibility: role of vascular endothelial growth factor. Semin Oncol. 29(Suppl 16): 3–9. 2002. View Article : Google Scholar : PubMed/NCBI

35 

Ferrara N: Role of vascular endothelial growth factor in physiologic and pathologic angiogenesis: therapeutic implications. Semin Oncol. 29(Suppl 16): 10–14. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Maeda K, Chung YS, Ogawa Y, Takatsuka S, Kang SM, Ogawa M, Sawada T and Sowa M: Prognostic value of vascular endothelial growth factor expression in gastric carcinoma. Cancer. 77:858–863. 1996. View Article : Google Scholar : PubMed/NCBI

37 

Kaya M, Wada T, Akatsuka T, Kawaguchi S, Nagoya S, Shindoh M, Higashino F, Mezawa F, Okada F and Ishii S: Vascular endothelial growth factor expression in untreated osteosarcoma is predictive of pulmonary metastasis and poor prognosis. Clin Cancer Res. 6:572–577. 2000.PubMed/NCBI

38 

Ferrara N, Gerber HP and LeCouter J: The biology of VEGF and its receptors. Nat Med. 9:669–676. 2003. View Article : Google Scholar : PubMed/NCBI

39 

Purow B: Notch inhibition as a promising new approach to cancer therapy. Adv Exp Med Biol. 727:305–319. 2012. View Article : Google Scholar : PubMed/NCBI

40 

Gale NW, Dominguez MG, Noguera I, Pan L, Hughes V, Valenzuela DM, Murphy AJ, Adams NC, Lin HC, Holash J, Thurston G and Yancopoulos GD: Haploinsufficiency of delta-like 4 ligand results in embryonic lethality due to major defects in arterial and vascular development. Proc Natl Acad Sci USA. 101:15949–15954. 2004. View Article : Google Scholar : PubMed/NCBI

41 

Duarte A, Hirashima M, Benedito R, Trindade A, Diniz P, Bekman E, Costa L, Henrique D and Rossant J: Dosage-sensitive requirement for mouse Dll4 in artery development. Genes Dev. 18:2474–2478. 2004. View Article : Google Scholar : PubMed/NCBI

42 

Skrtic A, Sokolic L, Borovecki A, Rosa J and Fenzl V: Immunohistochemical localization of CD31, NOTCH1 and JAGGED1 proteins in experimentally induced polycystic ovaries of immature rats. Acta Histochem. 113:262–269. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Benedito R, Roca C, Sörensen I, Adams S, Gossler A, Fruttiger M and Adams RH: The notch ligands Dll4 and Jagged1 have opposing effects on angiogenesis. Cell. 137:1124–1135. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

2014-April
Volume 31 Issue 4

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
Lin W, Zhao J, Cao Z, Zhuang Q, Zheng L, Zeng J, Hong Z and Peng J: Livistona chinensis seeds inhibit hepatocellular carcinoma angiogenesis in vivo via suppression of the Notch pathway. Oncol Rep 31: 1723-1728, 2014
APA
Lin, W., Zhao, J., Cao, Z., Zhuang, Q., Zheng, L., Zeng, J. ... Peng, J. (2014). Livistona chinensis seeds inhibit hepatocellular carcinoma angiogenesis in vivo via suppression of the Notch pathway. Oncology Reports, 31, 1723-1728. https://doi.org/10.3892/or.2014.3051
MLA
Lin, W., Zhao, J., Cao, Z., Zhuang, Q., Zheng, L., Zeng, J., Hong, Z., Peng, J."Livistona chinensis seeds inhibit hepatocellular carcinoma angiogenesis in vivo via suppression of the Notch pathway". Oncology Reports 31.4 (2014): 1723-1728.
Chicago
Lin, W., Zhao, J., Cao, Z., Zhuang, Q., Zheng, L., Zeng, J., Hong, Z., Peng, J."Livistona chinensis seeds inhibit hepatocellular carcinoma angiogenesis in vivo via suppression of the Notch pathway". Oncology Reports 31, no. 4 (2014): 1723-1728. https://doi.org/10.3892/or.2014.3051