SH003 selectively induces p73‑dependent apoptosis in triple‑negative breast cancer cells

  • Authors:
    • Eun Kyoung Choi
    • Seung‑Mi Kim
    • Seung‑Woo Hong
    • Jai‑Hee Moon
    • Jae‑Sik Shin
    • Jeong Hee Kim
    • Ih‑Yeon Hwang
    • Soo‑A Jung
    • Dae‑Hee Lee
    • Eun Young Lee
    • Seul Lee
    • Hyunwoo Kim
    • Daejin Kim
    • Yeong Seok Kim
    • Youn Kyung Choi
    • Hyo In Kim
    • Hyeong Sim Choi
    • Sung‑Gook Cho
    • Jeong Eun Kim
    • Kyu Pyo Kim
    • Yong Sang Hong
    • Won Keun Lee
    • Jung Shin Lee
    • Tae Won Kim
    • Seong‑Gyu Ko
    • Dong‑Hoon Jin
  • View Affiliations

  • Published online on: September 6, 2016     https://doi.org/10.3892/mmr.2016.5722
  • Pages: 3955-3960
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Triple-negative breast cancer (TNBC) is a breast cancer subtype that has an aggressive phenotype, is highly metastatic, has limited treatment options and is associated with a poor prognosis. In addition, metastatic TNBC has no preferred standard chemotherapy due to resistance to anthracyclines and taxanes. The present study demonstrated that a herbal extract, SH003, reduced cell viability and induced apoptosis in TNBC without cell cytotoxicity. Cell viability was examined using trypan blue exclusion and colony formation assays, which revealed a decrease in the cell viability. Additionally, apoptosis was determined using flow cytometry and a sub‑G1 assay, which revealed an increase in the proportion of cells in the sub‑G1 phase. The present study investigated the anticancer effect of SH003 in the Hs578T, MDA‑MB‑231 and ZR‑751 TNBC cell lines, and in the MCF7 and T47D non‑TNBC cell lines. Western blot analysis revealed that the expression levels of poly‑ADP‑ribose polymerase (PARP) cleavage protein in cells treated with SH003 were increased dose‑dependent manner, indicating that SH003 induced apoptosis via a caspase‑dependent pathway. Pre‑treatment with the caspase inhibitor Z‑VAD reduced SH003‑induced apoptosis was examined using trypan blue exclusion. Moreover, SH003 treatment enhanced the p73 levels in MDA‑MB‑231 cells but not in MCF7 cells. Transfection of p73 small interfering RNA (siRNA) in MDA‑MB0231 cells revealed that the apoptotic cell death induced by SH003 was significantly impaired in comparison with scramble siRNA transfected MDA‑MB‑231 cells. This was examined using trypan blue exclusion and flow cytometry analysis (sub‑G1). In addition, SH003 and paclitaxel exhibited synergistic anticancer effects on TNBC cells. The results indicate that SH003 exerts its anticancer effect via p73 protein induction and exhibits synergistic anticancer effects when combined with paclitaxel.

Introduction

Triple-negative breast cancer (TNBC) is one of the most common types of malignant tumors in women worldwide and is now the third leading cause of cancer-related mortality (1). TNBC is characterized by the absence of estrogen receptor (ER), progesterone receptor (PR) and human epidermal growth factor receptor 2 (HER2) (2). Thus, TNBC patients do not benefit from commonly used anti-estrogen and -herceptin-based therapies (3). In addition, patients with TNBC have been reported to have a poorer survival rate, and recurrence and distant metastases occurs more frequently than in patients with other types of breast cancer (4). Recent studies have revealed that TNBC comprises a heterogeneous group of tumors encompassing several molecular subtypes, such as luminal A, luminal B, HER2-enriched, claudin-low and basal-like tumors (57). Approximately 62% of basal-like TNBC and 43% of non-basal-like TNBC exhibit mutations in the tumor suppressor p53 (MTp53). The majority of p53 mutations observed in the tumor are loss-of-function mutations; however, some patients have been shown to exhibit oncogenic gain-of-function mutations. Therefore, targeting p53 may be less effective in TNBC treatment (8). Thus, successful treatment of p53-mutant TNBCs remains challenging. Doxorubicin (DOXO) and cisplatin (CDDP) are common treatment options for TNBC. However, acquired resistance and toxicity against these drugs eventually occurs, preventing effective treatment (9). Thus, novel therapies that overcome drug resistance and toxicity in p53-mutant TNBC cases are urgently required.

p73 is a member of the p53 gene family and has been shown to regulate p53 target genes in p53-deficient cancer cells (10). In contrast to p53, p73 is rarely mutated or lost in cancer (11). p53 deficient cancers are partly resistant to chemotherapy; however, not completely chemo-resistant. As p53 deficient cancers have other types of p53 family members such as p73, they may be able to replace the function of p53 in response to DNA damage (12). Therefore, identifying anticancer agents that can activate p73 may provide a chemotherapeutic approach for treating drug-resistant p53-mutant cancers. Recently, nonhydrolyzable ether analog of RRR-α-tocopherol in combination with DOXO or CDDP has been reported to target p53-mediated genes in a p73-dependent manner, enhancing the apoptosis of TNBC cells (9).

Traditional herbal medicines have recently been revisited for cancer therapy as herbal extracts or mixtures based on traditional medicines have exhibited anticancer effects with no or fewer side effects compared with current anticancer therapeutics, including chemical compounds and targeted antibodies (13). Previous studies have demonstrated the anticancer effects of herbal extracts from Astragalus membranaceus (Am), Angelica gigas (Ag) and Trichosanthes kirilowii Maximowicz (Tk) in different cancer cell types, such as leukemia, hepatocellular carcinoma, colon cancer, non-small cell lung cancer and gastric cancer cells (1419). Furthermore, extracts from a mixture of Am and Ag have been shown to affect various diseases, including hematological diseases and endocrine disorders (2022).

The present study demonstrated that SH003 extracted from a herbal mixture (Am, Ag and Tk) exhibited anticancer effects on TNBC via activation of the p73 pathway. Thus, SH003 may be useful for the treatment of TNBC.

Materials and methods

Cell culture and reagents

Established Hs578T, MDA-MB-231, ZR-75-1, MCF7 and T47D human breast cancer cell lines were purchased from the American Type Culture Collection (Manassas, VA, USA). All cell lines were maintained in RPMI-1640 medium (Gibco, Thermo Fisher Scientific Inc., Waltham, MA, USA) supplemented with 10% fetal bovine serum (FBS; Gibco, Thermo Fisher Scientific Inc.), 100 units of penicillin and 100 μl/ml streptomycin. All cells were cultured in a 5% CO2 incubator at 37°C. SH003 was extracted from Am, Ag or Tk, which were provided by Dr S.G. Ko (College of Korean Medicine, University of Kyung Hee, Seoul, Korea) as previously described (23).

Cell viability and cell death analysis

Cells (2×105 cells per plate) were seeded in a 60-mm plate and treated with various concentrations of SH003 (50, 100 or 200 μg/ml) for 48 h. Cell viability and cell death were assessed using a trypan blue exclusion method. Cell pellet was harvested and resuspend in 1 ml of phosphate-buffered saline (PBS). A total of 10 μl 0.4% trypan blue was gently mixed with 10 μl cell suspension. The mixture was applied to a hemocytometer and the number of trypan blue stained and non-stained cells were counted under a light microscope. The percentage of viable cells was calculated.

Colony formation assay

Cells were seeded at a density of 3×102 cells per well in a 6-well plate and were treated with various concentrations of SH003 (50, 100 or 200 μg/ml) for 24 h. The cells were cultured for 14 days and colonies were fixed with 4% paraformaldehyde and stained with a 0.01% crystal violet. Colony counts were performed manually using a light microscope and images of each plate were obtained.

RNA interference

Cells were transiently transfected with small interfering (si)RNA using the Lipofectamine RNAi MAX reagent (Invitrogen, Thermo Fisher Scientific, Inc., Waltham, MA, USA) according to the manufacturer's protocol. The siRNA sequence for transfection was p73-siRNA, 5′-GCAAUAAUCUCUCGCAGUAUU-3′ and scramble-siRNA, 5′-GGACUCUCGGAUUGUAAGAUU-3′

Western blot analysis

Cell lysates were prepared with radioimmunoprecipitation assay (RIPA) lysis buffer (50 mM Tris-HCl, pH 7.5; 50 mM NaCl, 1 μM EGTA and 1% Triton X-100) containing a protease inhibitor cocktail. Protein concentrations in extracts were determined using a Bradford assay (Bio Rad Laboratories, Inc., Hercules, CA, USA). Total cellular proteins (20 μg) were subjected to 10–15% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred to polyvinylidene difluoride membranes. The membranes were blocked with 5% non-fat dry milk in Tris-buffered saline with Tween-20 (TBST) buffer (20 mM Tris-HCl, pH 7.4, 150 mM NaCl, 0.1% Tween-20) and probed with anti-poly ADP ribose polymerase (PARP; cat. no. 9542; 1:1,000), anti-p73 (cat. no. 14620; 1:1,000), anti-caspase 3 (cat. no. 9661; 1:1000; Cell Signaling Technology, Beverly, MA, USA) or anti-β-actin (cat. no. sc-47778; 1:2,000; Santa Cruz Biotechnology Inc., Dallas, TX, USA) primary antibodies at 4°C overnight. Subsequently, the membranes were washed three times with TBST. Primary antibodies were detected following 2 h incubation at room temperature with a horseradish peroxidase-conjugated anti-mouse (cat. no. 7076; 1:2,000) or anti-rabbit secondary antibody (cat. no. 7074; 1:2,000; Cell Signaling Technology, Danvers, MA, USA). Blots were developed with an enhanced chemiluminescence detection kit (Amersham, Buckinghamshire, UK).

Cell cycle analysis

In total, 3×102 cells harvested by trypsinization were fixed in 1 ml of cold 70% ethanol for 24 h at −20°C. After washing cell pellets with 1 ml PBS, pellets were centrifuged at 300 × g for 5 min, discarded supernatant, resuspended in 1 ml staining solution (50 μg/ml propidium iodide, 50 μg/ml RNase and 0.1% Triton X 100 in citrate buffer, pH 7.8), incubated for 30 min and washed with PBS. Cell cycle distribution was analyzed using a FACSCalibur fluorescence-activated cell sorter and CellQuest version 3.0 software (BD Biosciences, San Jose, CA, USA).

Statistical analysis

SPSS version 22.0 (SPSS, Inc., Chicago, IL, USA) was used to perform statistical analysis. Data are presented as the mean ± standard deviation and multiple comparisons were conducted using one-way analysis of variance followed by Newman-Keuls post-hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

SH003 selectively inhibits the cell viability of TNBC cells

The viability of two breast cancer cell types, TNBC (Hs578T, MDA-MB-231 and ZR-75-1) and non-TNBC (MCF7 and T47D) was determined following exposure to SH003. Cells were treated with various concentrations (50, 100 and 200 μg/ml) of SH003. A significant decrease in cell viability was observed in TNBC cells (P<0.05 for the 100 μg/ml group and P<0.01 for the 200 μg/ml group compared with the untreated cells) but not in non-TNBC cells (Fig. 1A). MDA-MB-231 cells were further used as the cells showed the most effectively reduced cell viability in a dose-dependent manner. Additionally, colony formation analyses revealed a significant decrease in the number of MDA-MB-231 (TNBC) cells treated with SH003 but not MCF-7 (non-TNBC) cells (Fig. 1B; P<0.005 compared with the untreated cells). These results indicate that SH003 selectively decreases TNBC cell viability.

SH003 induces caspase-dependent cell death in TNBC cells

The effect of SH003 on cell death in MDA-MB-231 cells was determined using flow cytometry. A significant increase in the number of cells at the sub-G1 phase was observed following SH003 treatment (50, 100 and 200 μg/ml) (Fig. 2A) (P<0.05 and P<0.01 compared with the untreated cells). Apoptotic cell death and PARP cleavage in response to SH003 treatment were assessed in MDA-MB-231 cells using western blot analysis. The expression levels of cleaved PARP increased significantly in a dose-dependent manner compared with the untreated cells (P<0.05; Fig. 2B). SH003-induced apoptosis after pre-treatment with a pan-caspase inhibitor, Z-VAD was then examined. Pre-treatment with Z-VAD partially decreased the MDA-MB-231 cell death and levels of cleaved PARP induced by SH003 (Fig. 2C; P<0.05 compared with cells treated with SH003 only). Thus, SH003-induced cell death is partially caspase-dependent in TNBC cells.

Induction of p73 expression by SH003 leads to apoptosis in TNBC

A previous study indicated that p73 expression may prevent drug resistance and toxicity in p53-mutant TNBC (8). It was demonstrated that SH003 induced p73-mediated apoptosis in p53 mutant MDA-MB-231 cells. p73 expression in MDA-MB-231 cells was observed following treatment with SH003 using western blot analysis. The p73 protein levels in MDA-MB-231 cells treated with SH003 increased in a dose-dependent manner (Fig. 3A). To confirm that MDA-MB-231 cell death induced by SH003 was correlated with p73, the effect of knockdown of endogenous p73 using small interfering RNAs in MDA-MB-231 cells was examined. Cells were transfected with scrambled siRNA or p73 siRNA, followed by treatment with SH003. Transfected p73 siRNA decreased cell death and PARP cleavage compared with scrambled siRNA treatment (Fig. 3B; P<0.05 compared with SH003 single-treated scramble cells). Additionally, cell death was confirmed using flow cytometric analysis. The number of cells in the sub-G1 phase following SH003 treatment was decreased in the p73 siRNA-transfected cell line compared with the scrambled siRNA-transfected cell line (Fig. 3C; P<0.05 compared with SH003 single-treated scramble cells). These results indicated that the induction of p73 expression by SH003 leads to the apoptosis of MDA-MB-231 cells.

SH003 sensitizes paclitaxel-induced MDA-MB-231 cell death

Paclitaxel (taxane) is a commonly used treatment in conjunction with other anticancer agents for TNBC; however, paclitaxel treatment occasionally fails due to drug resistance. It was demonstrated that SH003 in combination with paclitaxel synergistically increases cell death in TNBC cell compared with individual treatments (Fig. 4A; P<0.05 compared with cells treated with SH003 or paclitaxel only). Western blot analyses indicated that SH003 in combination with paclitaxel increased the levels of cleaved caspase-3 in MDA-MB-231 cells but did not alter p73 expression (Fig. 4B). These results indicate that SH003 in combination with paclitaxel synergistically enhances apoptosis in TNBC cells.

Discussion

TNBC accounts for 10–20% of all types of breast cancer (24). TNBC is an aggressive histological subtype with limited treatment conditions and poor prognosis following standard chemotherapy. The anticancer effects of commonly used chemotherapeutic agents, such as paclitaxel, doxorubicin and cisplatin are limited to cure patients with TNBC due to acquired drug resistance and toxicity (9). The present study focused on anticancer therapy for TNBC to overcome resistance against conventional therapies.

The present chemotherapeutic agents for TNBC are DNA-damaging agents (25). In the DNA-damage pathway, tumor suppressor p53 is important in anticancer actions of DNA-damaging agents (26). A recent study reported that regulation of p53-mediated apoptotic signaling occurs in a p73-dependent manner, which results in enhanced apoptosis in p53-deficient TNBC (9). The functional and structural similarities of p53 and p73 have been previously reported (10). It is also known that p73 can replace the function of p53 in response to DNA damage in p53-deficient cancers. p73 is not frequently mutated in cancers and regulates p53 target genes, such as Bax and Noxa in p53-deficient cancers (27,28). The key role of p73 in anti-cancer effects for p53-deficient TNBC was identified.

Identification and development of traditional herbal medicines has increased due to their potential anticancer effects and minimal side effects. This study demonstrated that SH003 inhibited TNBC growth in a dose-dependent manner. Treatment with SH003 resulted in apoptotic cell death as shown by increased PARP cleavage, a caspase-dependent apoptotic marker. In addition, SH003-induced apoptosis was validated after pretreatment with the pan-caspase inhibitor, Z-VAD, as this partially decreased cell death in MDA-MB-231 cells.

Notably, apoptotic cell death induced by SH003 was associated with induction of p73 expression in TNBC. The anticancer effect of SH003 was validated upon siRNA-mediated knockdown of p73. The results showed that knockdown of p73 decreased apoptotic cell death induced by SH003 treatment. In addition, single treatment with paclitaxel did not result in any specific cell death, while SH003 in combination with paclitaxel synergistically increased cell death in TNBC. Therefore, SH003 in combination chemotherapies may aid in overcoming resistance to conventional chemotherapies in TNBC.

The apoptotic cell death induced by SH003 is associated with p73 expression, which indicates that the anticancer effects of SH003 are induced by p73-dependent apoptosis. This study showed that SH003 induced the expression of p73- and caspase-dependent apoptosis. Thus, this study revealed that a traditional herbal medicine, SH003, has a significant anticancer effect via p73-mediated apoptosis in TNBC cells and confirmed p73 as a promising therapeutic target for TNBC.

Abbreviations:

Am

Astragalus membranaceus

Ag

Angelica gigas

Tk

Trichosanthes kirilowii Maximowicz

TNBC

triple-negative breast cancer

Acknowledgments

This study was supported by grants from Basic Science Research Program through the the National Research Foundation of Korea (NRF) grant funded by the Korean government (MEST), Seoul, Republic of Korea (NRF-2013R1A2A2A01067394) to Professor Dong-Hoon Jin, and the Korean Medicine R&D Project of the Ministry of Health and Welfare to Professor Seong-Gyu Ko (B110043).

References

1 

Millis SZ, Gatalica Z, Winkler J, Vranic S, Kimbrough J, Reddy S and O'Shaughnessy JA: Predictive Biomarker Profiling of > 6000 Breast Cancer Patients Shows Heterogeneity in TNBC, With Treatment Implications. Clin Breast Cancer. 15:73–481. 2015. View Article : Google Scholar

2 

Isakoff SJ: Triple-negative breast cancer: Role of specific chemotherapy agents. Cancer J. 16:53–61. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Xu L, Yin S, Banerjee S, Sarkar F and Reddy KB: Enhanced anticancer effect of the combination of cisplatin and TRAIL in triple-negative breast tumor cells. Mol Cancer Ther. 10:550–557. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Haffty BG, Yang Q, Reiss M, Kearney T, Higgins SA, Weidhaas J, Harris L, Hait W and Toppmeyer D: Locoregional relapse and distant metastasis in conservatively managed triple negative early-stage breast cancer. J Clin Oncol. 24:5652–5657. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, Shyr Y and Pietenpol JA: Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. J Clin Invest. 121:2750–2767. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Cancer Genome Atlas Network: Comprehensive molecular portraits of human breast tumours. Nature. 490:61–70. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Prat A and Perou CM: Deconstructing the molecular portraits of breast cancer. Mol Oncol. 5:5–23. 2011. View Article : Google Scholar

8 

Aas T, Børresen AL, Geisler S, Smith-Sørensen B, Johnsen H, Varhaug JE, Akslen LA and Lønning PE: Specific P53 mutations are associated with de novo resistance to doxorubicin in breast cancer patients. Nat Med. 2:811–814. 1996. View Article : Google Scholar : PubMed/NCBI

9 

Tiwary R, Yu W, Sanders BG and Kline K: α-TEA cooperates with chemotherapeutic agents to induce apoptosis of p53 mutant, triple-negative human breast cancer cells via activating p73. Breast Cancer Res. 13:R12011. View Article : Google Scholar

10 

Levrero M, De Laurenzi V, Costanzo A, Gong J, Wang JY and Melino G: The p53/p63/p73 family of transcription factors: Overlapping and distinct functions. J Cell Sci. 113:1661–1670. 2000.PubMed/NCBI

11 

Kaelin WG Jr: The p53 gene family. Oncogene. 18:7701–7705. 1999. View Article : Google Scholar

12 

Rödicker F and Pützer BM: p73 is effective in p53-null pancreatic cancer cells resistant to wild-type TP53 gene replacement. Cancer Res. 63:2737–2741. 2003.PubMed/NCBI

13 

Maurya U and Srivastava S: Traditional Indian herbal medicine used as antipyretic, antiulcer, anti-diabetic and anticancer: A Review. IJRPC. 1:42011.

14 

Shin JW, Son JY, Kang JK, Han SH, Cho CK and Son CG: Trichosanthes kirilowii tuber extract induces G2/M phase arrest via inhibition of tubulin polymerization in HepG2 cells. J Ethnopharmacol. 115:209–216. 2008. View Article : Google Scholar

15 

Cho WC and Leung KN: In vitro and in vivo anti-tumor effects of Astragalus membranaceus. Cancer Lett. 252:43–54. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Cui R, He J, Wang B, Zhang F, Chen G, Yin S and Shen H: Suppressive effect of Astragalus membranaceus Bunge on chemical hepatocarcinogenesis in rats. Cancer Chemother Pharmacol. 51:75–80. 2003. View Article : Google Scholar

17 

Heo BG, Chon SU, Park YJ, Bae JH, Park SM, Park YS, Jang HG and Gorinstein S: Antiproliferative activity of Korean wild vegetables on different human tumor cell lines. Plant Foods Hum Nutr. 64:257–263. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Kim T, Choi HJ, Kim NJ and Kim DH: Anxiolytic-like effects of ginsenosides Rg3 and Rh2 from red ginseng in the elevated plus-maze model. Planta Med. 75:836–839. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Li LK, Kuang WJ, Huang YF, Xie HH, Chen G, Zhou QC, Wang BR and Wan LH: Anti-tumor effects of Astragalus on hepatocellular carcinoma in vivo. Indian J Pharmacol. 44:78–81. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Lv J, Zhao Z, Chen Y, Wang Q, Tao Y, Yang L, Fan TP and Liu C: The chinese herbal decoction danggui buxue tang inhibits angiogenesis in a rat model of liver fibrosis. Evid Based Complement Alternat Med. 2012:2849632012. View Article : Google Scholar : PubMed/NCBI

21 

Yang M, Chan GC, Deng R, Ng MH, Cheng SW, Lau CP, Ye JY, Wang L and Liu C: An herbal decoction of Radix astragali and Radix angelicae sinensis promotes hematopoiesis and thrombopoiesis. J Ethnopharmacol. 124:87–97. 2009. View Article : Google Scholar : PubMed/NCBI

22 

Zhang WL, Zheng KY, Zhu KY, Zhan JY, Bi CW, Chen JP, Du CY, Zhao KJ, Lau DT, Dong TT and Tsim KW: Chemical and biological assessment of Angelica herbal decoction: Comparison of different preparations during historical applications. Phytomedicine. 19:1042–1048. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Choi YK, Cho SG, Woo SM, Yun YJ, Park S, Shin YC and Ko SG: Herbal extract SH003 suppresses tumor growth and metastasis of MDA-MB-231 breast cancer cells by inhibiting STAT3-IL-6 signaling. Mediators Inflamm. 2014:4921732014. View Article : Google Scholar : PubMed/NCBI

24 

Schwentner L, Wolters R, Koretz K, Wischnewsky MB, Kreienberg R, Rottscholl R and Wöckel A: Triple-negative breast cancer: The impact of guideline-adherent adjuvant treatment on survival - a retrospective multi-centre cohort study. Breast Cancer Res Treat. 132:1073–1080. 2012. View Article : Google Scholar

25 

O'Reilly EA, Gubbins L, Sharma S, Tully R, Guang MH, Weiner-Gorzel K, McCaffrey J, Harrison M, Furlong F, Kell M and McCann A: The fate of chemoresistance in triple negative breast cancer (TNBC). BBA Clin. 3:257–275. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Essmann F and Schulze-Osthoff K: Translational approaches targeting the p53 pathway for anti-cancer therapy. Br J Pharmacol. 165:328–344. 2012. View Article : Google Scholar :

27 

Melino G, Bernassola F, Ranalli M, Yee K, Zong WX, Corazzari M, Knight RA, Green DR, Thompson C and Vousden KH: p73 induces apoptosis via PUMA transactivation and Bax mitochondrial translocation. J Biol Chem. 279:8076–8083. 2004. View Article : Google Scholar

28 

Flinterman M, Guelen L, Ezzati-Nik S, Killick R, Melino G, Tominaga K, Mymryk JS, Gäken J and Tavassoli M: E1A activates transcription of p73 and Noxa to induce apoptosis. J Biol Chem. 280:5945–5959. 2005. View Article : Google Scholar

Related Articles

Journal Cover

October-2016
Volume 14 Issue 4

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
Choi EK, Kim SM, Hong SW, Moon JH, Shin JS, Kim JH, Hwang IY, Jung SA, Lee DH, Lee EY, Lee EY, et al: SH003 selectively induces p73‑dependent apoptosis in triple‑negative breast cancer cells. Mol Med Rep 14: 3955-3960, 2016
APA
Choi, E.K., Kim, S., Hong, S., Moon, J., Shin, J., Kim, J.H. ... Jin, D. (2016). SH003 selectively induces p73‑dependent apoptosis in triple‑negative breast cancer cells. Molecular Medicine Reports, 14, 3955-3960. https://doi.org/10.3892/mmr.2016.5722
MLA
Choi, E. K., Kim, S., Hong, S., Moon, J., Shin, J., Kim, J. H., Hwang, I., Jung, S., Lee, D., Lee, E. Y., Lee, S., Kim, H., Kim, D., Kim, Y. S., Choi, Y. K., Kim, H. I., Choi, H. S., Cho, S., Kim, J. E., Kim, K. P., Hong, Y. S., Lee, W. K., Lee, J. S., Kim, T. W., Ko, S., Jin, D."SH003 selectively induces p73‑dependent apoptosis in triple‑negative breast cancer cells". Molecular Medicine Reports 14.4 (2016): 3955-3960.
Chicago
Choi, E. K., Kim, S., Hong, S., Moon, J., Shin, J., Kim, J. H., Hwang, I., Jung, S., Lee, D., Lee, E. Y., Lee, S., Kim, H., Kim, D., Kim, Y. S., Choi, Y. K., Kim, H. I., Choi, H. S., Cho, S., Kim, J. E., Kim, K. P., Hong, Y. S., Lee, W. K., Lee, J. S., Kim, T. W., Ko, S., Jin, D."SH003 selectively induces p73‑dependent apoptosis in triple‑negative breast cancer cells". Molecular Medicine Reports 14, no. 4 (2016): 3955-3960. https://doi.org/10.3892/mmr.2016.5722