Downregulation of Sp1 is involved in honokiol-induced cell cycle arrest and apoptosis in human malignant pleural mesothelioma cells

  • Authors:
    • Jung-Il Chae
    • Young-Joo Jeon
    • Jung-Hyun Shim
  • View Affiliations

  • Published online on: March 20, 2013     https://doi.org/10.3892/or.2013.2353
  • Pages: 2318-2324
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Malignant pleural mesothelioma (MPM) is an extremely aggressive type of cancer and is associated with a poor patient prognosis due to its rapid progression. Novel therapeutic agents such as honokiol (HNK) improve the clinical outcomes of cancer therapy, yet the mechanisms involved have not been fully elucidated. The present study examined the regulatory effects of HNK on the growth and apoptosis of MSTO-211H mesothelioma cells and investigated its anticancer mechanism. The results revealed that HNK significantly reduced the cell viability and increased the sub-G1 population in MSTO-211H cells and suppressed the expression of the specificity protein 1 protein (Sp1). HNK reduced the transcriptional activity of Sp1 regulatory proteins, including cyclin D1, Mcl-1 and survivin, and, thus, induced apoptosis signaling pathways by increasing Bax, reducing Bid and Bcl-xl and activating caspase-3 and PARP in mesothelioma cells. The results suggest that Sp1, a novel molecular target of HNK, may be related to cell cycle arrest and apoptosis induction through the modulation of signal transduction pathways in MPM.

Introduction

Malignant pleural mesothelioma (MPM) is a deadly disease that arises from the mesothelial layer of the pleural cavity and afflicts 2,000 to 3,000 individuals every year in the US (1,2). MPM has been linked to asbestos, iron and Simian virus 40 exposure (35) and represents a worldwide concern since its incidence has greatly increased during the last three decades (6). MPM progresses rapidly and is extraordinarily delibitating, particularly in the later stages. In addition, its poor prognosis stems from an overall survival period of only 9–17 months from the initial diagnosis (2). An aggressive multimodality therapy for MPM can offer survival benefits not only for a small subset of patients in the early stages of the disease but also for terminal patients with unresectable tumors who are not responsive to current therapeutic approaches (2,7). This suggests an urgent need for new therapeutic targets and effective therapies for improving the prognosis of MPM. Several studies have reported that novel plant-derived compounds act as antitumor agents through the modulation of biological pathways (8). This suggests that chemotherapeutic agents used in the treatment of MPM may activate apoptosis pathways.

The present study focused on honokiol (HNK) as this anticancer agent is widely used as the drug of choice in treating cancer. HNK, a biologically active biphenolic compound isolated from the Magnolia species, has multiple medicinal uses against oxidative stress, microbial infection and anxiety, among others (9). Noteworthy, HNK demonstrates antineoplastic properties in vitro against various types of cancers and is effective in vivo in treating angiosarcoma (10), colorectal carcinoma (11), breast cancer (12) and gastric cancer (13). Several studies have suggested that the cytotoxic effect of HNK is caused by the predominant activation of distinct apoptotic mechanisms in tumor cells (10,11,1416). Apart from apoptosis, the suppression of cancer cell proliferation is one of the most important strategies for antitumorigenesis.

Specificity protein 1 (Sp1), a constitutive transcription factor, plays an important role in various physiological processes such as cell cycle regulation, apoptosis and angiogenesis (17,18). In this regard, many studies have documented that Sp1 activity and expression levels are increased in various types of human cancers including prostate cancer, pancreatic adenocarcinoma, thyroid cancer, hepatocellular carcinomas, lung cancer, colorectal cancer, gastric cancer and breast cancer, but not in normal tissues (19,20).

Therefore, the present study examined the regulatory effects of HNK on the growth and apoptosis of MSTO-211H mesothelioma cells and investigated its anticancer mechanism in relation to Sp1.

Materials and methods

Cell culture and transfection

MSTO-211H cells obtained from the American Tissue Culture Collection (Manassas, VA) were maintained at 37°C in RPMI-1640 medium supplemented with 10% fetal bovine serum (FBS) and penicillin/streptomycin in the presence of 5% CO2. For transient transfection, 1.4×106 cells were plated onto a 60-mm cell culture dish, grown overnight, and transfected with DNA using Lipofectamine (Invitrogen).

MTS assay

The effects of HNK on cell viability were determined using the CellTiter 96 AQueous One Solution cell proliferation assay kit (Promega, Madison, WI) according to the manufacturer’s instructions. MSTO-211H cells were seeded onto a 96-well plate for 24 h and then treated with HNK for 24 and 48 h. The MTS solution was analyzed using the GloMax-Multi Microplate Mulimode reader (Promega) at 482 and 690 nm.

DAPI staining

The number of cells undergoing apoptosis following treatment with HNK was quantified using DAPI staining. Cells with nuclear condensation and fragmentation were determined using the nucleic acid stain 4′-6-diamidino-2-phenylindole (DAPI) (Sigma-Aldrich). Following a 48-h treatment of HNK at different concentrations (2, 4 and 6 μg/ml), MSTO-211H cells were harvested and fixed in 100% methanol at room temperature for 20 min. The cells were seeded on slides, stained with DAPI (2 mg/ml), and then monitored using a FluoView confocal laser microscope (FluoView FV10i, Olympus Corp., Tokyo, Japan).

Propidium iodide staining

Following HNK treatment of the MSTO-211H cells, detached cells were collected separately, and adherent cells were dissociated by trypsin-EDTA. The cells were washed with cold PBS and then pooled and centrifuged before being fixed in 70% ethanol overnight at −20°C. Prior to the flow cytometric analysis, the cells were centrifuged and incubated for 30 min at 37°C in PBS to allow for the release of low-molecular-weight DNA. After centrifugation, cell pellets were resuspended and treated with 150 mg/ml RNase A and 20 mg/ml propidium iodide (PI) using a MACSQuant Analyzer (Miltenyi Biotec GmbH, Bergisch Gladbach, Germany).

Western blot analysis

MSTO-211H cells treated with HNK were washed three times with ice-cold phosphate-buffered saline (PBS) and harvested in an ice-cold PRO-PREP™ protein extraction solution (Intron Biotechnology, Inc., Korea) containing a protease inhibitor. Protein concentrations were measured using the Bradford protein assay. Protein samples were separated on an SDS-PAGE gel and transferred to a PVDF membrane (Immobilon-P, Millipore) using a semi-dry blotting apparatus. ECL western blotting was performed according to the manufacturer’s instructions (Thermo Scientific, Rockford, IL).

In vitro pull-down assays

Cell lysates were incubated with HNK-Sepharose 4B (or Sepharose 4B only as a control) beads (50 μl, 50% slurry) in reaction buffer (50 mM Tris, pH 7.5, 5 mM EDTA, 150 mM NaCl, 1 mM dithiothreitol, 0.01% Nonidet P-40, 2 μg/ml bovine serum albumin, 0.02 mM phenylmethylsulfonyl fluoride and 1X protease inhibitor cocktail). After incubation with gentle rocking overnight at 4°C, the beads were washed five times with a buffer solution (50 mM Tris, pH 7.5, 5 mM EDTA, 150 mM NaCl, 1 mM dithiothreitol, 0.01% Nonidet P-40 and 0.02 mM phenylmethylsulfonyl fluoride) and proteins bound to the beads were analyzed by western blotting using the anti-Sp1 antibody.

Luciferase assay

MSTO-211H cells were grown to 50–80% confluence on a 60-mm cell dish and were transfected with Lipofectamine (Invitrogen) with 1 μg each of cyclin D1-luc, Mcl-1-luc and survivin-luc and 0.5 μg of pCMV/β-gal plasmids. The luciferase reporter assay kit from Promega was used following the manufacturer’s protocol. Luciferase activity was measured with the Lumat LB 9507 luminometer (EG&G Berthold). The values shown represent an average of three experiments for each sample.

Statistical analysis

The statistical significance of differences was assessed using the Student’s t-test. A P-value <0.05 relative to the control was considered to indicate a statistically significant result.

Results

Growth inhibitory effect of HNK on a human mesothelioma cell line

We investigated the effects of HNK on a human mesothelioma cell line. The structure of HNK is shown in Fig. 1A. To ascertain the efficiency of HNK as an anticancer drug, the effects of HNK were tested using an MTS assay using MSTO-211H cells. As shown in Fig. 1B, the efficiency of HNK in altering the cell viability of MSTO-211H cells was assayed after 24 and 48 h of incubation in HNK-containing medium at different concentrations (2, 4 or 6 μg/ml). The viability curves revealed that HNK reduced MSTO-211H cell viability at 24 and 48 h, in a concentration-dependent manner (P<0.05). In particular, a maximum reduction in cell viability was observed at 48 h. Apoptosis-induced changes in the cell morphology in the HNK-containing medium were observed. After 48 h, the apoptotic phenotype showed cell rounding, cytoplasmic blebbing and irregularities in shape, indicating a sharp increase in the apoptosis of HNK-treated MSTO-211H cells in a concentration-dependent manner (Fig. 1C).

HNK induces cell cycle arrest at the G1 phase and apoptosis in a human mesothelioma cell line

Cancer cell growth can be suppressed by cell cycle arrest or apoptosis induction or both (8). Morphological changes induced by HNK were examined following treatment of MSTO-211H cells with HNK at a concentration of 2, 4 and 6 μg/ml for 48 h. In the DAPI staining analysis, MSTO-211H cells were observed via fluorescence microscopy following exposure to DAPI, which specifically stains nuclei. The results indicated the presence of nuclear condensation and perinuclear apoptotic bodies in the MSTO-211H cells following HNK treatment at concentrations of 4 and 6 μg/ml for 48 h (Fig. 2A). The percentage of apoptosis was determined to be 6±1.6, 26±11.3 and 20±20.3% at concentrations of 2, 4 and 6 ng/ml HNK compared with the untreated control cells.

For the determination of whether the HNK-mediated growth inhibition of MSTO-211H cells was attributable to cell cycle arrest, cell cycle distribution was analyzed by FACS analysis. As shown in Fig. 2B, a significant increase in the number of sub-G1 cells was noted when compared with the control (5.3±1.9, 7.2±3.5 and 34.7±4.6% in the presence of 2, 4 and 6 μg/ml HNK, respectively, in MSTO-211H cells). The histogram in Fig. 2C shows the quantification of FACS data.

HNK suppresses Sp 1 expression in a human mesothelioma cell line

The potential interaction between HNK and apoptosis-related proteins was examined since HNK has been found to have marked effects on apoptosis and cell cycle arrest in various cancer cell lines. Therefore, an in vitro pull-down assay was performed using HNK-conjugated Sepharose 4B beads. Sp1 was found to bind to HNK-conjugated Sepharose beads but not to Sepharose beads alone (Fig. 3A), providing support for the direct interaction between HNK and Sp1 and suggesting that Sp1 may be a possible target of HNK in human MSTO-211H cells. The effects of HNK treatment on the levels of Sp1 were examined by western blotting. As shown in Fig. 3B, HNK treatment led to a sharp decrease in the level of Sp1 in MSTO-211H cells at 48 h after treatment.

In order to better characterize the apoptotic action of HNK, the cleavage of the DNA repair enzyme poly(adenosine diphosphate-ribose) polymerase (PARP) as a substrate for activated caspase-3 and caspase-3 expression levels were confirmed by western blotting (Fig. 3C). There were increases in cleaved PARP and activated caspase-3 following HNK (6 μg/ml) treatment of MSTO-211H cells in a time-dependent manner.

HNK modulates regulators of cell cycle arrest and apoptosis

Sp1 activation has been shown to regulate the expression of various gene products involved in cell survival and proliferation (18,21). Based on the results for DNA fragmentation and cell cycle distribution, the potential relationship between the induction of apoptosis and cell cycle arrest by HNK and the transcriptional activity of cell cycle regulatory proteins and apoptosis-related transcription factors was examined. We investigated the effects of HNK on cell cycle regulator cyclin D1, anti-apoptotic proteins Mcl-1 and survivin dependent gene transcriptions. When the MSTO-211H cells transiently transfected with cyclin D1-luc (Fig. 4A), Mcl-1-luc (Fig. 4B), and survivin-luc (Fig. 4C) constructs were treated with 2, 4 and 6 μg/ml HNK, significant decreases in transcriptional activity mediated by these proteins were observed in a dose-dependent manner. The cyclin D1, Mcl-1 and survivin protein expression levels were examined using western blotting. The results indicated that these levels decreased gradually after HNK treatment (Fig. 5).

HNK alters the expression of proteins associated with the cell cycle and survival

The expression of genes involved in the regulation of apoptosis was analyzed. The apoptotic cell death by HNK was well correlated with the activation of caspase-3 through the cleavage of the pro-form during HNK treatment. The upregulation and downregulation of Bax and Bid, respectively, and Bcl-xL expression appeared to be involved in apoptotic cell death. In addition, PARP cleavage was induced by HNK (Fig. 6). These results indicate that downregulation of cyclin D1, Mcl-1 and survivin and activation of caspase-3 were involved in the HNK-mediated growth arrest and apoptotic cell death in MSTO-211H cells.

Discussion

Compounds of natural origin such as topotecan, irinotecan, etoposide, teniposide and paclitaxil are widely employed in clinical trials as chemotherapeutic agents (22). Therefore, the evaluation of natural compounds with anticancer properties has been considered to play an important role in the development of anticancer drug. The present study focused on the anticancer effects of HNK, a pharmacologically active component found in the traditional Chinese medicinal herb, Magnolia species. Previous studies have demonstrated the anticancer properties of HNK, including cell cycle arrest, apoptosis and necrosis in a variety of cancer cell lines such as human breast cancer (23), human colorectal carcinoma RKO (24), human squamous lung cancer CH27 (15), human pancreatic cancer (25), adult T-cell leukemia (26) and hepatocellular carcinoma cells (27). This suggests that HNK may be involved in the apoptotic pathway. However, the exact mechanism underlying the anticancer activity of HNK remains unclear.

In this study, our findings along with the findings of previous research, suggest that HNK may induce apoptosis in human MPM cells. In this study, the apoptotic effects of HNK on MSTO-211H cells were examined through an MTS assay. This assay, a traditional method for discovering anticancer drugs, can be used to determine the cytotoxic effect and proliferation of cell lines (28,29). In this study, this assay was used to elucidate the apoptotic effects of HNK on MSTO-211H cells (Fig. 1).

As shown in Fig. 2, HNK inhibited the proliferation of MSTO-211H cells through cell cycle arrest at G0/G1 and induction of apoptosis.

Sp1, a transcription factor, is overexpressed in various human cancer cell lines (3034) and plays a role in the regulation of genes involved in most cellular processes (35). In this regard, Sp1 may exhibit transcriptional activity for promoters of genes involved in the progression, differentiation and oncogenesis of the cell cycle (19). In addition, Sp1 may be inhibited by compounds of natural origin with anticancer properties. For example, recent studies have demonstrated that flavonoids suppress Sp1 expression (36) and, thus, inhibit the known Sp1 target genes cyclin D1, Mcl-1 and survivin in a selective manner (3741). The results of the present study demonstrated the potential chemotherapeutic properties of HNK. That is, HNK suppressed Sp1 expression via direct interactions with Sp1, preventing Sp1 from binding to G-C-rich promoters. In addition, HNK inhibited the transcriptional activity and expression of Sp1 downstream proteins, including cyclin D1, Mcl-1 and survivin, in a dose-dependent manner (Figs. 4 and 5). HNK reduced Bid and Bcl-xL, increased Bax, and activated caspase-3 and PARP, suggesting that HNK regulated Sp1 and ultimately led to apoptotic cell death (Fig. 6).

The results regarding the chemopreventive effect of HNK as a compound of natural origin on MPM cells revealed that HNK downregulated Sp1 expression and Sp1 target transcription factors, including cyclin D1, Mcl-1 and survivin, thereby inducing the apoptosis of MSTO-211H cells. To conclude, this study is the first to suggest that the apoptosis induced by HNK in MSTO-211H cells may be mediated by the downregulation of Sp1. The results suggest that the antiproliferative and apoptotic effects of HNK are mediated through the suppression of Sp1-regulated gene products.

Acknowledgements

This research was supported by the Basic Science Research Program of the National Research Foundation Korea (NRF), funded by the Ministry of Education, Science and Technology (2011-0008463); the Next-Generation BioGreen 21 Program (PJ008116062011) of the Rural Development Administration of the Republic of Korea.

References

1 

Cugell DW and Kamp DW: Asbestos and the pleura: a review. Chest. 125:1103–1117. 2004. View Article : Google Scholar

2 

Tsao AS, Wistuba I, Roth JA and Kindler HL: Malignant pleural mesothelioma. J Clin Oncol. 27:2081–2090. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Broaddus VC: Asbestos, the mesothelial cell and malignancy: a matter of life or death. Am J Respir Cell Mol Biol. 17:657–659. 1997. View Article : Google Scholar : PubMed/NCBI

4 

Morinaga K, Kishimoto T, Sakatani M, Akira M, Yokoyama K and Sera Y: Asbestos-related lung cancer and mesothelioma in Japan. Ind Health. 39:65–74. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Dufresne A, Begin R, Churg A and Masse S: Mineral fiber content of lungs in patients with mesothelioma seeking compensation in Quebec. Am J Respir Crit Care Med. 153:711–718. 1996. View Article : Google Scholar : PubMed/NCBI

6 

Britton M: The epidemiology of mesothelioma. Semin Oncol. 29:18–25. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Zellos L and Sugarbaker DJ: Current surgical management of malignant pleural mesothelioma. Curr Oncol Rep. 4:354–360. 2002. View Article : Google Scholar : PubMed/NCBI

8 

Gupta SC, Kim JH, Prasad S and Aggarwal BB: Regulation of survival, proliferation, invasion, angiogenesis, and metastasis of tumor cells through modulation of inflammatory pathways by nutraceuticals. Cancer Metastasis Rev. 29:405–434. 2010. View Article : Google Scholar : PubMed/NCBI

9 

Liou KT, Shen YC, Chen CF, Tsao CM and Tsai SK: Honokiol protects rat brain from focal cerebral ischemia-reperfusion injury by inhibiting neutrophil infiltration and reactive oxygen species production. Brain Res. 992:159–166. 2003. View Article : Google Scholar

10 

Bai X, Cerimele F, Ushio-Fukai M, et al: Honokiol, a small molecular weight natural product, inhibits angiogenesis in vitro and tumor growth in vivo. J Biol Chem. 278:35501–35507. 2003. View Article : Google Scholar : PubMed/NCBI

11 

Chen F, Wang T, Wu YF, et al: Honokiol: a potent chemotherapy candidate for human colorectal carcinoma. World J Gastroenterol. 10:3459–3463. 2004.PubMed/NCBI

12 

Wolf I, O’Kelly J, Wakimoto N, et al: Honokiol, a natural biphenyl, inhibits in vitro and in vivo growth of breast cancer through induction of apoptosis and cell cycle arrest. Int J Oncol. 30:1529–1537. 2007.PubMed/NCBI

13 

Sheu ML, Liu SH and Lan KH: Honokiol induces calpain-mediated glucose-regulated protein-94 cleavage and apoptosis in human gastric cancer cells and reduces tumor growth. PLoS One. 2:e10962007. View Article : Google Scholar : PubMed/NCBI

14 

Ishitsuka K, Hideshima T, Hamasaki M, et al: Honokiol overcomes conventional drug resistance in human multiple myeloma by induction of caspase-dependent and -independent apoptosis. Blood. 106:1794–1800. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Yang SE, Hsieh MT, Tsai TH and Hsu SL: Down-modulation of Bcl-XL, release of cytochrome c and sequential activation of caspases during honokiol-induced apoptosis in human squamous lung cancer CH27 cells. Biochem Pharmacol. 63:1641–1651. 2002. View Article : Google Scholar : PubMed/NCBI

16 

Shigemura K, Arbiser JL, Sun SY, et al: Honokiol, a natural plant product, inhibits the bone metastatic growth of human prostate cancer cells. Cancer. 109:1279–1289. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Chu S and Ferro TJ: Sp1: regulation of gene expression by phosphorylation. Gene. 348:1–11. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Deniaud E, Baguet J, Mathieu AL, Pages G, Marvel J and Leverrier Y: Overexpression of Sp1 transcription factor induces apoptosis. Oncogene. 25:7096–7105. 2006. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Sankpal UT, Goodison S, Abdelrahim M and Basha R: Targeting Sp1 transcription factors in prostate cancer therapy. Med Chem. 7:518–525. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Jutooru I, Chadalapaka G, Sreevalsan S, et al: Arsenic trioxide downregulates specificity protein (Sp) transcription factors and inhibits bladder cancer cell and tumor growth. Exp Cell Res. 316:2174–2188. 2010. View Article : Google Scholar

22 

Mann J: Natural products in cancer chemotherapy: past, present and future. Nat Rev Cancer. 2:143–148. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Park EJ, Min HY, Chung HJ, et al: Down-regulation of c-Src/EGFR-mediated signaling activation is involved in the honokiol-induced cell cycle arrest and apoptosis in MDA-MB-231 human breast cancer cells. Cancer Lett. 277:133–140. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Wang T, Chen F, Chen Z, et al: Honokiol induces apoptosis through p53-independent pathway in human colorectal cell line RKO. World J Gastroenterol. 10:2205–2208. 2004.PubMed/NCBI

25 

Arora S, Bhardwaj A, Srivastava SK, et al: Honokiol arrests cell cycle, induces apoptosis, and potentiates the cytotoxic effect of gemcitabine in human pancreatic cancer cells. PLoS One. 6:e215732011. View Article : Google Scholar : PubMed/NCBI

26 

Ishikawa C, Arbiser JL and Mori N: Honokiol induces cell cycle arrest and apoptosis via inhibition of survival signals in adult T-cell leukemia. Biochim Biophys Acta. 1820:879–887. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Rajendran P, Li F, Shanmugam MK, et al: Honokiol inhibits signal transducer and activator of transcription-3 signaling, proliferation, and survival of hepatocellular carcinoma cells via the protein tyrosine phosphatase SHP-1. J Cell Physiol. 227:2184–2195. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Riss TL and Moravec RA: Use of multiple assay endpoints to investigate the effects of incubation time, dose of toxin, and plating density in cell-based cytotoxicity assays. Assay Drug Dev Technol. 2:51–62. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Li Y, Huang W, Huang S, Du J and Huang C: Screening of anti-cancer agent using zebrafish: comparison with the MTT assay. Biochem Biophys Res Commun. 422:85–90. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Chiefari E, Brunetti A, Arturi F, et al: Increased expression of AP2 and Sp1 transcription factors in human thyroid tumors: a role in NIS expression regulation? BMC Cancer. 2:352002. View Article : Google Scholar : PubMed/NCBI

31 

Hosoi Y, Watanabe T, Nakagawa K, et al: Up-regulation of DNA-dependent protein kinase activity and Sp1 in colorectal cancer. Int J Oncol. 25:461–468. 2004.PubMed/NCBI

32 

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

33 

Yao JC, Wang L, Wei D, et al: Association between expression of transcription factor Sp1 and increased vascular endothelial growth factor expression, advanced stage, and poor survival in patients with resected gastric cancer. Clin Cancer Res. 10:4109–4117. 2004. View Article : Google Scholar

34 

Zannetti A, Del Vecchio S, Carriero MV, et al: Coordinate up-regulation of Sp1 DNA-binding activity and urokinase receptor expression in breast carcinoma. Cancer Res. 60:1546–1551. 2000.PubMed/NCBI

35 

Davie JR, He S, Li L, et al: Nuclear organization and chromatin dynamics - Sp1, Sp3 and histone deacetylases. Adv Enzyme Regul. 48:189–208. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Lee KA, Lee YJ, Ban JO, et al: The flavonoid resveratrol suppresses growth of human malignant pleural mesothelioma cells through direct inhibition of specificity protein 1. Int J Mol Med. 30:21–27. 2012.

37 

Culver C, Melvin A, Mudie S and Rocha S: HIF-1α depletion results in SP1-mediated cell cycle disruption and alters the cellular response to chemotherapeutic drugs. Cell Cycle. 10:1249–1260. 2011.

38 

Blume SW, Snyder RC, Ray R, Thomas S, Koller CA and Miller DM: Mithramycin inhibits SP1 binding and selectively inhibits transcriptional activity of the dihydrofolate reductase gene in vitro and in vivo. J Clin Invest. 88:1613–1621. 1991. View Article : Google Scholar : PubMed/NCBI

39 

Chintharlapalli S, Papineni S, Lei P, Pathi S and Safe S: Betulinic acid inhibits colon cancer cell and tumor growth and induces proteasome-dependent and -independent downregulation of specificity proteins (Sp) transcription factors. BMC Cancer. 11:3712011. View Article : Google Scholar

40 

Pietrzak M and Puzianowska-Kuznicka M: p53-dependent repression of the human MCL-1 gene encoding an anti-apoptotic member of the BCL-2 family: the role of Sp1 and of basic transcription factor binding sites in the MCL-1 promoter. Biol Chem. 389:383–393. 2008. View Article : Google Scholar : PubMed/NCBI

41 

Tapias A, Ciudad CJ, Roninson IB and Noe V: Regulation of Sp1 by cell cycle-related proteins. Cell Cycle. 7:2856–2867. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June 2013
Volume 29 Issue 6

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
Chae J, Jeon Y and Shim J: Downregulation of Sp1 is involved in honokiol-induced cell cycle arrest and apoptosis in human malignant pleural mesothelioma cells. Oncol Rep 29: 2318-2324, 2013
APA
Chae, J., Jeon, Y., & Shim, J. (2013). Downregulation of Sp1 is involved in honokiol-induced cell cycle arrest and apoptosis in human malignant pleural mesothelioma cells. Oncology Reports, 29, 2318-2324. https://doi.org/10.3892/or.2013.2353
MLA
Chae, J., Jeon, Y., Shim, J."Downregulation of Sp1 is involved in honokiol-induced cell cycle arrest and apoptosis in human malignant pleural mesothelioma cells". Oncology Reports 29.6 (2013): 2318-2324.
Chicago
Chae, J., Jeon, Y., Shim, J."Downregulation of Sp1 is involved in honokiol-induced cell cycle arrest and apoptosis in human malignant pleural mesothelioma cells". Oncology Reports 29, no. 6 (2013): 2318-2324. https://doi.org/10.3892/or.2013.2353