Skip to main content
Erschienen in: BMC Cancer 1/2010

Open Access 01.12.2010 | Research article

Oridonin induces apoptosis and senescence in colorectal cancer cells by increasing histone hyperacetylation and regulation of p16, p21, p27 and c-myc

verfasst von: Feng-Hou Gao, Xiao-Hui Hu, Wei Li, Hua Liu, Yan-Jie Zhang, Zhu-Ying Guo, Mang-Hua Xu, Shi-Ting Wang, Bin Jiang, Feng Liu, Ying-Zheng Zhao, Yong Fang, Fang-Yuan Chen, Ying-Li Wu

Erschienen in: BMC Cancer | Ausgabe 1/2010

Abstract

Background

Oridonin, a tetracycline diterpenoid compound, has the potential antitumor activities. Here, we evaluate the antitumor activity and action mechanisms of oridonin in colorectal cancer.

Methods

Effects of oridonin on cell proliferation were determined by using a CCK-8 Kit. Cell cycle distribution was determined by flow cytometry. Apoptosis was examined by analyzing subdiploid population and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay. Senescent cells were determined by senescence-associated β-galactosidase activity analysis. Semi-quantitative RT-PCR was used to examine the changes of mRNA of p16, p21, p27 and c-myc. The concomitant changes of protein expression were analyzed with Western blot. Expression of AcH3 and AcH4 were examined by immunofluorescence staining and Western blots. Effects of oridonin on colony formation of SW1116 were examined by Soft Agar assay. The in vivo efficacy of oridonin was detected using a xenograft colorectal cancer model in nude mice.

Results

Oridonin induced potent growth inhibition, cell cycle arrest, apoptosis, senescence and colony-forming inhibition in three colorectal cancer cell lines in a dose-dependent manner in vitro. Daily i.p. injection of oridonin (6.25, 12.5 or 25 mg/kg) for 28 days significantly inhibited the growth of SW1116 s.c. xenografts in BABL/C nude mice. With western blot and reverse transcription-PCR, we further showed that the antitumor activities of oridonin correlated with induction of histone (H3 and H4) hyperacetylation, activation of p21, p27 and p16, and suppression of c-myc expression.

Conclusion

Oridonin possesses potent in vitro and in vivo anti-colorectal cancer activities that correlated with induction of histone hyperacetylation and regulation of pathways critical for maintaining growth inhibition and cell cycle arrest. Therefore, oridonin may represent a novel therapeutic option in colorectal cancer treatment.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2407-10-610) contains supplementary material, which is available to authorized users.
Feng-Hou Gao, Xiao-Hui Hu contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

F-HG and X-HH carried out the data collection and data analysis, and drafted the manuscript. WL, HL and Y-JZ participated in the design of the study, performed the statistical analysis and participated in the interpretation of data. Z-YG, M-HX and S-TW participated in the design of the study and the acquisition and interpretation of data. BJ, FL, Y-ZZ, YF and F-YC were involved in the interpretation of data and critically revised the manuscript. Y-LW conceived the study, participated in its design and coordination, and helped in statistical analysis and drafting of the manuscript. All authors have read and approved the final manuscript.

Background

Colorectal cancer (CRC) is one of the most frequently diagnosed malignancies in both men and women, with more than 1,000,000 new cases annually worldwide [1]. Advances in therapies over the past decade have led to improved outcomes for many patients. Although curative resection is the major treatment option, approximately half of all patients eventually develop distant metastases. Liver metastases (LM) occur in more than 50% of CRC patients, but curative liver resection is possible only in 15% of them, resulting in 5-year survival rates of 30% on average [24]. Improving resectability rates and hopefully patient's prognosis by adding up front active chemotherapy and biological agents in metastatic CRC is a challenging opportunity for both medical and surgical oncologists [5]. Thus, effective new cytotoxic chemotherapy is needed for these diseases.
The terpenoids constitute the largest family of natural products; over 22,000 individual compounds of this class have been described, and the number of defined structures has doubled every decade since the 1970s [6]. In plants, terpenoids represent a chemical defense against environmental stress and provide a repair mechanism for wounds and injuries. Interestingly, effective ingredients in several plant-derived medicinal extracts are also terpenoid compounds of monoterpenoid, sesquiterpenoid, diterpenoid, triterpenoid and carotenoid groups. Experimental study shows that many of them have strong anti-tumor activity [7]. For example, Tanshinone IIA, the major active diterpene quinine in the herbal product from the roots of Salvia miltiorrhiza, is a commonly used Chinese plant remedy which seems to have some activity against breast cancer [8, 9]. Celastrol, a quinone methide triterpenoid, isolated from the Chinese Thunder of God Vine (Tripterygium wilfordii Hook F.), as well as triptolide, are currently being investigated in the search for prevention of tumor cell invasion [10]. Plant-derived terpenoids provide a challenging field to identify new potent natural anticancer compound for the therapy of colorectal cancer.
Oridonin, an ent-kaurane diterpenoid isolated from Rabdosia rubescens, is an important traditional Chinese herbal remedy. Studies showed that oridonin induced apoptosis in a variety of cancer cells including those from prostate, breast, non-small cell lung cancer, acute leukemia, glioblastoma multiforme and human melanoma cells. Cell culture experiments have indicated that oridonin inhibits cell cycle progression and induces apoptosis as well as enhance the phagocytosis of apoptotic cells by macrophages [11, 12]. Oridonin has also immunosuppressive properties both in vitro and in vivo [13]. However, mechanisms underlying the antitumor activity of oridonin and whether oridonin has the anti-colorectal cancer activity remain largely unknown.
In this report, we found that oridonin could induce potent growth inhibition, cell cycle arrest, apoptosis and senescence of colorectal cancer cells in vitro and in vivo. The antitumor activities of oridonin correlated with induction of histone (H3 and H4) hyperacetylation, activation of p21, p27 and p16, and suppression of c-myc expression.

Methods

Cell Culture and Reagents

The colorectal cancer cell lines SW1116, HT29 and HCT116 from Shanghai Institutes for Biological Sciences were incubated in humidified room air containing 5% CO2 at 37°C and cultured in McCOY'S 5A medium (Sigma, USA) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin-streptomycin (GIBCO BRL, Grand Island,NY). Cells were routinely grown in 100 mm plastic tissue culture dishes (Nunc, Roskilde, Denmark) and harvested with a solution of trypsin-EDTA when they were in logarithmic phase of growth. Cells were maintained at these culture conditions for all experiments. Oridonin (purity > 98%) was purchased from CHENGDU MUST BIO-TECHNOLOGY CO.LTD. It was dissolved in DMSO at a stock concentration of 100 mmol/L and store at -20°C. The stock solution was further diluted with cell culture medium to yield final oridonin concentrations.

Cell Proliferation Assay

Cells were seeded into 96-well plates at 2,000 to 3,000 live cells per well and treated with Oridonin (6.25-100 μM) for 3 days. The antiproliferative effect of Oridonin was assessed using Cell Count Kit-8 (Dojindo Molecular Technologies, Inc., Gaithersburg, MD).

Cell Cycle Analysis with Flow Cytometry

Cells treated with or without Oridonin (12.5 and 25 μmol/L) were harvested for flow cytometry analysis on day 1. Cells were fixed and stained with 0.1 mg/mL propidium iodide for DNA analysis with Becton Dickinson FACScan (Franklin Lakes, NJ) as described previously [14].

Detection of Apoptosis

Apoptosis was evaluated with flow cytometry and on cell smears using the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay (In situ Cell Death Detection kit, AP; Boehringer Mannheim GmbH, Mannheim, Germany). Samples were incubated with 50 μL of reaction mixture in a humidified chamber at 37°C for 90 minutes as described previously [15]. The percentage of apoptotic cells was determined by counting at least 1,000 cells from 10 to 20 high-power fields (×200) under both phase-contrast and fluorescent microscopy.

Cell Senescence Assay

Senescence-associated expression of β-galactosidase activity [16] was done with a Senescence Detection kit (BioVision, Mountain View, CA) on fixed cells treated with or without Oridonin (12.5 and 25 μmol/L). The development of cytoplasmic blue was detected and photographed using a Nikon (Nikon Instruments, Inc., Lewisville, TX) inverted microscope equipped with a color CCD camera.

RNA Extraction and Semi-quantitative RT-PCR

Total RNA was extracted from cell cultures using TRI REAGENT (Molecular Research Center, Inc., OH) according to the manufacturer's protocol. The mRNA levels of the genes analyzed were measured by RT-PCR amplification. Sequences for mRNAs from the nucleotide data bank (National Center for Biotechnology Information) were used to design primer pairs for RT-PCR reactions (Primer Express, Applied Biosystems, CA). The following specific oligonucleotide primers were used respectively for p16 (p16-F: 5'-CAC GGC CGC GGC CCG GGG TC -3' and p16-R: 5'-GGC CCG GTG CAG CAC CAC CA -3' ), p21(p21-F: 5'-AGG CGC CAT GTC AGA ACC GGC TGG -3' and p21-R: 5'-GGA AGG TAG AGC TTG GGC AGG C-3' ), p27 (p27-F: 5'-ATG TCA AAC GTG CGA GTG TCT AAC -3' and p27-R: 5'-TTA CGT TTG ACG TCT TCT GAG GCC A-3' ), c-myc (c-myc-F: 5'-ATT CTC TGC TCT CCT CGA -3' and c-myc-R: 5'-TCT TGG CAG CAG GAT AGT -3' ) with GAPDH as internal control (GAPDH-F: 5'-TCC CAT CAC CAT CTT CCA G-3' and GAPDH-R:5'-ATG AGT CCT TCC ACG ATA CC-3';). PCR cycles were adjusted to have linear amplification for all the targets. Each RT-PCR reaction was repeated at least three times. A semiquantitative analysis of mRNA levels was carried out by the ''GEL DOC UV SYSTEM'' (Biorad Company, CA).

Western-blot analysis

Attached cells were collected by scraping them off into a lysis buffer, and the detached cells in the supernatant were collected by centrifugation before resuspension in the lysis buffer. Protein concentration was determined by the bicinchoninic acid (BCA) method according to the manufacturer's (Pierce, Rockford, IL, U.S.A.) instructions after trichloroacetic acid precipitation. The protein lysates were mixed with equal volume of Laemmli buffer (62.5 mM Tris-HCl pH 6.8, 2% SDS, 50 mM DTT, 10% glycerol, 0.01% bromophenol blue), boiled for 3 min at 100°C, and then resolved by SDS-PAGE on a 10 to 12% gel using a mini gel apparatus (Bio-Rad). Bromophenol Blue (0.01%) was added to the samples before an equal amount of proteins was loaded in each lane for electrophoresis and blotting. The PVDF membrane was incubated with a primary antibody against cdc2, cdc25c and cyclinB (Santa Cruz Biotechnology), p16 (PharMingen, San Diego, CA, U.S.A.), p21 (PharMingen, San Diego, CA, U.S.A.), p27 (PharMingen, San Diego, CA, U.S.A.), c-Myc (N-262; Santa Cruz Biotechnology, Santa Cruz, CA, U.S.A.), Acetylated histone H3 (AcH3) and H4 (AcH4), phospho-Histone H3 (Ser10) (Upstate Biotechnology, Lake Placid, NY), histone 3, histone 4 and GAPDH (Santa Cruz Biotechnology) for 2 h. Signals were detected using a horseradish peroxidase-conjugated secondary antibody and an enhanced chemiluminescence detection kit (ECL; Amersham Biosciences, Pittsburgh, PA) [17] and were quantitated by an Eagle Eye II Image System with installed density-analysis software (Stratagene, La Jolla, CA, U.S.A.).

Immunofluorescent Staining

Cells were fixed with 4% paraformaldehyde in PBS for 15 min at room temperature for anti-acetyl histone H4 (06-598; Upstate Biotechnology) and anti-acetyl histone H3 (06-599; Upstate Biotechnology) staining. Cells were permeabilized with 0.2% Triton X-100 (EM Science, Gibbstown, NJ) in PBS for 10 min at room temperature. FITC-labeled secondary antibody (F-0382; Sigma) were applied at the concentration of 1:500. Images were taken with Nikon E800 scope. Senescence-associated heterochromatin staining was conducted as described [18].

CFE in Soft Agar

Tumor cells were resuspended in DMEM with 0.3% agar and plated in 24-well plates at 2,000 per well on top of a 0.5 mL precast semisolid 1% agar underlayer following treatment with Oridonin ( 0, 6.25, 12.5, 25, 50 or 100 μmol/L) for 2 weeks as described previously [15]. The CFE was defined as the percentage of plated cells that formed colonies relative to an untreated control.

Murine model and oridonin treatment

Five-week-old pathogen-free athymic nude mice were purchased from Experimental Animal Centre of SIBS. (Shanghai, PR China). BALB/C nude mice were bred and maintained in a specific pathogen-free environment. Mice were allowed free access to mice standard food pellets and tapwater. Twice a week cages were cleaned and water changed. Temperature was controlled at 21°C ± 2°C. The light was on a 12 hours light-12 hour dark cycle, with light on at 8 am. Xenograft model in nude mice was established by subcutaneous inoculation of 1 × 106 SW1116 cells into the right flank. The nude mice received oridonin treatment (6.25, 12.5 or 25 mg/kg per day) when tumor was measurable. Caliper measurements of the longest perpendicular tumor diameters were performed every day to estimate the tumor volume, using the following formula: 4π/3 × (width/2)2 × (length/2), representing the 3-dimensional volume of an ellipse [19]. Animals were killed when their tumors reached 2 cm or when the mice became moribund. TUNEL assay was performed to detect in situ apoptosis on tissue section using a DeadEnd Colorimetric TUNEL System (Promega) according to the manufacturer's instructions. Senescence-associated expression of β-galactosidase activity [20] was detected with a Senescence Detection kit (BioVision, Mountain View, CA) in situ senescence on tissue section. Animal related experiments were performed according to the Guide for the Care and Use of Laboratory Animals (NIH Publications No. 80-23, revised 1996) and approved by the committee for human treatment of animals at Shanghai Jiao Tong University School of Medicine.

Statistical Analysis

The effects of oridonin on cell proliferation, CFE, cell cycle arrest, apoptosis, and xenograft growth in SCID mice were analyzed with two-way ANOVA and presented as the mean ± SD.

Results

Oridonin suppresses colorectal cell proliferation

To investigate the possible effect of oridonin on the proliferation of colorectal cancer cells, three colorectal cancer cell lines HCT116, HT29, SW1116 were used. As shown in Figure 1, oridonin could inhibit proliferation of the three colorectal cancer cells in a time- and dose-dependent manner. These tumor cells showed different sensitivity to the oridonin treatment. It seemed that HT29 cells are more sensitive to oridonin treatment than HCT116 and SW1116 cells. The growth of HT29 cells was greatly inhibited by oridonin at 6.25 μM for 24 hours, which become more obvious on day 3. However, 12.5 μM oridonin was needed to obtain 50% inhibition in HCT1116 and SW1116 cells on day 3. At 25 μM, growth of all three colorectal cancer cell lines was completely inhibited.

Oridonin induces cell cycle arrest and augments apoptosis

We next examined the effects of oridonin on cell cycle distribution and apoptosis. HCT116 and SW1116 cells were treated with oridonin at low dose (12.5 μM for HCT116 or 25 μM for SW1116) or at higher dose (25 μM for HCT116 or 50 μM for SW1116) for 24 hours. Low dose oridonin causes obvious G2/M arrest in HCT116 and SW1116 cells. As shown in Figure 2A and 2B, the population of G2/M cells increased from 23.5% and 18.2% to 44.1% and 39.6%, respectively in HCT116 and SW1116 cells. Further, the G2/M related protein cyclin B, cdc2, cdc25c and phosphorylated histone H3 were detected in HCT116 and SW1116 cells (Figure 2C and 2E). Oridonin treatment for 12, 24 h causes down-regulation of all these proteins, indicating that oridonin induces G2 arrest in these cells.
While at higher concentration (25 μM for HCT116 or 50 μM for SW1116), a marked increase of subdiploid peak was observed, indicating the increase of apoptotic cells. This apoptosis inducing effect of oridonin was further confirmed by the terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay (Figure 2D and 2F). These results suggested cell cycle arrest and apoptosis induction involved in oridonin induced cell proliferation inhibition.

Oridonin induces cellular senescence

Cellular senescence has been identified as one of the mechanisms mediating the anticancer effects of chemotherapies [16]. One of the morphologic changes that were frequently observed in the oridonin-treated cells is the flattening of the adherent cells with increased granularity, which is a typical morphologic change associated with cellular senescence (Figure 3A and 3C). By examining senescence-associated expression of β-galactosidase activity, we confirmed that cellular senescence was indeed induced in those flattened HCT116 and SW1116 cells treated by oridonin (12.5 or 25 μM) for three days (Figure 3B and 3D), indicating that senescence inducing also contribute to the proliferation inhibition effect of oridonin.

Oridonin activates p21, p16 and p27 expression and down-regulates the expression of oncogene c-myc

To perform detailed temporal analysis of gene expression alteration, SW1116 cells were treated with 25 μM oridonin for 0, 24, 48 and 72 hours. Then, the mRNA and protein levels of p16, p21, p27 were examined. As shown in Figure 4A and 4C, after treated oridonin for 24 hours, the mRNA and protein level of p21 but not p16 and p27 were significantly increased. Treatment with oridonin for 48 and 72 hours resulted in increase of the p16, p21 and p27. A series of studies have documented that c-myc regulates a wide range of genes involved in cell proliferation, differentiation, and apoptosis [21]. We also detected the expression of c-myc in oridonin treated SW1116 cells. Treatment with oridonin (25 μM) dramatically reduced c-myc mRNA transcription and c-myc protein levels as early as 24 hours. These data suggested that suppression of c-myc and up-regulation of p16, p21 and p27 correlated with oridonin responsiveness of colorectal cancer cells.

Oridonin induces global changes in chromatin structure

Cellular senescence is known to be associated with changes in chromatin structure [18, 22]. Therefore, we examined global chromatin modifications associated with cellular senescence in SW1116 cell lines treated with oridonin (25 μM) for 3 days in vitro. As shown in Figure 5A, after 72 h of oridonin treatment, heterochromatin formation were observed with DAPI staining, and a marked global increase of total acetylation of histone H3 and histone H4 were detected by immunofluorescence. Time course analysis showed that protein levels of acetylation of histone H3 and histone H4 gradually increased, especially for histone H4 (Figure 5B and 5C). Hence, oridonin treatment resulted in global changes of histone modifications that have been associated with cellular senescence.

Oridonin Suppresses CFE

To examine the suppressive effects of oridonin on CFE, SW1116 cells were treated with 0, 6.25, 12.5, 25, 50, 100 μM oridonin for 14 days. Our results showed that oridonin exerted dose-dependant suppression of CFE in SW1116 cells (P < 0.01; Figure 6A). Treatment with 6.25 μM oridonin for 2 week causes over 50% inhibition of CFE. With the increase in drug concentration, more significant suppressive effects were observed. At higher concentration (50 and 100 μM), the formation of colony was completely inhibited (Figure 6A).

Oridonin Suppresses the Growth of SW1116 Xenografts in BALB/C nude mice

We next further assessed the in vivo anti-colorectal cancer effects of oridonin. BALB/C nude mice xenografts were treated with oridonin by daily i.p injection at 6.25 or 12.5 or 25 mg/kg for up to 28 days. Such treatment resulted in significant suppression of xenograft growth , compared with the PBS or DMSO (1% in PBS) treated group (P < 0.05; Figure 6B and 6C). In the low-dose (6.25 mg/kg) group, significant tumor growth inhibition effect was observed until 21 days, while in the middle dose (12.5 mg/kg) group, significant tumor growth inhibition effect was observed on day 14. More interesting, the high dose (25 mg/kg) administration almost completely inhibited tumor growth at the onset phase. It is worth noting that no obvious changes in body weight were observed in the oridonin treated groups, compared with the PBS or DMSO (1% in PBS) treated groups (Figure 6D). In H & E stained tumor section, sparse tumor cells and areas of necrosis foci can be seen in oridonin treated mice, while massive tumor cells were observed in PBS or DMSO (1% in PBS) treated mice (Figure 7A). TUNEL assay (Figure 7B) and Senescence-associated-β-galactosidase (SA-β-Gal) staining (Figure 7C) were also performed to detect the degree of apoptosis and senescence. The results showed that oridonin induces apoptosis and senescence of colorectal cells in vivo in a dose dependent manner.

Discussion

Chemotherapy remains the primary treatment for systemic malignancies. It is now clear that drug-induced damage is not invariably lethal, but can instead initiate a series of post-damage responses including apoptosis, mitotic catastrophe, and cellular senescence [23, 24]. Therefore, the integrity of these damage responses might also influence treatment sensitivity. In the current study, we showed that the potent anti-cancer activities of oridonin in colorectal cancer are correlated with induction of apoptosis, cell-cycle arrest and cellular senescence.
Oridonin treatment resulted in significant growth arrest in colorectal cancer cells and this in vitro effect was time and concentration dependent. This finding is in agreement with our previous results with oridonin in acute promyelocytic leukemia cells and others [14]. More importantly, we further showed that oridonin possesses strong inhibitory activities on tumorigenicity of colorectal cancer cells. Oridonin at 6.25 μM could inhibit the colony-forming by 50%. With the increase in drug concentration, the formation of colony was almost completely inhibited. In consistent with these results in vitro, treatment with 6.25 mg/kg oridonin in SW1116 xenografts BALB/C nude mice for 4 weeks was able to significantly decrease the growth of xenografts. These results provided strong evidence to support the notion that oridonin has strong activity against colorectal cancer.
Apoptosis is a well-characterized post-damage program and diverse anticancer agents can induce apoptosis through common pathways [24]. It has been reported previously that the antitumor effect of oridonin was due to its apoptosis induction activity [14, 2528]. In agreement with these reports, our data showed oridonin could induce apoptosis of colorectal cancer cells in vitro and in vivo. In addition to apoptosis induction, we found that oridonin could induce senescence of colorectal cancer cells. In fibroblasts and epithelial cells, senescence is controlled by the p53 and Rb tumor suppressor pathways, although the contribution of each pathway to the program depends on species and cell type [20]. Here, oridonin-induced arrest was accompanied by substantial increases in p21, p16 (an Rb regulator linked exclusively to senescence) and SA-β-gal activity. It is interesting to note that oridonin-induced senescence associated with G2 arrest. In most cases, the senescence is associated with G1/G0 arrest. However, many reports also showed that G2/M arrest is associated with senescence [2932]. For example, some drugs including DNA damage agent Adriamycin, DNA methylating agent Temozolomide, HDAC inhibitor sodium butyrate could induce G2/M arrest and senescence-like phenotype via p21 induction. Similar to these results, oridonin could also up-regulate the expression of p21, p27 and p16, which may contribute to the observed senescent phenotype.
The expression of cyclin-dependent kinase inhibitor p21 and p27 has been implicated in chemotherapy-induced cell cycle arrest in numerous human cancers [33, 34]. In this study, we confirmed that oridonin is capable of activating p21 and p27 gene in colorectal cancer cells. Overexpression of c-myc had been frequently detected in colorectal cancer and is associated with shorter survival and tumor anaplasia [3537]. C-myc has also been reported to promote cell cycle reentry and proliferation through repression of p21 and p27 expression [38]. Therefore, the suppression of c-myc expression by oridonin may render substantial therapeutic benefits in colorectal cancer patients by inhibiting the driving activities of c-myc in cell proliferation and cell cycle progression. In line with previous reports, our data showed oridonin- induced p21 and p27 and down-regulated c-myc in vitro.
Epigenetic changes are tightly related to neoplastic transformation in colorectal cancers. Histone modifications, recently recognized as a 'histone code' that affects chromatin structure and gene expression also play an important role in the establishment of gene silencing during tumorigenesis. Alterations in histone modifications appear to be primary mediators of epigenetic inheritance in cancer cells [3941]. The potential reversibility of epigenetic states in the tumor cell is an attractive target for cancer therapy [42]. Our data showed oridonin induced histone (H3 and H4) hyperacetylation in vitro. We also found that the levels of accumulated AcH3 and AcH4 correlated with the degree of in vitro growth suppression in the oridonin-sensitive colorectal cancer cell lines, suggesting that the anti-colorectal cancer effects of oridonin were at least partly mediated through histone H3 and H4 hyperacetylation.

Conclusions

In summary, we showed that oridonin possesses potent in vitro and in vivo anti-colorectal cancer activities by suppressing cell proliferation, promoting apoptosis, inducing cell cycle arrest and cellular senescence. These results may lay the groundwork for further studies using specific genetically engineered models to establish the causal relationship between oridonin anti-tumor activity and specific genetic pathways and to identify molecular markers that will predict drug responsiveness and guide the development of future clinical therapies.

Acknowledgements

This work was supported in part by the National Natural Science Foundation of China (81070433, 91013008), Science and Technology Committee of Shanghai (08JC1413400), Science and Technology Committee of Baoshan District (08-E-13), Renji hospital and college of basic medicine cooperation foundation (ZD0704) and NO.3 People's Hospital affiliated to Shanghai Jiao-Tong University School of Medicine (syz07-04).
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

F-HG and X-HH carried out the data collection and data analysis, and drafted the manuscript. WL, HL and Y-JZ participated in the design of the study, performed the statistical analysis and participated in the interpretation of data. Z-YG, M-HX and S-TW participated in the design of the study and the acquisition and interpretation of data. BJ, FL, Y-ZZ, YF and F-YC were involved in the interpretation of data and critically revised the manuscript. Y-LW conceived the study, participated in its design and coordination, and helped in statistical analysis and drafting of the manuscript. All authors have read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Boursi B, Arber N: Current and future clinical strategies in colon cancer prevention and the emerging role of chemoprevention. Curr Pharm Des. 2007, 13 (22): 2274-2282. 10.2174/138161207781368783.CrossRefPubMed Boursi B, Arber N: Current and future clinical strategies in colon cancer prevention and the emerging role of chemoprevention. Curr Pharm Des. 2007, 13 (22): 2274-2282. 10.2174/138161207781368783.CrossRefPubMed
2.
Zurück zum Zitat Mayo SC, Pawlik TM: Current management of colorectal hepatic metastasis. Expert Rev Gastroenterol Hepatol. 2009, 3 (2): 131-144. 10.1586/egh.09.8.CrossRefPubMed Mayo SC, Pawlik TM: Current management of colorectal hepatic metastasis. Expert Rev Gastroenterol Hepatol. 2009, 3 (2): 131-144. 10.1586/egh.09.8.CrossRefPubMed
3.
Zurück zum Zitat Hebbar M, Ychou M, Ducreux M: Current place of high-dose irinotecan chemotherapy in patients with metastatic colorectal cancer. J Cancer Res Clin Oncol. 2009, 135 (6): 749-752. 10.1007/s00432-009-0580-x.CrossRefPubMed Hebbar M, Ychou M, Ducreux M: Current place of high-dose irinotecan chemotherapy in patients with metastatic colorectal cancer. J Cancer Res Clin Oncol. 2009, 135 (6): 749-752. 10.1007/s00432-009-0580-x.CrossRefPubMed
4.
Zurück zum Zitat Berri RN, Abdalla EK: Curable metastatic colorectal cancer: recommended paradigms. Curr Oncol Rep. 2009, 11 (3): 200-208. 10.1007/s11912-009-0029-z.CrossRefPubMed Berri RN, Abdalla EK: Curable metastatic colorectal cancer: recommended paradigms. Curr Oncol Rep. 2009, 11 (3): 200-208. 10.1007/s11912-009-0029-z.CrossRefPubMed
5.
Zurück zum Zitat Mihaylova Z, Raynov J: Neoadjuvant chemotherapy and targeted therapy in patients with liver metastases from colorectal cancer; medical oncologist's point of view. J BUON. 2008, 13 (3): 323-331.PubMed Mihaylova Z, Raynov J: Neoadjuvant chemotherapy and targeted therapy in patients with liver metastases from colorectal cancer; medical oncologist's point of view. J BUON. 2008, 13 (3): 323-331.PubMed
7.
Zurück zum Zitat Salminen A, Lehtonen M, Suuronen T, Kaarniranta K, Huuskonen J: Terpenoids: natural inhibitors of NF-kappaB signaling with anti-inflammatory and anticancer potential. Cell Mol Life Sci. 2008, 65 (19): 2979-2999. 10.1007/s00018-008-8103-5.CrossRefPubMed Salminen A, Lehtonen M, Suuronen T, Kaarniranta K, Huuskonen J: Terpenoids: natural inhibitors of NF-kappaB signaling with anti-inflammatory and anticancer potential. Cell Mol Life Sci. 2008, 65 (19): 2979-2999. 10.1007/s00018-008-8103-5.CrossRefPubMed
8.
Zurück zum Zitat Wang X, Wei Y, Yuan S, Liu G, Lu Y, Zhang J, Wang W: Potential anticancer activity of tanshinone IIA against human breast cancer. Int J Cancer. 2005, 116 (5): 799-807. 10.1002/ijc.20880.CrossRefPubMed Wang X, Wei Y, Yuan S, Liu G, Lu Y, Zhang J, Wang W: Potential anticancer activity of tanshinone IIA against human breast cancer. Int J Cancer. 2005, 116 (5): 799-807. 10.1002/ijc.20880.CrossRefPubMed
9.
Zurück zum Zitat Nizamutdinova IT, Lee GW, Son KH, Jeon SJ, Kang SS, Kim YS, Lee JH, Seo HG, Chang KC, Kim HJ: Tanshinone I effectively induces apoptosis in estrogen receptor-positive (MCF-7) and estrogen receptor-negative (MDA-MB-231) breast cancer cells. Int J Oncol. 2008, 33 (3): 485-491.PubMed Nizamutdinova IT, Lee GW, Son KH, Jeon SJ, Kang SS, Kim YS, Lee JH, Seo HG, Chang KC, Kim HJ: Tanshinone I effectively induces apoptosis in estrogen receptor-positive (MCF-7) and estrogen receptor-negative (MDA-MB-231) breast cancer cells. Int J Oncol. 2008, 33 (3): 485-491.PubMed
10.
Zurück zum Zitat Sethi G, Ahn KS, Pandey MK, Aggarwal BB: Celastrol, a novel triterpene, potentiates TNF-induced apoptosis and suppresses invasion of tumor cells by inhibiting NF-kappaB-regulated gene products and TAK1-mediated NF-kappaB activation. Blood. 2007, 109 (7): 2727-2735.PubMed Sethi G, Ahn KS, Pandey MK, Aggarwal BB: Celastrol, a novel triterpene, potentiates TNF-induced apoptosis and suppresses invasion of tumor cells by inhibiting NF-kappaB-regulated gene products and TAK1-mediated NF-kappaB activation. Blood. 2007, 109 (7): 2727-2735.PubMed
11.
Zurück zum Zitat Hsieh TC, Wijeratne EK, Liang JY, Gunatilaka AL, Wu JM: Differential control of growth, cell cycle progression, and expression of NF-kappaB in human breast cancer cells MCF-7, MCF-10A, and MDA-MB-231 by ponicidin and oridonin, diterpenoids from the chinese herb Rabdosia rubescens. Biochem Biophys Res Commun. 2005, 337 (1): 224-231. 10.1016/j.bbrc.2005.09.040.CrossRefPubMed Hsieh TC, Wijeratne EK, Liang JY, Gunatilaka AL, Wu JM: Differential control of growth, cell cycle progression, and expression of NF-kappaB in human breast cancer cells MCF-7, MCF-10A, and MDA-MB-231 by ponicidin and oridonin, diterpenoids from the chinese herb Rabdosia rubescens. Biochem Biophys Res Commun. 2005, 337 (1): 224-231. 10.1016/j.bbrc.2005.09.040.CrossRefPubMed
12.
Zurück zum Zitat Ikezoe T, Yang Y, Bandobashi K, Saito T, Takemoto S, Machida H, Togitani K, Koeffler HP, Taguchi H: Oridonin, a diterpenoid purified from Rabdosia rubescens, inhibits the proliferation of cells from lymphoid malignancies in association with blockade of the NF-kappa B signal pathways. Mol Cancer Ther. 2005, 4 (4): 578-586. 10.1158/1535-7163.MCT-04-0277.CrossRefPubMed Ikezoe T, Yang Y, Bandobashi K, Saito T, Takemoto S, Machida H, Togitani K, Koeffler HP, Taguchi H: Oridonin, a diterpenoid purified from Rabdosia rubescens, inhibits the proliferation of cells from lymphoid malignancies in association with blockade of the NF-kappa B signal pathways. Mol Cancer Ther. 2005, 4 (4): 578-586. 10.1158/1535-7163.MCT-04-0277.CrossRefPubMed
13.
Zurück zum Zitat Liu J, Yang F, Zhang Y, Li J: Studies on the cell-immunosuppressive mechanism of Oridonin from Isodon serra. Int Immunopharmacol. 2007, 7 (7): 945-954. 10.1016/j.intimp.2007.03.001.CrossRefPubMed Liu J, Yang F, Zhang Y, Li J: Studies on the cell-immunosuppressive mechanism of Oridonin from Isodon serra. Int Immunopharmacol. 2007, 7 (7): 945-954. 10.1016/j.intimp.2007.03.001.CrossRefPubMed
14.
Zurück zum Zitat Gao F, Tang Q, Yang P, Fang Y, Li W, Wu Y: Apoptosis inducing and differentiation enhancement effect of oridonin on the all-trans-retinoic acid-sensitive and -resistant acute promyelocytic leukemia cells. Int J Lab Hematol. 2010, 32 (1 Pt 1): e114-122. 10.1111/j.1751-553X.2009.01147.x.CrossRefPubMed Gao F, Tang Q, Yang P, Fang Y, Li W, Wu Y: Apoptosis inducing and differentiation enhancement effect of oridonin on the all-trans-retinoic acid-sensitive and -resistant acute promyelocytic leukemia cells. Int J Lab Hematol. 2010, 32 (1 Pt 1): e114-122. 10.1111/j.1751-553X.2009.01147.x.CrossRefPubMed
15.
Zurück zum Zitat Li XN, Parikh S, Shu Q, Jung HL, Chow CW, Perlaky L, Leung HC, Su J, Blaney S, Lau CC: Phenylbutyrate and phenylacetate induce differentiation and inhibit proliferation of human medulloblastoma cells. Clin Cancer Res. 2004, 10 (3): 1150-1159. 10.1158/1078-0432.CCR-0747-3.CrossRefPubMed Li XN, Parikh S, Shu Q, Jung HL, Chow CW, Perlaky L, Leung HC, Su J, Blaney S, Lau CC: Phenylbutyrate and phenylacetate induce differentiation and inhibit proliferation of human medulloblastoma cells. Clin Cancer Res. 2004, 10 (3): 1150-1159. 10.1158/1078-0432.CCR-0747-3.CrossRefPubMed
16.
Zurück zum Zitat Schmitt CA: Senescence, apoptosis and therapy--cutting the lifelines of cancer. Nat Rev Cancer. 2003, 3 (4): 286-295. 10.1038/nrc1044.CrossRefPubMed Schmitt CA: Senescence, apoptosis and therapy--cutting the lifelines of cancer. Nat Rev Cancer. 2003, 3 (4): 286-295. 10.1038/nrc1044.CrossRefPubMed
17.
Zurück zum Zitat Gao FH, Wang Q, Wu YL, Li X, Zhao KW, Chen GQ: c-Jun N-terminal kinase mediates AML1-ETO protein-induced connexin-43 expression. Biochem Biophys Res Commun. 2007, 356 (2): 505-511. 10.1016/j.bbrc.2007.03.009.CrossRefPubMed Gao FH, Wang Q, Wu YL, Li X, Zhao KW, Chen GQ: c-Jun N-terminal kinase mediates AML1-ETO protein-induced connexin-43 expression. Biochem Biophys Res Commun. 2007, 356 (2): 505-511. 10.1016/j.bbrc.2007.03.009.CrossRefPubMed
18.
Zurück zum Zitat Narita M, Nunez S, Heard E, Lin AW, Hearn SA, Spector DL, Hannon GJ, Lowe SW: Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell. 2003, 113 (6): 703-716. 10.1016/S0092-8674(03)00401-X.CrossRefPubMed Narita M, Nunez S, Heard E, Lin AW, Hearn SA, Spector DL, Hannon GJ, Lowe SW: Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell. 2003, 113 (6): 703-716. 10.1016/S0092-8674(03)00401-X.CrossRefPubMed
19.
Zurück zum Zitat LeBlanc R, Catley LP, Hideshima T, Lentzsch S, Mitsiades CS, Mitsiades N, Neuberg D, Goloubeva O, Pien CS, Adams J, et al: Proteasome inhibitor PS-341 inhibits human myeloma cell growth in vivo and prolongs survival in a murine model. Cancer Res. 2002, 62 (17): 4996-5000.PubMed LeBlanc R, Catley LP, Hideshima T, Lentzsch S, Mitsiades CS, Mitsiades N, Neuberg D, Goloubeva O, Pien CS, Adams J, et al: Proteasome inhibitor PS-341 inhibits human myeloma cell growth in vivo and prolongs survival in a murine model. Cancer Res. 2002, 62 (17): 4996-5000.PubMed
20.
Zurück zum Zitat Schmitt CA, Fridman JS, Yang M, Lee S, Baranov E, Hoffman RM, Lowe SW: A senescence program controlled by p53 and p16INK4a contributes to the outcome of cancer therapy. Cell. 2002, 109 (3): 335-346. 10.1016/S0092-8674(02)00734-1.CrossRefPubMed Schmitt CA, Fridman JS, Yang M, Lee S, Baranov E, Hoffman RM, Lowe SW: A senescence program controlled by p53 and p16INK4a contributes to the outcome of cancer therapy. Cell. 2002, 109 (3): 335-346. 10.1016/S0092-8674(02)00734-1.CrossRefPubMed
21.
Zurück zum Zitat Wong SC, Chan JK, Lee KC, Hsiao WL: Differential expression of p16/p21/p27 and cyclin D1/D3, and their relationships to cell proliferation, apoptosis, and tumour progression in invasive ductal carcinoma of the breast. J Pathol. 2001, 194 (1): 35-42. 10.1002/path.838.CrossRefPubMed Wong SC, Chan JK, Lee KC, Hsiao WL: Differential expression of p16/p21/p27 and cyclin D1/D3, and their relationships to cell proliferation, apoptosis, and tumour progression in invasive ductal carcinoma of the breast. J Pathol. 2001, 194 (1): 35-42. 10.1002/path.838.CrossRefPubMed
22.
Zurück zum Zitat Zhang R, Poustovoitov MV, Ye X, Santos HA, Chen W, Daganzo SM, Erzberger JP, Serebriiskii IG, Canutescu AA, Dunbrack RL, et al: Formation of MacroH2A-containing senescence-associated heterochromatin foci and senescence driven by ASF1a and HIRA. Dev Cell. 2005, 8 (1): 19-30. 10.1016/j.devcel.2004.10.019.CrossRefPubMed Zhang R, Poustovoitov MV, Ye X, Santos HA, Chen W, Daganzo SM, Erzberger JP, Serebriiskii IG, Canutescu AA, Dunbrack RL, et al: Formation of MacroH2A-containing senescence-associated heterochromatin foci and senescence driven by ASF1a and HIRA. Dev Cell. 2005, 8 (1): 19-30. 10.1016/j.devcel.2004.10.019.CrossRefPubMed
23.
Zurück zum Zitat Chang BD, Broude EV, Dokmanovic M, Zhu H, Ruth A, Xuan Y, Kandel ES, Lausch E, Christov K, Roninson IB: A senescence-like phenotype distinguishes tumor cells that undergo terminal proliferation arrest after exposure to anticancer agents. Cancer Res. 1999, 59 (15): 3761-3767.PubMed Chang BD, Broude EV, Dokmanovic M, Zhu H, Ruth A, Xuan Y, Kandel ES, Lausch E, Christov K, Roninson IB: A senescence-like phenotype distinguishes tumor cells that undergo terminal proliferation arrest after exposure to anticancer agents. Cancer Res. 1999, 59 (15): 3761-3767.PubMed
24.
Zurück zum Zitat Johnstone RW, Ruefli AA, Lowe SW: Apoptosis: a link between cancer genetics and chemotherapy. Cell. 2002, 108 (2): 153-164. 10.1016/S0092-8674(02)00625-6.CrossRefPubMed Johnstone RW, Ruefli AA, Lowe SW: Apoptosis: a link between cancer genetics and chemotherapy. Cell. 2002, 108 (2): 153-164. 10.1016/S0092-8674(02)00625-6.CrossRefPubMed
25.
Zurück zum Zitat Huang J, Wu L, Tashiro S, Onodera S, Ikejima T: Reactive oxygen species mediate oridonin-induced HepG2 apoptosis through p53, MAPK, and mitochondrial signaling pathways. J Pharmacol Sci. 2008, 107 (4): 370-379. 10.1254/jphs.08044FP.CrossRefPubMed Huang J, Wu L, Tashiro S, Onodera S, Ikejima T: Reactive oxygen species mediate oridonin-induced HepG2 apoptosis through p53, MAPK, and mitochondrial signaling pathways. J Pharmacol Sci. 2008, 107 (4): 370-379. 10.1254/jphs.08044FP.CrossRefPubMed
26.
Zurück zum Zitat Jin S, Shen JN, Wang J, Huang G, Zhou JG: Oridonin induced apoptosis through Akt and MAPKs signaling pathways in human osteosarcoma cells. Cancer Biol Ther. 2007, 6 (2): 261-268. 10.4161/cbt.6.2.3621.CrossRefPubMed Jin S, Shen JN, Wang J, Huang G, Zhou JG: Oridonin induced apoptosis through Akt and MAPKs signaling pathways in human osteosarcoma cells. Cancer Biol Ther. 2007, 6 (2): 261-268. 10.4161/cbt.6.2.3621.CrossRefPubMed
27.
Zurück zum Zitat Zhou GB, Kang H, Wang L, Gao L, Liu P, Xie J, Zhang FX, Weng XQ, Shen ZX, Chen J, et al: Oridonin, a diterpenoid extracted from medicinal herbs, targets AML1-ETO fusion protein and shows potent antitumor activity with low adverse effects on t(8;21) leukemia in vitro and in vivo. Blood. 2007, 109 (8): 3441-3450. 10.1182/blood-2006-06-032250.CrossRefPubMedPubMedCentral Zhou GB, Kang H, Wang L, Gao L, Liu P, Xie J, Zhang FX, Weng XQ, Shen ZX, Chen J, et al: Oridonin, a diterpenoid extracted from medicinal herbs, targets AML1-ETO fusion protein and shows potent antitumor activity with low adverse effects on t(8;21) leukemia in vitro and in vivo. Blood. 2007, 109 (8): 3441-3450. 10.1182/blood-2006-06-032250.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Cheng Y, Qiu F, Ye YC, Tashiro S, Onodera S, Ikejima T: Oridonin induces G2/M arrest and apoptosis via activating ERK-p53 apoptotic pathway and inhibiting PTK-Ras-Raf-JNK survival pathway in murine fibrosarcoma L929 cells. Arch Biochem Biophys. 2009, 490 (1): 70-75. 10.1016/j.abb.2009.08.011.CrossRefPubMed Cheng Y, Qiu F, Ye YC, Tashiro S, Onodera S, Ikejima T: Oridonin induces G2/M arrest and apoptosis via activating ERK-p53 apoptotic pathway and inhibiting PTK-Ras-Raf-JNK survival pathway in murine fibrosarcoma L929 cells. Arch Biochem Biophys. 2009, 490 (1): 70-75. 10.1016/j.abb.2009.08.011.CrossRefPubMed
29.
Zurück zum Zitat Hirose Y, Berger MS, Pieper RO: p53 effects both the duration of G2/M arrest and the fate of temozolomide-treated human glioblastoma cells. Cancer Res. 2001, 61 (5): 1957-1963.PubMed Hirose Y, Berger MS, Pieper RO: p53 effects both the duration of G2/M arrest and the fate of temozolomide-treated human glioblastoma cells. Cancer Res. 2001, 61 (5): 1957-1963.PubMed
30.
Zurück zum Zitat Shay JW, Roninson IB: Hallmarks of senescence in carcinogenesis and cancer therapy. Oncogene. 2004, 23 (16): 2919-2933. 10.1038/sj.onc.1207518.CrossRefPubMed Shay JW, Roninson IB: Hallmarks of senescence in carcinogenesis and cancer therapy. Oncogene. 2004, 23 (16): 2919-2933. 10.1038/sj.onc.1207518.CrossRefPubMed
31.
Zurück zum Zitat Terao Y, Nishida J, Horiuchi S, Rong F, Ueoka Y, Matsuda T, Kato H, Furugen Y, Yoshida K, Kato K, et al: Sodium butyrate induces growth arrest and senescence-like phenotypes in gynecologic cancer cells. Int J Cancer. 2001, 94 (2): 257-267. 10.1002/ijc.1448.CrossRefPubMed Terao Y, Nishida J, Horiuchi S, Rong F, Ueoka Y, Matsuda T, Kato H, Furugen Y, Yoshida K, Kato K, et al: Sodium butyrate induces growth arrest and senescence-like phenotypes in gynecologic cancer cells. Int J Cancer. 2001, 94 (2): 257-267. 10.1002/ijc.1448.CrossRefPubMed
32.
Zurück zum Zitat Vigneron A, Roninson IB, Gamelin E, Coqueret O: Src inhibits adriamycin-induced senescence and G2 checkpoint arrest by blocking the induction of p21waf1. Cancer Res. 2005, 65 (19): 8927-8935. 10.1158/0008-5472.CAN-05-0461.CrossRefPubMed Vigneron A, Roninson IB, Gamelin E, Coqueret O: Src inhibits adriamycin-induced senescence and G2 checkpoint arrest by blocking the induction of p21waf1. Cancer Res. 2005, 65 (19): 8927-8935. 10.1158/0008-5472.CAN-05-0461.CrossRefPubMed
33.
Zurück zum Zitat Shankar S, Singh G, Srivastava RK: Chemoprevention by resveratrol: molecular mechanisms and therapeutic potential. Front Biosci. 2007, 12: 4839-4854. 10.2741/2432.CrossRefPubMed Shankar S, Singh G, Srivastava RK: Chemoprevention by resveratrol: molecular mechanisms and therapeutic potential. Front Biosci. 2007, 12: 4839-4854. 10.2741/2432.CrossRefPubMed
34.
Zurück zum Zitat Lee JT, Lehmann BD, Terrian DM, Chappell WH, Stivala F, Libra M, Martelli AM, Steelman LS, McCubrey JA: Targeting prostate cancer based on signal transduction and cell cycle pathways. Cell Cycle. 2008, 7 (12): 1745-1762.CrossRefPubMedPubMedCentral Lee JT, Lehmann BD, Terrian DM, Chappell WH, Stivala F, Libra M, Martelli AM, Steelman LS, McCubrey JA: Targeting prostate cancer based on signal transduction and cell cycle pathways. Cell Cycle. 2008, 7 (12): 1745-1762.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Masramon L, Arribas R, Tortola S, Perucho M, Peinado MA: Moderate amplifications of the c-myc gene correlate with molecular and clinicopathological parameters in colorectal cancer. Br J Cancer. 1998, 77 (12): 2349-2356.CrossRefPubMedPubMedCentral Masramon L, Arribas R, Tortola S, Perucho M, Peinado MA: Moderate amplifications of the c-myc gene correlate with molecular and clinicopathological parameters in colorectal cancer. Br J Cancer. 1998, 77 (12): 2349-2356.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Lagerholm S, Dutta S, Nair P: Non-invasive detection of c-myc p64, c-myc p67 and c-erbb-2 in colorectal cancer. Scand J Gastroenterol. 2005, 40 (11): 1343-1350. 10.1080/00365520510023549.CrossRefPubMed Lagerholm S, Dutta S, Nair P: Non-invasive detection of c-myc p64, c-myc p67 and c-erbb-2 in colorectal cancer. Scand J Gastroenterol. 2005, 40 (11): 1343-1350. 10.1080/00365520510023549.CrossRefPubMed
37.
Zurück zum Zitat Matsushita K, Takenouchi T, Shimada H, Tomonaga T, Hayashi H, Shioya A, Komatsu A, Matsubara H, Ochiai T: Strong HLA-DR antigen expression on cancer cells relates to better prognosis of colorectal cancer patients: Possible involvement of c-myc suppression by interferon-gamma in situ. Cancer Sci. 2006, 97 (1): 57-63. 10.1111/j.1349-7006.2006.00137.x.CrossRefPubMed Matsushita K, Takenouchi T, Shimada H, Tomonaga T, Hayashi H, Shioya A, Komatsu A, Matsubara H, Ochiai T: Strong HLA-DR antigen expression on cancer cells relates to better prognosis of colorectal cancer patients: Possible involvement of c-myc suppression by interferon-gamma in situ. Cancer Sci. 2006, 97 (1): 57-63. 10.1111/j.1349-7006.2006.00137.x.CrossRefPubMed
38.
Zurück zum Zitat Nasi S, Ciarapica R, Jucker R, Rosati J, Soucek L: Making decisions through Myc. FEBS Lett. 2001, 490 (3): 153-162. 10.1016/S0014-5793(01)02118-4.CrossRefPubMed Nasi S, Ciarapica R, Jucker R, Rosati J, Soucek L: Making decisions through Myc. FEBS Lett. 2001, 490 (3): 153-162. 10.1016/S0014-5793(01)02118-4.CrossRefPubMed
39.
Zurück zum Zitat Karpinski P, Sasiadek MM, Blin N: Aberrant epigenetic patterns in the etiology of gastrointestinal cancers. J Appl Genet. 2008, 49 (1): 1-10.CrossRefPubMed Karpinski P, Sasiadek MM, Blin N: Aberrant epigenetic patterns in the etiology of gastrointestinal cancers. J Appl Genet. 2008, 49 (1): 1-10.CrossRefPubMed
40.
Zurück zum Zitat Konishi K, Issa JP: Targeting aberrant chromatin structure in colorectal carcinomas. Cancer J. 2007, 13 (1): 49-55. 10.1097/PPO.0b013e31803c72fe.CrossRefPubMed Konishi K, Issa JP: Targeting aberrant chromatin structure in colorectal carcinomas. Cancer J. 2007, 13 (1): 49-55. 10.1097/PPO.0b013e31803c72fe.CrossRefPubMed
41.
Zurück zum Zitat Kondo Y, Issa JP: Epigenetic changes in colorectal cancer. Cancer Metastasis Rev. 2004, 23 (1-2): 29-39. 10.1023/A:1025806911782.CrossRefPubMed Kondo Y, Issa JP: Epigenetic changes in colorectal cancer. Cancer Metastasis Rev. 2004, 23 (1-2): 29-39. 10.1023/A:1025806911782.CrossRefPubMed
42.
Zurück zum Zitat Atadja P: Development of the pan-DAC inhibitor panobinostat (LBH589): successes and challenges. Cancer Lett. 2009, 280 (2): 233-241. 10.1016/j.canlet.2009.02.019.CrossRefPubMed Atadja P: Development of the pan-DAC inhibitor panobinostat (LBH589): successes and challenges. Cancer Lett. 2009, 280 (2): 233-241. 10.1016/j.canlet.2009.02.019.CrossRefPubMed
Metadaten
Titel
Oridonin induces apoptosis and senescence in colorectal cancer cells by increasing histone hyperacetylation and regulation of p16, p21, p27 and c-myc
verfasst von
Feng-Hou Gao
Xiao-Hui Hu
Wei Li
Hua Liu
Yan-Jie Zhang
Zhu-Ying Guo
Mang-Hua Xu
Shi-Ting Wang
Bin Jiang
Feng Liu
Ying-Zheng Zhao
Yong Fang
Fang-Yuan Chen
Ying-Li Wu
Publikationsdatum
01.12.2010
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2010
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-10-610

Weitere Artikel der Ausgabe 1/2010

BMC Cancer 1/2010 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.