Phosphoinositide 3-kinase/Akt pathway is involved in pingyangmycin‑induced growth inhibition, apoptosis and reduction of invasive potential in EOMA mouse hemangioendothelioma cells

  • Authors:
    • Li‑Xia Peng
    • Ping Zhao
    • Hong‑Sheng Zhao
    • Er Pan
    • Bin‑Bin Yang
    • Qin Li
  • View Affiliations

  • Published online on: October 15, 2015     https://doi.org/10.3892/mmr.2015.4447
  • Pages: 8275-8281
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Pingyangmycin (PYM), a glycopeptide antibiotic, has been recommended as a stand treatment for hemangioma. However, the underlying mechanisms of its anti‑tumor effects have remained elusive. The purpose of the present study was to explore the effects of PYM on the biological behavior of the EOMA mouse hemangioendothelioma cell line and investigate the possible mechanisms. The effects of PYM on EOMA cell viability were determined by an MTT assay, apoptosis was evaluated by Annexin V/propidium iodide staining and flow cytometric analysis, and cell invasion ability was determined using a Transwell invasion assay. In order to investigate the underlying mechanism of action of PYM, the expression of angiogenic signaling proteins was determined by western blot analysis. PYM treatment (0.5‑500 µg/ml) inhibited cell growth in a time- and dose‑dependent manner. PYM at 100 µg/ml significantly induced apoptosis and reduced the invasive ability of EOMA cells. Effects of PYM on cell viability, apoptosis and invasion ability were completely blocked by co‑treatment with phosphoinositide 3‑kinase (PI3K) activator insulin‑like growth factor‑1 (IGF‑1). Furthermore, treatment with PYM reduced the expression of PI3K and phosphorylated Akt. In conclusion, the present study indicated that the PI3K/Akt pathway is likely to be involved in the anti-cancer effects of PYM on EOMA cells.

Introduction

Pingyangmycin (PYM) was isolated from Treptomyces verticillus var. pingyangensis n.sp in 1969 and has been used for clinical treatment of various types of cancer since 1978 (15). The most successful clinical use of PYM is in the treatment of vascular malformations and hemangioma (612). Injection of PYM has been shown to have beneficial effects in >90% of infants with low-flow orbital or periorbital venous malformation (7) and to improve infantile hemangiomas in oral and maxillofacial regions in 100% of cases (11). Based on these results, PYM was recommended as a standard treatment for vascular malformations and hemangioma (13). However, to the best of our knowledge, the underlying molecular mechanism of action of PYM in treating hemangioma has remained elusive. The determination of its mechanism of action may aid in expanding the treatment spectrum of PYM to other cancer types.

As one of most common tumor types in infants, hemangioma have an incidence rate of 5–10% at the end of the first year of life worldwide (14). The etiology of hemangioma has remained elusive; however, angiogenesis (sprouting of new vessels from existing vessels) and vasculogenesis (de novo formation of new blood vessels from stem cells) were proposed as mechanisms of neovascularization in hemangioma (15). Numerous signaling pathways are implicated in the process of angiogenesis and vasculogenesis, including the human vascular endothelial growth factor/receptor pathway (16), hypoxia-inducible factor pathway (17), angiopoietin signaling (18) and the notch pathway (19). These pathways have provided therapeutic targets for the development of molecular targeted therapies.

Recent studies indicated that the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway was involved in the process of angiogenesis. PI3K/Akt/mTOR is one of the major signaling pathways which is crucial for cancer-cell survival, proliferation and angiogenesis. This pathway acts to promote cancer-cell survival by inhibiting pro-apoptotic factors and activating anti-apoptotic factors (20). mTOR is the key kinase downstream of the PI3K/Akt pathway. Studies have reported that mTOR inhibitor rapamycin has anti-angiogenic effects on endothelial cells (21,22). Considering that mTOR is the central downstream signaling molecule of the PI3K/Akt pathway, these results implied that de-regulation of the PI3K/Akt/mTOR pathway may be involved in the pathology of hemangioma. Thus, the present study posed the hypothesis that PYM may have inhibitory effect on hemangioma-derived endothelial cells by affecting the PI3K/Akt pathway, which may provide a mechanistic basis for the anti-cancer effects of PYM on hemangioma.

Originally, PYM was reported to exert its anti-cancer effects by DNA strand breaks (23). Recent studies have reported mechanisms including activation of the p53 pathway (24), downregulation of B-cell lymphoma 2 (Bcl-2) and upregulation of Bcl-2-associated X protein (Bax) (25) and induction of apoptosis by activation of caspase-3 (26) in vitro and in vivo, which may be downstream cellular responses to PYM treatment. The present study investigated the underlying mechanisms of the anti-cancer activity of PYM on hemangioma by examining its effects on cell viability, cell cycle, invasive potential and the expression of proteins involved in the angiogenesis-associated PI3K/Akt pathway using an MTT assay, flow cytometry, Transwell assay and western blot analysis. The discoveries of the present study may aid in expanding the treatment spectrum of PYM to other cancer types.

Materials and methods

Reagents

PYM was purchased from Harbin Laiboten Pharmaceutical (Harbin, China). It was dissolved in dimethyl-sulfoxide (DMSO) to obtain an 8 mg/ml stock solution, which was stored at 4°C. LY294002 was purchased from Promega Corp. (Madison, WI, USA) and insulin-like growth factor-1 (IGF-1) was purchased from Sino Biological (Beijing, China).

Cell culture

EOMA mouse hemangioendothelioma endothelial cells were purchased from the American Type Tissue Collection (Manassas, VA, USA) and were cultured in Dulbecco's modified Eagle's medium (DMEM; Hyclone, Logan, UT, USA) supplemented with 10% fetal bovine serum (Gen-View Scientific, Inc.,confirm El Monte, FL, USA), 100 IU/ml penicillin (Sigma-Aldrich, St. Louis, MO, USA) and 100 µg/ml streptomycin (Sigma-Aldrich) at 37°C, 5% CO2 and a humidified atmosphere.

Cell viability assay

The effect of PYM, LY294002 and IGF-1 on the proliferation of EOMA cells was determined using an MTT assay (Sigma-Aldrich). EOMA cells were seeded at a density of 1×103/well in 96-well plates and incubated at 37°C for 24 h. The cells were treated with various concentrations of PYM (0.5–100 µg/ml), LY294002 (0.5 nM), PYM (100 µg/ml) plus IGF-1 (100 ng/ml) or an equal volume of DMSO for the control and incubated for 24, 48 or 72 h. Subsequently, 20 µl MTT solution (5 mg/ml) was added to each well. After 4 h of incubation at 37°C, the supernatants were carefully removed and 150 µl DMSO was added. The optical density (OD) value was measured at 490 nm using a Multiscan MK3 microplate reader (Thermo Fisher Scientific, Waltham, MA, USA).

Determination of apoptotic rates

Apoptosis was determined by flow cytometry using an Annexin V/propidium iodide (PI) apoptosis kit (Thermo Fisher Scientific, Inc.). Briefly, after incubation with PYM (100 µg/ml), LY294002 (0.5 nM), PYM (100 µg/ml) plus IGF-1 (100 ng/ml) or an equal volume of DMSO for the control for 48 h, the cells were collected, washed with phosphate-buffered saline (PBS) and re-suspended in 0.5 ml 1X Annexin-binding buffer at a density of 5×105 cells/ml. Annexin V-fluorescein isothiocyanate and PI were added and the cells were incubated for 10 min at room temperature. Samples were immediately analyzed by flow cytometry (FACSCalibur; BD Biosciences, San Jose, CA, USA) using BD Accuri C6 software version 2.8 (BD Biosciences).

Cell invasion assay

The invasive capacity of the cells was determined using Transwell chambers (Corning-Costar, Corning, NY, USA; cat no. 3422). Cells were incubated with PYM (100 µg/ml), LY294001221 (0.5 nM), PYM (100 µg/ml) plus IGF-1 (100 ng/ml) or an equal volume of DMSO for the negative control in serum-free DMEM for 12 h. Cells were then harvested and added to the upper chamber of a Transwell insert (1×104 cells in 100 µl), which was coated with a Matrigel® mix (Corning Incorporated, Corning, NY, USA). The lower chamber was filled with complete medium. After 48 h of incubation at 37°C, the cells on the upper surface of the membrane were removed. The cells attached to the lower surface of the membrane were fixed with 4% paraformaldehyde (Beyotime Institute of Biotechnology, Shanghai, China) for 20 min, stained with hematoxylin (Beyotime Institute of Biotechnology) for 5–10 min and counted under a microscope (Olympus CX41; Olympus Corp., Tokyo, Japan).

Western blot analysis

The effects of PYM, LY294002 or PYM plus IGF-1 on the expression of PI3K and Akt in EOMA cells was determined by western blot analysis. After incubation with PYM (100 µg/ml), LY294001221 (0.5 nM), PYM (100 µg/ml) plus IGF-1 (100 ng/ml) or an equal volume of DMSO for the negative control, cells were washed with PBS (Beyotime Institute of Biotechnology) and lysed with lysis buffer (Beyotime Institute of Biotechnology). Following centrifugation at 14,000 × g for 10 min at 4°C, the supernatants were collected and the protein concentration was determined using the Pierce Bicinchoninic Acid Protein Assay kit (Thermo Fisher Scientific). Equal amounts of protein (20 µg) were separated by 10% SDS-PAGE (Beyotime Institute of Biotechnology) and transferred to polyvinylidene difluoride membranes (Millipore, Billerica, MA, USA). After washing with Tris-buffered saline (TBS; Beyotime Institute of Biotechnology), the membranes were blocked with TBS containing 5% skimmed milk and incubated with primary antibodies against PI3K (rabbit monoclonal; Abcam, Cambridge, UK; cat. no. ab40755; 1:2,000; overnight incubation at 4°C), Akt (rabbit monoclonal; Abcam; cat. no. ab179463; 1:10,000; overnight incubation at 4°C), phosphorylated (p)-Akt (rabbit monoclonal; Abcam; cat. no. ab81283; 1:10,000; overnight incubation at 4°C) or GAPDH (rabbit polyclonal; Abcam; cat. no. ab181602; 1:10,000; overnight incubation at 4°C). After washing with TBS containing Tween 20 (Beyotime Institute of Biotechnology), the membranes were incubated with secondary antibody (horseradish peroxidase-conjugated goat anti-rabbit immunoglobulin G; Wuhan Boster Biological Technology, Ltd., Wuhan, China; cat. no. BA1055; 1:5,000; overnight incubation at 4°C). Then the membranes were developed using an enhanced chemiluminescence system (Alpha Fluochem Q; Thermo Fisher Scientific, Inc.). Finally, the protein bands were quantified by densitometry using Image-Pro Plus 6.0 software (Media Cybernetics, Rockville, MD, USA) and normalized to GAPDH.

Statistical analysis

All statistical analyses were performed using SPSS 19.0 statistical software program (SPSS Inc., Chicago, IL, USA) or GraphPad Prism 6.0 (GraphPad Software, La Jolla, CA, USA). Values are expressed as the mean ± standard deviation of three independent experiments. P<0.05 was considered to indicate a statistically significant difference.

Results

PYM inhibits the proliferation, induces apoptosis and reduces the invasive ability of EOMA cells

The EOMA cell line is derived from hemangioendothelioma, a type of hemangioma, of an adult mouse. In the present study, EOMA cells were used as a pathological model of hemangioma. First, the effects of PYM treatment on the biological behavior of EOMA cells were tested. Treatment with PYM (0.5–500 µM) inhibited the growth of EOMA cells in a time-and dose-dependant manner (Fig. 1A). In particular, after treatment with PYM at 500 µM for 72 h, cell growth was inhibited by 70%. There were fewer differences between 48- and 72-h treatments with 100 µM PYM compared to lower doses of PYM. Based on these results, 100 µM PYM was selected as the concentration to be used in the subsequent experiments.

Necrosis and apoptosis are two main modes of cell death. To explore the mode of growth inhibition induced by PYM, flow cytometry was used to determine the apoptotic rate after treatment with PYM. As shown in Fig. 1B, PYM treatment at 100 µM for 48 h significantly induced a higher apoptotic rate than that of untreated cells (18.9 vs. 2.5%). Thus, apoptosis represented a mode of cell death caused by PYM treatment.

Tumor invasion is an important step of tumor progression and in the development of metastasis. Therefore, inhibition of the invasive ability of tumor cells is a desired property of anti-tumor agents. In the present study, a Transwell assay demonstrated that PYM treatment at 100 µM for 48 h markedly reduced the number of EOMA cells that transgressed though the filters of the Transwell membranes (Fig. 1C), which indicated that the invasive ability of EOMA cells was inhibited by PYM.

PYM reduces the expression of PI3K, Akt and p-Akt proteins

The effects of PYM on the expression of angiogenesis-associated PI3K/Akt signaling proteins in EOMA cells were determined by western blot analysis. After treatment with PYM (100 µM), the expression levels of PI3K, Akt and p-Akt were markedly reduced (Fig. 2). These results provided direct evidence that PYM treatment affects the PI3K/Akt signaling pathway. However, the association between reduction of protein expression and changes in the biological behavior of EOMA cells following PYM treatment require further study.

PI3K inhibitor LY294002 and PYM inhibit the expression of PI3K and p-Akt, while the effects of PYM are blocked by PI3K activator IGF-1

To explore the association between inhibition of the PI3K/Akt pathway and changes in the biological behavior of EOMA cells in response to PYM treatment, the PI3K inhibitor LY294002 and the PI3K activator IGF-1 were employed. LY294002 was used as a positive control for comparison with the effects of PYM, while IGF-1 was used to explore the possible role of the PI3K/Akt pathway in the mechanism of action of PYM.

As shown in Fig. 3, treatment with LY294002 or PYM significantly reduced the levels of PI3K and p-Akt expression. However, when the EOMA cells were treated by PYM together with IGF-1, the expression of PI3K was significantly increased and p-Akt was not inhibited. These results provided direct evidence that PYM exerts its anti-cancer effects by inhibiting PI3K/Akt signaling.

IGF-1 reverses the effects of PYM on the viability, apoptosis and invasive ability of EOMA cells

The effects of PI3K activator IGF-1 on the anti-cancer effects of PYM in EOMA cells were further investigated. Cell viability studies showed that, compared with PYM at 100 µg/ml, LY294002 at 0.5 nM was slightly more potent (Fig. 4A). Furthermore, the anti-proliferative effects of PYM on EOMA cells were attenuated by co-treatment with IGF-1 (100 ng/ml). These results suggested that inhibition of PI3K-associated signaling pathways may be involved in the anti-proliferative effects of PYM.

In the apoptosis assay, as shown in Fig. 4B, treatment with PYM or PI3K inhibitor LY294002 resulted in a significant induction of apoptosis by 22.9 and 16.9%, respectively, compared with that in the control group (4.2%). However, combined treatment with PYM and IGF-1 resulted in a similar apoptotic rate (7.5%) to that in the control group. These results were consistent with those of the MTT assay and confirmed the involvement of the PI3K pathway in PYM-induced apoptosis of EOMA cells.

Finally, in the Transwell invasion assays, treatment with PI3K inhibitor LY294002 significantly reduced the invasion ability of EOMA cells (P<0.01) and treatment with PYM caused a similar extent of reduction of the invasive potential (Fig. 4C). However, co-treatment with PI3K activator IGF-1 blocked this inhibitory effect of PYM. Again, these results demonstrated that PYM exerts it effects via a PI3K-associated mechanism.

In conclusion, PYM affected the biological behavior of EOMA cells, including cell viability, cell apoptosis and invasive ability, by inhibiting the PI3K/Akt pathway.

Discussion

PYM is a glycopeptide antibiotic which was identified and developed into a pharmaceutical drug in China, and which has been clinically used for the treatment of various tumor types (15). The most common and effective use of PYM is in the treatment of hemangioma, which is a benign type of tumor derived from endothelial cells. After treatment with PYM alone or in combination with dexamethasone, patients with hemangioma were cured at rates of 70–100% (2,912). The effects of PYM on hemangioma-derived endothelial cells have been studied in vitro. PYM treatment (100–300 µg/ml) resulted in a significant and dose-dependent induction of apoptosis in human hemangioma-derived endothelial cells (HemECs) (24). These results were in line with the results of the present study, which showed that PYM inhibited the proliferation of EOMA cells in a dose- and time-dependent manner and significantly induced apoptosis at 100 µg/ml. Thus, induction of apoptosis may be an important mechanism of action of drugs used for the clinical treatment of hemangioma.

It is thought that PYM exerts its anti-proliferative activity by inhibiting the synthesis of DNA and DNA strands breaks in oral cancer cells (23), and these damages may lead to the apoptosis or necrosis of KB cells (27). However, to date, the underlying mechanisms of PYM-mediated DNA damage and induction of apoptosis have not been sufficiently elucidated. Yang et al (28) reported that PYM may induce apoptosis by activating c-Jun N-terminal kinases and inhibiting the activity of extracellular signal-regulated kinases 1 and -2 in KB cells. Zhao et al (25) reported that patients with maxillofacial squamous cell carcinomas subjected to microwave-induced hyperthermia followed by intravenous injection of PYM, exhibited an increased number of apoptotic cancer cells; this effect was likely to be mediated via downregulation of Bcl-2 and upregulation of Bax. In HemECs, the induction of apoptosis was attributed to the activation of the p53 pathway (24). The present study demonstrated that the PI3K/Akt pathway has a significant role in PYM-induced inhibition of cell viability and invasive ability as well as induction of apoptosis in EOMA cells.

PI3K/Akt/mTOR signaling is the key regulatory pathway for certain essential cellular processes, including cell survival, growth and differentiation. As this pathway is over-activated in various cancer types, it is considered as an ideal target for anti-cancer drugs (2931). In the present study, PYM at 100 µg/ml acted in a similar manner to PI3K inhibitor LY294002 at 0.5 nM; the two compounds significantly inhibited EOMA-cell growth, induced apoptosis and decreased the invasive ability of the cells. Furthermore, all of these PYM-induced effects were blocked by PI3K activator IGF-1. Finally, western blot analysis confirmed that PYM treatment significantly decreased the expression of PI3K and p-Akt. It was therefore concluded that PYM affects the biological behavior of EOMA cells by inhibiting the PI3K/Akt pathway.

mTOR is one of main downstream signal regulators of the PI3K/Akt pathway. It has a central role in positively regulating cell growth, survival and other cell functions (32). When mTOR binds rapamycin-insensitive companion of mTOR (rictor), the resulting mTORC2 complex can activate the PI3K/Akt pathway by phosphorylating Akt (33), which forms a positive feedback loop to promote tumor-cell proliferation. Zheng et al (34) reported that small hairpin RNA-mediated knockdown of rictor in EOMA cells reduced the phosphorylation of Akt, which suppressed cell proliferation and invasion. The effects of PYM on mTOR and associated binding units should be explored in future studies in order to further specify the exact mechanisms of action of PYM.

Similar to the effects of other bleomycins, such as those of a combined drug composed of 69% bleomycin A2 29.3% bleomycin B2 and 1.7% of PYM (35), PYM has adverse effects of pulmonary fibrosis (36). Recent studies showed that bleomycin-induced pulmonary fibrosis may be associated with PI3Kγ. In vivo, PI3Kγ knockout mice exhibited reduced mortality and fibrosis compared with those of C57Bl/6j mice after instillation of bleomycin (37). In vitro, PI3Kγ inhibitor AS605240 protected against bleomycin-induced pulmonary injury, angiogenesis and fibrosis through the modulation of leukocyte, fibroblast and endothelial cell functions (37,38). The above studies implied that bleomycin induces fibrosis by activating the PI3Kγ pathway; however, in the present study, PYM was demonstrated to inhibit the PI3K/Akt pathway. A reasonable explanation for this apparent inconsistency in results may be the possibility that PYM selectively activates PI3Kγ, while specifically inhibiting other PI3K sub-family members. This hypothesis should be examined in future studies.

In conclusion, the results of the present study showed that PYM inhibited the viability, induced apoptosis and reduced the invasive ability of EOMA cells by inhibiting the PI3K/Akt pathway. Further studies should focus on the effect of PYM on downstream signaling proteins of the PI3K/Akt pathway and sub-families of PI3K.

Acknowledgments

This study was supported by the Science and Technology Planning Project of Guangdong Province, China (no. 2011B060300019).

References

1 

Meisheng X: Histopathologic study of esophageal squamous cell carcinoma treated preoperatively with Pingyangmycin. Chin Med J (Engl). 92:343–348. 1979.

2 

Zheng JW, Zhou Q, Yang XJ, He Y, Wang YA, Ye WM, Zhu HG and Zhang ZY: Intralesional injection of Pingyangmycin may be an effective treatment for epulis. Med Hypotheses. 72:453–454. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Liang XH, He YW, Tang YL, Wu JL, Cao XP, Xiao GZ and Mao ZY: Thermochemotherapy of lower lip squamous cell carcinoma without metastases: An experience of 31 cases. J Craniomaxillofac Surg. 38:260–265. 2010. View Article : Google Scholar

4 

Guan JY, He XF, Chen Y, Zeng QL, Mei QL and Li YH: Percutaneous intratumoral injection with pingyangmycin lipiodol emulsion for the treatment of recurrent sacrococcygeal chordomas. J Vasc Interv Radiol. 22:1216–1220. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Qi W, Guo J, Wu S, Su B, Zhang L, Pan J and Zhang J: Synergistic effect of nanosecond pulsed electric field combined with low-dose of pingyangmycin on salivary adenoid cystic carcinoma. Oncol Rep. 31:2220–2228. 2014.PubMed/NCBI

6 

Bai N, Chen YZ, Fu YJ, Wu P and Zhang WN: A clinical study of pingyangmycin sclerotherapy for venous malformation: An evaluation of 281 consecutive patients. J Clin Pharm Ther. 39:521–526. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Jia R, Xu S, Huang X, Song X, Pan H, Zhang L, He F, Lin M, Ge S and Fan X: Pingyangmycin as first-line treatment for low-flow orbital or periorbital venous malformations: Evaluation of 33 consecutive patients. JAMA Ophthalmol. 132:942–948. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Yue H, Qian J, Elner VM, Guo J, Yuan YF, Zhang R and Ge Q: Treatment of orbital vascular malformations with intralesional injection of pingyangmycin. Br J Ophthalmol. 97:739–745. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Luo QF and Zhao FY: The effects of Bleomycin A5 on infantile maxillofacial haemangioma. Head Face Med. 7:112011. View Article : Google Scholar : PubMed/NCBI

10 

Luo Q and Zhao F: How to use bleomycin A5 for infantile maxillofacial haemangiomas: Clinical evaluation of 82 consecutive cases. J Craniomaxillofac Surg. 39:482–486. 2011. View Article : Google Scholar

11 

Hou J, Wang M, Tang H, Wang Y and Huang H: Pingyangmycin sclerotherapy for infantile hemangiomas in oral and maxillofacial regions: An evaluation of 66 consecutive patients. Int J Oral Maxillofac Surg. 40:1246–1251. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Yang Y, Sun M, Cheng X, Hu X, Zhang P, Ma Q, Li J, Tian L and Lei D: Bleomycin A5 plus dexamethasone for control of growth in infantile parotid hemangiomas. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 108:62–69. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Zheng JW, Zhou Q, Yang XJ, Wang YA, Fan XD, Zhou GY, Zhang ZY and Suen JY: Treatment guideline for hemangiomas and vascular malformations of the head and neck. Head Neck. 32:1088–1098. 2010. View Article : Google Scholar

14 

Greenberger S and Bischoff J: Infantile hemangioma-mechanism(s) of drug action on a vascular tumor. Cold Spring Harb Perspect Med. 1:a0064602011. View Article : Google Scholar

15 

Greenberger S and Bischoff J: Pathogenesis of infantile haemangioma. Br J Dermatol. 169:12–19. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Verheul HM and Pinedo HM: The role of vascular endothelial growth factor (VEGF) in tumor angiogenesis and early clinical development of VEGF-receptor kinase inhibitors. Clin Breast Cancer. 1(Suppl 1): S80–S84. 2000. View Article : Google Scholar

17 

Boye E and Olsen BR: Signaling mechanisms in infantile hemangioma. Curr Opin Hematol. 16:202–208. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Augustin HG, Koh GY, Thurston G and Alitalo K: Control of vascular morphogenesis and homeostasis through the angiopoietin-Tie system. Nat Rev Mol Cell Biol. 10:165–177. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Phng LK and Gerhardt H: Angiogenesis: A team effort coordinated by notch. Dev Cell. 16:196–208. 2009. View Article : Google Scholar : PubMed/NCBI

20 

Bauer TM, Patel MR and Infante JR: Targeting PI3 kinase in cancer. Pharmacol Ther. 146:53–60. 2015. View Article : Google Scholar

21 

Phung TL, Eyiah-Mensah G, O'Donnell RK, Bieniek R, Shechter S, Walsh K, Kuperwasser C and Benjamin LE: Endothelial Akt signaling is rate-limiting for rapamycin inhibition of mouse mammary tumor progression. Cancer Res. 67:5070–5075. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Guba M, von Breitenbuch P, Steinbauer M, Koehl G, Flegel S, Hornung M, Bruns CJ, Zuelke C, Farkas S, Anthuber M, et al: Rapamycin inhibits primary and metastatic tumor growth by antiangiogenesis: Involvement of vascular endothelial growth factor. Nat Med. 8:128–135. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Tai KW, Chang YC, Chou LS and Chou MY: Cytotoxic effect of pingyangmycin on cultured KB cells. Oral Oncol. 34:219–223. 1998. View Article : Google Scholar : PubMed/NCBI

24 

Tu JB, Li QY, Jiang F, Hu XY, Ma RZ, Dong Q, Zhang H, Pattar P and Li SX: Pingyangmycin stimulates apoptosis in human hemangioma-derived endothelial cells through activation of the p53 pathway. Mol Med Rep. 10:301–305. 2014.PubMed/NCBI

25 

Zhao J, Wang SZ, Tang XF, Liu N, Zhao D and Mao ZY: Analysis of thermochemotherapy-induced apoptosis and the protein expressions of Bcl-2 and Bax in maxillofacial squamous cell carcinomas. Med Oncol. 28(Suppl 1): S354–S359. 2011. View Article : Google Scholar

26 

Huang Y, Li P, Xia S, Zhuo Y and Wu L: Proapoptotic effect and the mechanism of action of pingyangmycin on cavernous hemangiomas. Exp Ther Med. 7:473–477. 2014.PubMed/NCBI

27 

Tai KW, Chou MY, Hu CC, Yang JJ and Chang YC: Induction of apoptosis in KB cells by pingyangmycin. Oral Oncol. 36:242–247. 2000. View Article : Google Scholar : PubMed/NCBI

28 

Yang LC, Yang SH, Tai KW, Chou MY and Yang JJ: MEK inhibition enhances bleomycin A5-induced apoptosis in an oral cancer cell line: Signaling mechanisms and therapeutic opportunities. J Oral Pathol Med. 33:37–45. 2004. View Article : Google Scholar

29 

Fruman DA and Rommel C: PI3K and cancer: Lessons, challenges and opportunities. Nat Rev Drug Discov. 13:140–156. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Hassan B, Akcakanat A, Holder AM and Meric-Bernstam F: Targeting the PI3-kinase/Akt/mTOR signaling pathway. Surg Oncol Clin N Am. 22:641–664. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Khan KH, Yap TA, Yan L and Cunningham D: Targeting the PI3K-AKT-mTOR signaling network in cancer. Chin J Cancer. 32:253–265. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Sun SY: MTOR kinase inhibitors as potential cancer therapeutic drugs. Cancer Lett. 340:1–8. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Alessi DR, Pearce LR and Garcia-Martinez JM: New insights into mTOR signaling: MTORC2 and beyond. Sci Signal. 2:pe272009. View Article : Google Scholar : PubMed/NCBI

34 

Zheng NN, Ding XD and Zhang HP: Targeting rictor inhibits mouse vascular tumor cell proliferation and invasion in vitro and tumor growth in vivo. Neoplasma. 60:41–45. 2013. View Article : Google Scholar

35 

Aouida M and Ramotar D: A new twist in cellular resistance to the anticancer drug bleomycin-A5. Curr Drug Metab. 11:595–602. 2010. View Article : Google Scholar : PubMed/NCBI

36 

Raisfeld IH: Pulmonary toxicity of bleomycin analogs. Toxicol Appl Pharmacol. 56:326–336. 1980. View Article : Google Scholar : PubMed/NCBI

37 

Russo RC, Garcia CC, Barcelos LS, Rachid MA, Guabiraba R, Roffê E, Souza AL, Sousa LP, Mirolo M, Doni A, et al: Phosphoinositide 3-kinase γ plays a critical role in bleomycin-induced pulmonary inflammation and fibrosis in mice. J Leukoc Biol. 89:269–282. 2011. View Article : Google Scholar

38 

Wei X, Han J, Chen ZZ, Qi BW, Wang GC, Ma YH, Zheng H, Luo YF, Wei YQ and Chen LJ: A phosphoinositide 3-kinase-gamma inhibitor, AS605240 prevents bleomycin-induced pulmonary fibrosis in rats. Biochem Biophys Res Commun. 397:311–317. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2015
Volume 12 Issue 6

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
Peng LX, Zhao P, Zhao HS, Pan E, Yang BB and Li Q: Phosphoinositide 3-kinase/Akt pathway is involved in pingyangmycin‑induced growth inhibition, apoptosis and reduction of invasive potential in EOMA mouse hemangioendothelioma cells. Mol Med Rep 12: 8275-8281, 2015
APA
Peng, L., Zhao, P., Zhao, H., Pan, E., Yang, B., & Li, Q. (2015). Phosphoinositide 3-kinase/Akt pathway is involved in pingyangmycin‑induced growth inhibition, apoptosis and reduction of invasive potential in EOMA mouse hemangioendothelioma cells. Molecular Medicine Reports, 12, 8275-8281. https://doi.org/10.3892/mmr.2015.4447
MLA
Peng, L., Zhao, P., Zhao, H., Pan, E., Yang, B., Li, Q."Phosphoinositide 3-kinase/Akt pathway is involved in pingyangmycin‑induced growth inhibition, apoptosis and reduction of invasive potential in EOMA mouse hemangioendothelioma cells". Molecular Medicine Reports 12.6 (2015): 8275-8281.
Chicago
Peng, L., Zhao, P., Zhao, H., Pan, E., Yang, B., Li, Q."Phosphoinositide 3-kinase/Akt pathway is involved in pingyangmycin‑induced growth inhibition, apoptosis and reduction of invasive potential in EOMA mouse hemangioendothelioma cells". Molecular Medicine Reports 12, no. 6 (2015): 8275-8281. https://doi.org/10.3892/mmr.2015.4447