The transcription factor c-Myc is important in cell fate decisions and is frequently overexpressed in cancer cells, making it an attractive therapeutic target. Natural compounds are among the current strategies aimed at targeting c-Myc, but their modes of action still need to be characterized. To explore the mechanisms underlying the anticancer activity of a natural diterpenoid, oridonin, we conducted miRNA expression profiling and statistical analyses that strongly suggested that c-Myc was a potential molecular target of oridonin. Furthermore, experimental data showed that oridonin significantly reduced c-Myc protein levels in vitro and in vivo and that this reduction was mediated by the ubiquitin-proteasome system. Fbw7, a component of the ubiquitin-proteasome system and an E3 ubiquitin ligase of c-Myc, was upregulated rapidly in K562 cells and other leukemia and lymphoma cells, resulting in the rapid turnover of c-Myc. In cell lines harboring mutations in the WD domain of Fbw7, the degradation of c-Myc induced by oridonin was attenuated during short-term treatment. GSK-3, an Fbw7 priming kinase, was also activated by oridonin, along with an increase in T58-phosphorylated c-Myc. Furthermore, the knockdown of Fbw7 or the forced expression of stable c-Myc resulted in reduced sensitization to oridonin-induced apoptosis. Our observations help to clarify the anticancer mechanisms of oridonin and shed light on the application of this natural compound as an Fbw7-c-Myc pathway targeting agent in cancer treatment. Mol Cancer Ther; 11(5); 1155–65. ©2012 AACR.

The protein c-Myc is a helix-loop-helix leucine zipper transcription factor that plays essential roles in cell growth, proliferation, and apoptosis. Several lines of evidence support the role of c-Myc as a central transcriptional hub, including (i) previous reports that approximately 15% of the genes in the human genome are regulated by c-Myc (1), (ii) demonstrations that 48 transcription factors are direct targets of c-Myc in a model of human B lymphoid tumor (2), and (iii) the finding that c-Myc transcriptionally regulates the expression of miRNAs (3, 4), a family of regulatory small RNAs that functions by targeting protein-coding genes (5). Given its importance, c-Myc is tightly regulated at different biologic levels, including F-box and WD repeat domain–containing 7 (Fbw7)-mediated posttranslational regulation (6, 7). However, the deregulation of c-Myc has been observed in a wide range of human cancers, particularly leukemias and lymphomas, due to gene amplification, translocation, transactivation, or increased protein stability. Deregulated c-Myc increases genomic instability, blocks differentiation, and is able to induce lymphoid and myeloid neoplasia, making it an attractive target for cancer treatment (8–10).

Various strategies aimed at targeting c-Myc for cancer treatment are under investigation, including the use of antisense oligonucleotides, siRNAs, small molecules, and natural compounds (8, 10–12). Among these approaches, natural compounds have received particular attention in recent years due to their potent anticancer activities and relative safety. For example, it was recently shown that ascofuranone and curcumin, both as natural compounds, target c-Myc and exhibit anticancer activities (11, 12). However, the mechanisms by which natural compounds target c-Myc still need to be investigated.

Oridonin is a natural diterpenoid compound (Fig. 1A) that can be isolated from the Chinese medicinal herb Rabdosia rubescens and other plants in the genus Isodon, which are traditionally used in China and Japan for the treatment of various human diseases (13, 14). Oridonin exhibits proapoptotic activity against a variety of cancer cells, including those of hematologic malignancies, and shows no obvious side effects in murine models (15, 16). Although several proteins, including caspase-3, extracellular signal—regulated kinase (ERK), p53, Bax/Bcl-xL, AML-ETO, and NF-κB, have been found to be involved in the anticancer activity of oridonin (16–19), the underlying mechanisms remain largely unknown, and further mechanistic studies are needed to provide evidence supporting the use of oridonin as a drug for cancer treatment.

Figure 1.

Effects of oridonin on K562 cells and K562 xenograft tumors. A, MTT assays showing cell viability upon oridonin treatment. B, soft agar assays showing colony-forming ability. Representative photomicrographs are shown below. *, P < 0.05; **, P < 0.01. C, top, contour diagrams of flow cytometry of cells after 24-hour treatment; bottom, Western blotting showing activation of caspases by 20 μmol/L oridonin. D, oridonin delayed tumor growth in a K562 xenograft model.

Figure 1.

Effects of oridonin on K562 cells and K562 xenograft tumors. A, MTT assays showing cell viability upon oridonin treatment. B, soft agar assays showing colony-forming ability. Representative photomicrographs are shown below. *, P < 0.05; **, P < 0.01. C, top, contour diagrams of flow cytometry of cells after 24-hour treatment; bottom, Western blotting showing activation of caspases by 20 μmol/L oridonin. D, oridonin delayed tumor growth in a K562 xenograft model.

Close modal

To gain a deeper understanding of the molecular action of oridonin, we profiled miRNA expression in K562 cells in the absence or presence of oridonin and scored transcription factors reported to regulate the differentially expressed miRNAs, finally confirming that one of the miRNA regulators, c-Myc, was a molecular target of oridonin in vitro and in vivo. This observation is supported by a previous report that oridonin treatment decreased c-Myc expression in K562 cells (20). We then explored the mechanism by which oridonin targets c-Myc and revealed that oridonin activates the E3 ubiquitin ligase Fbw7 and the GSK-3 kinase, which accelerates the ubiquitin-proteasome–dependent degradation of c-Myc and results in the apoptosis of K562 cells and a delay of tumor growth in a K562 xenograft model. Our findings provide molecular evidence supporting the use of oridonin as an effective Fbw7 activator, and therefore, a c-Myc targeting agent, in the treatment of c-Myc–overexpressing hematologic malignancies or other cancers.

Reagents

Oridonin (Kangrun Pharmaceutical), MG-132 (Calbiochem), and cycloheximide (Beyotime) were dissolved in dimethyl sulfoxide (DMSO) to make stock solutions and freshly used at concentrations of 20 μmol/L, 10 μmol/L, and 50 μg/mL, respectively, unless otherwise specified. The final concentrations of DMSO were kept below 0.1% in all cell cultures.

Cell lines and transfection

The K562 and HL-60 cell lines were kindly provided by Dr. Shi-Mei Zhuang (Sun Yat-sen University, Guangdong, PR China). The B16BL6 cell line was purchased from KeyGen Biotech. Other cell lines were obtained from the cell bank of Chinese Academy of Sciences (Shanghai, China). The K562 cell line was authenticated on the basis of BCR-ABL expression and sequencing within the past 12 months, whereas other cell lines were not authenticated by the authors. All transient transfections of plasmids and siRNAs were carried out with the Neon Transfection System (Life Technologies). All siRNAs were purchased from GenePharma and transfected at a final concentration of 50 nmol/L. Sequences for siRNAs are as follows:

  • si-Myc: 5′-CGAUGUUGUUUCUGUGGAA-(dT)2-3′,

  • si-Fbw7: 5′-ACAGGACAGUGUUUACAAA-(dT)2-3′,

  • si-GSK-3: 5′-AUCUUUGGAGCCACUGAUU-(dT)2-3′

  • Negative control: 5′-UUCUCCGAACGUGUCACGU-(dT)2-3′.

Cell growth, proliferation, and apoptosis assays

Cells were cultured in 96-well cell culture plates and treated the next day as indicated. Cell viability was evaluated with MTT assays (Promega).

Cell proliferation was assessed with soft agar assays. Briefly, 2,000 cells from different treatments were mixed with a 0.35% gel (Sigma) and seeded over a 0.6% bottom gel. The colonies that contained more than 50 cells were counted and photographed under a DM LB2 microscope (Leica) 7 days later.

Apoptosis was analyzed with a FACSCalibur flow cytometer (BD Biosciences) immediately after cells were fluorescein isothiocyanate (FITC)-Annexin V/propidium iodide (PI) double-stained. The data were analyzed by FlowJo software (TreeStar).

miRNA expression profiling

The miRNA expression array including 235 human-specific miRNA probes and 8 control probes were generated as previously described (21). A 22-nucleotide (nt) oligonucleotide with a sequence complementary to a spotted control was added to the small RNA (12–28 nt) sample as a reference for normalization. The gel-purified small RNAs were dephosphorylated, radiolabeled, and hybridized to membranes. Signals were visualized with a Typhoon 8600 imager (GE Healthcare) and quantified by ImageMaster TotalLab V2.00 (GE Healthcare).

RNA expression analysis

Northern blot detection of mature miRNAs and pre-miRNAs was conducted according to the Bartel laboratory protocol (http://web.wi.mit.edu/bartel/pub/protocols.html).

For semiquantitative real-time PCR (RT-PCR), 2 μg of total RNA was used for cDNA synthesis. For quantitative RT-PCR, a SYBR RT-PCR kit (TaKaRa) was used. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as an internal control. The data were analyzed by StepOne software (Applied Biosystems). PCR primers are listed in Supplementary Table S1.

Western blot analysis

Cells were lysed as described (22). Equal amounts of proteins were loaded and separated by SDS-PAGE, transferred to polyvinylidene fluoride membranes, and detected by immunoblotting with the SuperECL Plus Detection Reagent (Applygen) or the ECL Advance Detection Kit (GE Healthcare). GAPDH was used as a loading control. See Supplementary Table S2 for information about the antibodies used.

Immunoprecipitations

Cells were lysed with immunoprecipitated lysis buffer (Beyotime) at 6 × 106 cells/mL for control samples and 1.2 × 107 cells/mL for oridonin-treated samples. Proteins were immunoprecipitated from equal volumes of cell lysates with anti-c-Myc antibody or rabbit IgG (Millipore) as a control and precipitated with Protein G Magnetic Beads (Millipore), then washed twice with lysis buffer, and subjected to Western blot analysis. All buffers contained leupeptin, phenylmethylsulfonylfluoride, and N-ethylmaleimide (Sigma).

Expression plasmids

The cDNAs of FLAG-tagged Fbw7α, Fbw7β, Fbw7γ, and wild-type c-Myc were PCR-amplified from K562 cells and cloned into the pcDNA3 vector. For construction of the c-Myc-T58A/S62A mutation plasmid, the Multipoints Mutagenesis Kit (TaKaRa) was used. Primer sequences are as follows:

  • Fbw7α-F: attaagcttgccaccatggattacaaggacgacgatgacaagatgaatcaggaactgctctctg,

  • Fbw7β-F: attaagcttgccaccatggattacaaggacgacgatgacaagatgtgtgtcccgagaagcg,

  • Fbw7γ-F: attaagcttgccaccatggattacaaggacgacgatgacaagatgtcaaaaccgggaaaacc,

  • Fbw7-R: gacgaattc tcacttcatgtccacatcaaagtc,

  • c-Myc-F: gggaattcctggatttttttcgggtagtgg,

  • c-Myc-R: tttctcgagttacgcacaagagttccgtagct,

  • c-Myc-T58A/S62A: gctgcccgctccgcccctggcacctagccgccgctc.

Efficacy of oridonin in K562 xenograft tumor models

All procedures for the experiments on mice were approved by the Animal Care and Use Committee of Sun Yat-sen University. Each 6-week-old nonobese severe-combined immunodeficient (NOD/SCID) mouse (Sun Yat-sen University) was subcutaneously injected with 6.75 × 106 K562 cells. When the tumors reached a volume of 150 to 300 mm3, the mice were randomized into 2 groups (6 mice per group) and treated intraperitoneally with oridonin (15 mg/kg) or vehicle (2% DMSO) everyday for 12 consecutive days. The tumor volumes (length × width2 × 0.5236) were measured and the body weights were monitored. All mice were euthanized 1 day after the last treatment and the tumors were cut into 8-μm sections and assessed for c-Myc expression by immunohistochemistry as previously described (23).

Statistical analysis

The data shown represent the mean of 3 independent experiments (3 replicates each). The error bars indicate the SD. Statistical differences were assessed by the Student t test. P < 0.05 was considered statistically significant.

Oridonin exhibits anticancer activity against K562 cells both in vitro and in vivo

We first investigated the in vitro effects of oridonin in K562 cells. Oridonin inhibited cell growth in a concentration- and time-dependent manner, with an IC50 of 14.6 μmol/L after a 24-hour treatment (Fig. 1A). The colony-forming ability of K562 cells in soft agar was also markedly impaired (Fig. 1B), implying a potent antiproliferative activity of oridonin. Increased percentages of Annexin V–positive cells, together with the activation of caspase-3 and caspase-9 (Fig. 1C), indicated a proapoptotic activity of oridonin. To further evaluate the efficacy of oridonin, we conducted xenograft tumor studies. Treatment with oridonin dramatically delayed the tumor growth (Fig. 1D, P < 0.05), whereas the body weight changes showed no apparent difference between the treatment and control groups (data not shown). These results show that oridonin exhibits potent anticancer effects on K562 cells both in vitro and in vivo.

miRNA expression profiling reveals that c-Myc is a target of oridonin

To explore the anticancer mechanisms of oridonin, we generated oligonucleotide DNA arrays for detection of miRNA expression because accumulating evidence suggested that miRNA expression profiling could be used as a tool to understand responses to therapy in human cancers (24, 25). We found that 14 miRNAs were upregulated and 12 miRNAs were downregulated by more than 2-fold upon oridonin treatment, primarily representing the let-7 miRNA family and the miR-17-92 paralogous clusters (including the miR-17-92 cluster, miR-106a-363 cluster, and miR-106b-25 cluster), respectively (Fig. 2A). The expression changes of several representative miRNAs were confirmed by Northern blotting (Supplementary Fig. S1A). Notably, the miRNA precursors (pre-miRNAs) of the members of the miR-17-92 paralogous clusters were also dramatically decreased by oridonin (Fig. 2B), which is suggestive of regulation upstream of pre-miRNA processing, likely at the transcriptional level. Accordingly, we began to search for transcription factors that were affected by oridonin and hence, resulted in the differential expression of the miRNAs mentioned above. As described in the Supplementary Materials and Methods, we scored transcription factors that had been reported to regulate any oridonin-induced differentially expressed miRNA and listed in Supplementary Table S3 the top 12 transcription factors that showed close correlation with oridonin in terms of miRNA regulation. Notably, c-Myc received a negative score with the highest absolute value, suggesting a strong negative regulation of c-Myc activity by oridonin. When the correlation between oridonin activity and the gene expression of these candidate transcription factors in NCI-48 cell lines was examined, c-myc still showed a high positive correlation coefficient, with an r value of 0.390 or a ρ value of 0.378 (Supplementary Table S3). c-Myc protein expression was also positively correlated with oridonin activity in these cell lines with an r value of 0.452 (P < 0.01) or a ρ value of 0.367 (P < 0.05). This significant positive correlation was also experimentally verified in 10 cell lines of leukemia and solid tumor origin (Fig. 2C). Overall, these data suggest that c-Myc is a potential molecular target of oridonin and that the activity of oridonin is highly correlated with c-Myc expression.

Figure 2.

miRNA expression profiling reveals that c-Myc is a potential molecular target of oridonin (Ori). A, a histogram showing miRNAs with more than 2-fold change based on miRNA expression profiling in K562 cells. B, Northern blotting showing pre-miRNA expression. U6 small nuclear RNA (snRNA) was used as a loading control. CT, DMSO control. C, correlation between c-Myc protein expression and IC50 (24 hour) of oridonin in a panel of cancer cell lines. The expression of c-Myc in B16BL6 was set to 1.

Figure 2.

miRNA expression profiling reveals that c-Myc is a potential molecular target of oridonin (Ori). A, a histogram showing miRNAs with more than 2-fold change based on miRNA expression profiling in K562 cells. B, Northern blotting showing pre-miRNA expression. U6 small nuclear RNA (snRNA) was used as a loading control. CT, DMSO control. C, correlation between c-Myc protein expression and IC50 (24 hour) of oridonin in a panel of cancer cell lines. The expression of c-Myc in B16BL6 was set to 1.

Close modal

Oridonin decreases c-Myc expression in vitro and in vivo

To determine the impact of oridonin on c-Myc, K562 cells were treated with oridonin and Western blot analyses were carried out. As shown in Fig. 3A and D, oridonin decreased c-Myc protein levels in a time- and concentration-dependent manner. In addition, K562 xenograft tumors treated with oridonin showed remarkably reduced levels of c-Myc compared with the control group (Fig. 3B). The oridonin-induced reduction of c-Myc resulted in the altered expression of certain miRNA primary transcripts (pri-miRNA; Fig. 3C and Supplementary Fig. S1B), all of which were reported to be transcriptional targets of c-Myc (3, 26). These changes in pri-miRNA levels were similar to but more significant than those caused by c-Myc siRNA (Fig. 3C). Overall, these results show that oridonin targets c-Myc in vitro and in vivo, by reducing c-Myc protein levels and thereby impairing its transcriptional activity.

Figure 3.

Oridonin (Ori) decreases c-Myc expression and impairs its transcription activity. A, oridonin decreases c-Myc protein expression in a time-dependent manner in K562 cells. B, immunohistochemistry of c-Myc in K562 xenograft tumors. The sections were developed by diaminobenzidine and counterstained with hematoxylin. Bar, 20 μm. C, qRT-PCR analysis of pri-miRNA expression. Top, confirmation of c-Myc expression by Western blotting. D, concentration-dependent reduction of c-Myc by oridonin (24 hours) positively correlates with the cytotoxic activity of oridonin. Top, MTT assays; bottom, Western blot analyses. CT, control; NC, negative control.

Figure 3.

Oridonin (Ori) decreases c-Myc expression and impairs its transcription activity. A, oridonin decreases c-Myc protein expression in a time-dependent manner in K562 cells. B, immunohistochemistry of c-Myc in K562 xenograft tumors. The sections were developed by diaminobenzidine and counterstained with hematoxylin. Bar, 20 μm. C, qRT-PCR analysis of pri-miRNA expression. Top, confirmation of c-Myc expression by Western blotting. D, concentration-dependent reduction of c-Myc by oridonin (24 hours) positively correlates with the cytotoxic activity of oridonin. Top, MTT assays; bottom, Western blot analyses. CT, control; NC, negative control.

Close modal

We next explored the correlation between oridonin potency and the reduction of c-Myc in 4 leukemia and lymphoma cell lines including K562, KU812, HL-60, and Daudi. In all of these cell lines, the cell growth was inhibited by oridonin in a concentration-dependent manner (Fig. 3D). Interestingly, the expression change of c-Myc was almost entirely consistent with the change in cell viability (Fig. 3D), which further shows that the reduction of c-Myc is correlated with the anticancer effects of oridonin and that c-Myc is an important target of oridonin.

Oridonin promotes the ubiquitin-proteasome–dependent degradation of c-Myc

Given the importance of c-Myc in diverse cellular processes, we further explored the mechanisms by which c-Myc is regulated. RT-PCR showed that the c-myc mRNA level remained unchanged after oridonin treatment (Fig. 4A), which was also confirmed by qRT-PCR (data not shown) and suggested that oridonin did not affect the transcription or mRNA stability of c-myc. We also conducted dual luciferase assays to rule out the existence of oridonin-responsive elements within the 5′-untranslated region (UTR) or 3′UTR of c-myc that could affect the translation of c-myc mRNA (Supplementary Fig. S2A). Thus, on the basis of the above results, it appears unlikely that oridonin regulates c-Myc by affecting the synthesis of its mRNA or protein.

Figure 4.

Oridonin (Ori) promotes the ubiquitin (Ub)-proteasome–mediated degradation of c-Myc in K562 cells. A, cells were treated with 20 μmol/L oridonin for 24 hours, and the mRNA levels of c-myc were examined with RT-PCR. B, cycloheximide (CHX) chase assays showing the half-life of the c-Myc protein. C, cells pretreated with MG-132 for 2 hours were then exposed to oridonin for 1 hour and lysed for immunoprecipitation (IP). D, cells preincubated with MG-132 for 2 hours were then treated with oridonin (20 μmol/L) for the time indicated and lysed for Western blot (WB) analysis. CT, control.

Figure 4.

Oridonin (Ori) promotes the ubiquitin (Ub)-proteasome–mediated degradation of c-Myc in K562 cells. A, cells were treated with 20 μmol/L oridonin for 24 hours, and the mRNA levels of c-myc were examined with RT-PCR. B, cycloheximide (CHX) chase assays showing the half-life of the c-Myc protein. C, cells pretreated with MG-132 for 2 hours were then exposed to oridonin for 1 hour and lysed for immunoprecipitation (IP). D, cells preincubated with MG-132 for 2 hours were then treated with oridonin (20 μmol/L) for the time indicated and lysed for Western blot (WB) analysis. CT, control.

Close modal

Previous studies have shown that the c-Myc protein has a very short half-life and is degraded through the ubiquitin-proteasome pathway. In the presence of oridonin, the half-life of c-Myc in K562 cells was further reduced, as observed with cycloheximide chase assays (Fig. 4B), indicating the accelerated degradation of c-Myc protein. We next evaluated the effect of oridonin on c-Myc ubiquitination. Immunoprecipitations were carried out and the results showed that ubiquitinated c-Myc accumulated upon oridonin treatment (Fig. 4C). When we used MG-132 to inhibit the 26S proteasome activity in K562 cells, the oridonin-induced downregulation of c-Myc was significantly antagonized (Fig. 4D). Similar results were observed in KU812, HL-60, and Daudi cells (Supplementary Fig. S2B). We therefore concluded that oridonin downregulated c-Myc by promoting its ubiquitin-proteasome–dependent degradation rather than by decreasing its synthesis.

Oridonin activates the Fbw7 E3 ubiquitin ligase

The process of ubiquitination has been shown to require the consecutive action of an E1 ubiquitin–activating enzyme, an E2 ubiquitin–conjugating enzyme, and an E3 ubiquitin ligase. Of these enzymes, the E3 ligase is considered the most important component, as it confers substrate specificity (27). To determine how oridonin affects the ubiquitination of c-Myc, we examined the protein expression levels of Fbw7, an E3 ubiquitin ligase shown to be responsible for the ubiquitin-mediated degradation of c-Myc (6, 7) and other oncogenes such as cyclin E (28, 29) and mTOR (30). Oridonin treatment significantly increased the expression of Fbw7α and Fbw7γ in a time-dependent manner, which was accompanied by the remarkable downregulation of c-Myc, cyclin E, and mTOR, among which the reduction of c-Myc was the most significant (Fig. 5A and Supplementary Fig. S3A). We also detected the expression of the deubiquitinating enzyme USP28, which could interact with Fbw7α and thereby antagonize the degradation of c-Myc (31). As shown in Fig. 5A, oridonin did not affect the expression of USP28. In addition, we carried out immunoprecipitations to assess the interaction of c-Myc with Fbw7. As expected, c-Myc–associated Fbw7 increased significantly in oridonin-treated cells (Fig. 5A).

Figure 5.

Activation of Fbw7 and GSK-3 by oridonin (Ori). A, activation of Fbw7 in K562 cells as shown by Western blot analysis and immunoprecipitation (IP). B, cells preincubated with MG-132 for 2 hours were then treated with oridonin (20 μmol/L) for the indicated time and lysed for Western blotting. C, oridonin decreases phosphorylated GSK-3 and increases T58-phosphorylated c-Myc in K562 cells. D, inhibition of GSK-3 by siRNA (top) or LiCl (bottom) partially attenuated oridonin-induced c-Myc degradation. The intensities of the bands of c-Myc were quantitated and normalized to those of GAPDH. CT, control; NC, negative control.

Figure 5.

Activation of Fbw7 and GSK-3 by oridonin (Ori). A, activation of Fbw7 in K562 cells as shown by Western blot analysis and immunoprecipitation (IP). B, cells preincubated with MG-132 for 2 hours were then treated with oridonin (20 μmol/L) for the indicated time and lysed for Western blotting. C, oridonin decreases phosphorylated GSK-3 and increases T58-phosphorylated c-Myc in K562 cells. D, inhibition of GSK-3 by siRNA (top) or LiCl (bottom) partially attenuated oridonin-induced c-Myc degradation. The intensities of the bands of c-Myc were quantitated and normalized to those of GAPDH. CT, control; NC, negative control.

Close modal

To further show the importance of Fbw7 in oridonin-induced c-Myc degradation, we used 2 T-cell acute lymphoblastic leukemia (T-ALL) cell lines, Jurkat, and CCRF-CEM, which harbor an R505C and an R465C missense mutation in Fbw7, respectively (32, 33). When exposed to oridonin, the decrease of c-Myc in these 2 cell lines was less evident than that in K562 (Fig. 5B), implying that the abundance of functional Fbw7 affects the efficiency of oridonin-induced c-Myc degradation. Taken together, our results strongly indicate that oridonin promotes the degradation of c-Myc through the activation of the Fbw7 E3 ligase.

Oridonin activates GSK-3 and enhances the phosphorylation of c-Myc at T58

GSK-3 is an Fbw7 priming kinase that phosphorylates most known Fbw7 substrates. It was previously shown that the GSK-3–mediated phosphorylation of c-Myc T58 was needed for the efficient binding of c-Myc by Fbw7 (34, 35). We investigated GSK-3 activity in K562 cells upon oridonin treatment. Phosphorylated/inactive GSK-3 decreased, whereas the total GSK-3 protein levels remained unchanged (Fig. 5C), indicating the activation of GSK-3. Accordingly, T58-phosphorylated c-Myc increased significantly as expected (Fig. 5C). When we transiently inhibited GSK-3 with either siRNA or LiCl (a selective GSK-3 inhibitor), the oridonin-induced degradation of c-Myc was somewhat delayed (Fig. 5D), suggesting a dependence on GSK-3 in this process.

The Fbw7-c-Myc pathway contributes to the anticancer activities of oridonin

Because oridonin was able to upregulate Fbw7, we evaluated the effects of Fbw7 overexpression in K562 cells. As shown in Fig. 6A, the overexpression of the 3 isoforms of Fbw7 (α, β, and γ) all decreased c-Myc levels, although to different extents. At the cellular level, Fbw7 remarkably inhibited the growth of K562 cells in culture medium and in soft agar (Fig. 6A), and substantially increased the fraction of apoptotic cells as determined by fluorescence-activated cell-sorting (FACS) analysis (Fig. 6B). The anticancer property of Fbw7 was comparable with that of c-Myc silencing (Fig. 6A and B) except for a slighter effect on the anchorage-independent growth in soft agar (Fig. 6A). These results reveal that the forced expression of Fbw7 inhibits growth and induces apoptosis in K562 cells, which could also be achieved by silencing c-Myc expression.

Figure 6.

The Fbw7-c-Myc pathway contributes to the anticancer activities of oridonin. A, Fbw7 overexpression or c-Myc knockdown decreases cell viability and colony-forming ability in K562 cells. Relative intensities of c-Myc levels are indicated in the left. B, cell apoptosis shown by contour diagrams of flow cytometry. C, knockdown of Fbw7 or forced expression of stable c-Myc (c-Myc-mut) restores cell viability in the presence of oridonin. D, the proposed molecular mechanisms of action of oridonin. The red arrows denote activation whereas the green arrow denotes inhibition. NC, negative control; Ub, ubiquitin.

Figure 6.

The Fbw7-c-Myc pathway contributes to the anticancer activities of oridonin. A, Fbw7 overexpression or c-Myc knockdown decreases cell viability and colony-forming ability in K562 cells. Relative intensities of c-Myc levels are indicated in the left. B, cell apoptosis shown by contour diagrams of flow cytometry. C, knockdown of Fbw7 or forced expression of stable c-Myc (c-Myc-mut) restores cell viability in the presence of oridonin. D, the proposed molecular mechanisms of action of oridonin. The red arrows denote activation whereas the green arrow denotes inhibition. NC, negative control; Ub, ubiquitin.

Close modal

We further investigated the role of the Fbw7-c-Myc pathway in the anticancer activities of oridonin. The knockdown of Fbw7 by siRNA that targeted the common region of Fbw7 resulted in the accumulation of c-Myc and led to the reduced sensitization of cells to oridonin-induced apoptosis, as shown by the presence of fewer apoptotic cells and more viable cells (Fig. 6C). In addition, the overexpression of stable c-Myc that was mutated at both T58 and S62 (both within the Cdc4 phospho-degron) and unable to be ubiquitinated partially conferred resistance to oridonin-induced apoptosis compared with the overexpression of wild-type c-Myc (Fig. 6C). These results strongly suggest the importance of Fbw7 upregulation and c-Myc degradation in the proapoptotic ability of oridonin.

In this study, we explored the mechanism of action of the diterpenoid compound oridonin from a new point of view. We analyzed miRNA expression data in the context of oridonin treatment and determined that a transcriptional regulator of most of the differentially expressed miRNAs, c-Myc, was a molecular target of oridonin. By activating the E3 ubiquitin ligase Fbw7 and the GSK-3 kinase, oridonin induces the rapid turnover of c-Myc and the reprogramming of c-Myc–regulated miRNAs, resulting in growth inhibition and apoptosis. These mechanisms are summarized in Fig. 6D.

Research indicates that c-Myc acts as a double-edged sword in cell fate decisions because it can promote cell growth or induce apoptosis (36, 37). Our results showed that inhibition of c-Myc expression led to severe cell death (Fig. 6B), suggesting that the deregulated c-Myc in leukemia and lymphoma cells was oncogenic rather that tumor-suppressive. Because of its oncogenic properties, c-Myc has been investigated and shown to be an effective target for cancer therapy (8, 10, 38). Because proteins are the executors of cell function, promoting the degradation of the c-Myc protein by oridonin may be more efficient than other strategies aimed at disrupting c-myc expression. A question arose as to whether the oridonin-induced reduction of c-Myc was concomitant with apoptosis or a cause of apoptosis. The latter possibility was validated because the reduction in c-Myc occurred independently of the activation of caspases (Supplementary Fig. S2C), and the oridonin-induced apoptosis could be partially blocked by the forced expression of c-Myc in K562 cells. Thus, as a natural compound, oridonin targets c-Myc and exhibits antiproliferative and proapoptotic activities in c-Myc–overexpressing hematologic malignancies and other cancer cells.

The mechanism underlying the oridonin-induced c-Myc downregulation was still unclear, although the decrease of c-Myc by oridonin was also reported in prior work (20, 39). It was previously shown that treatment of SW1116 colorectal cancer cells with 25 μmol/L oridonin reduced c-myc transcription and protein levels after 24 hours or a longer period of time (39). However, our work showed that oridonin did not affect the mRNA level of c-myc in K562 cells after a 24-hour treatment but dramatically induced the degradation of c-Myc protein as early as 40 minutes. The rapid degradation of c-Myc was also observed in other malignant hematologic cell lines, including KU812, HL-60, and Daudi (Supplementary Fig. S2B). We showed, for the first time, that a ubiquitin-proteasome-dependent pathway was involved in oridonin-induced c-Myc downregulation. There are 2 rate-limiting steps in proteasomal degradation: the specific modification of substrate proteins, such as phosphorylation, and the enzymatic attachment of ubiquitin to substrate proteins (40). In this study, we showed that oridonin acts upon these 2 steps to achieve its most potent effect on c-Myc degradation in K562 cells (Fig. 6D). However, we observed that the transient inhibition of GSK-3 could not fully antagonize the oridonin-induced degradation of c-Myc. This observation may suggest that the activation of GSK-3 was less important than the activation of Fbw7 in oridonin-induced c-Myc degradation or that the residual kinase activity of GSK-3 was sufficient for the phosphorylation of c-Myc. We also examined the phosphorylation levels of AKT, an upstream kinase of GSK3. Oridonin significantly reduced pAKT levels (data not shown), which was consistent with previous reports (41) and suggested that the downregulation of pAKT may be involved in the activities of oridonin.

The ubiquitin-proteasome system (UPS) plays a crucial role in fine-tuning the abundance of key proteins in diverse cellular processes (27). The deregulation of the UPS has been observed in many types of cancer, and studies targeting the UPS for cancer therapy are being conducted (27). As a component of the UPS, Fbw7 ubiquitin ligase plays a vital role in cell fate decisions (35, 42). It was recently shown that Fbw7 controlled the fate of hematopoietic stem cells (43) and affected the proliferation and lymphomagenesis of mouse T cells (44), both through the regulation of c-Myc expression. In addition, as a tumor suppressor, Fbw7 is inactivated in numerous human malignancies. Because the overexpression of c-Myc is frequently observed in human malignancies, promotion of the degradation of oncogenic c-Myc through the activation of Fbw7 may provide a novel therapeutic approach. Our finding that oridonin upregulates Fbw7 in malignant cells of hematologic origin (Fig. 5A and Supplementary Fig. S3B) is therefore promising for the development of Fbw7-based cancer therapy. It is worth noting that among the several substrate proteins examined, the expression change in c-Myc was the most significant, implying a more important role for c-Myc in the response of cancer cells to oridonin. Besides, other prosurvival proteins, such as MCL1 and KLF5, have been shown to be targeted by Fbw7 (45–47), raising the possibility that these proteins are also involved in the anticancer activities of oridonin.

Interestingly, it has recently been reported that several stressors, including hypoxia, nickel compounds (48), or UV irradiation–induced DNA damage (49), could enhance the Fbw7-mediated degradation of c-Myc by targeting USP28, another component of the UPS. However, these types of stress are unsuitable or difficult to use as clinical therapeutic approaches. As a natural anticancer compound, oridonin is also able to influence the UPS by quickly upregulating the expression of Fbw7 and enhancing the binding of Fbw7 to its substrate rather than by affecting the expression of USP28. From this point of view, oridonin can be seen as an agonist of Fbw7, and as a result, has potential clinical implications. As to the mechanism underlying the Fbw7 activation, we found that the mRNA levels of Fbw7 were not affected by oridonin after a 2-hour treatment, whereas a 24-hour treatment did upregulate the mRNA levels of Fbw7β (data not shown), which might be mediated by p53 (50). Further mechanistic studies are being conducted.

In summary, we showed the potent anticancer activity of oridonin in vitro and in vivo and determined that the underlying mechanism involves the regulation of an E3 ligase–substrate pair (Fbw7-c-Myc). It appears that the exposure of leukemia and lymphoma cells to oridonin activates Fbw7 and GSK-3, and in this way, effectively triggers the Fbw7-mediated ubiquitination and proteasome-dependent degradation of c-Myc, leading to growth inhibition and apoptosis. Our results also provide evidence for the use of miRNA expression profiling as a tool to understand the molecular mechanisms of drug actions by retracing the expression changes in upstream transcription factors. In addition, it is possible that the altered expression of oncogenic miRNAs (e.g., the miR-17-92 cluster) and tumor suppressor miRNAs (e.g., let-7a) caused by c-Myc downregulation accounts for the anticancer activity of oridonin. It will be valuable to identify and study the roles of the target proteins of these miRNAs as well as other c-Myc downstream genes in the context of oridonin treatment to better understand the anticancer activities of this natural compound.

No potential conflicts of interest were disclosed.

Conception and design: L.-H. Qu, H.-L. Huang, H.-Y. Weng, H. Zhou

Development of methodology: H.-L. Huang, H.-Y. Weng, L.-Q. Wang, Q.-J. Huang, H. Zhou

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): L.-H. Qu, H.-L. Huang, H.-Y. Weng, L.-Q. Wang, Q.-J. Huang, P.-P. Zhao, H. Zhou

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): L.-H. Qu, H.-L. Huang, H.-Y. Weng, C.-H. Yu, Q.-J. Huang, J.-Z. Wen, H. Zhou

Writing, review, and/or revision of the manuscript: L.-H. Qu, H.-L. Huang, H.-Y. Weng, C.-H. Yu, J.-Z. Wen, H. Zhou

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): L.-H. Qu, H. Zhou

Study supervision: L.-H. Qu, H. Zhou

This research was supported by the National Natural Science Foundation of China (No. 30830066, 81070589, and 30870530) and the National Basic Research Program (No. 2011CB 811300) from the Ministry of Science and Technology of China.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Fernandez
PC
,
Frank
SR
,
Wang
L
,
Schroeder
M
,
Liu
S
,
Greene
J
, et al
Genomic targets of the human c-Myc protein
.
Genes Dev
2003
;
17
:
1115
29
.
2.
Zeller
KI
,
Zhao
X
,
Lee
CW
,
Chiu
KP
,
Yao
F
,
Yustein
JT
, et al
Global mapping of c-Myc binding sites and target gene networks in human B cells
.
Proc Natl Acad Sci U S A
2006
;
103
:
17834
9
.
3.
Chang
TC
,
Yu
D
,
Lee
YS
,
Wentzel
EA
,
Arking
DE
,
West
KM
, et al
Widespread microRNA repression by Myc contributes to tumorigenesis
.
Nat Genet
2008
;
40
:
43
50
.
4.
O'Donnell
KA
,
Wentzel
EA
,
Zeller
KI
,
Dang
CV
,
Mendell
JT
. 
c-Myc-regulated microRNAs modulate E2F1 expression
.
Nature
2005
;
435
:
839
43
.
5.
Bartel
DP
. 
MicroRNAs: genomics, biogenesis, mechanism, and function
.
Cell
2004
;
116
:
281
97
.
6.
Welcker
M
,
Orian
A
,
Jin
J
,
Grim
JE
,
Harper
JW
,
Eisenman
RN
, et al
The Fbw7 tumor suppressor regulates glycogen synthase kinase 3 phosphorylation-dependent c-Myc protein degradation
.
Proc Natl Acad Sci U S A
2004
;
101
:
9085
90
.
7.
Yada
M
,
Hatakeyama
S
,
Kamura
T
,
Nishiyama
M
,
Tsunematsu
R
,
Imaki
H
, et al
Phosphorylation-dependent degradation of c-Myc is mediated by the F-box protein Fbw7
.
EMBO J
2004
;
23
:
2116
25
.
8.
Vita
M
,
Henriksson
M
. 
The Myc oncoprotein as a therapeutic target for human cancer
.
Semin Cancer Biol
2006
;
16
:
318
30
.
9.
Hoffman
B
,
Amanullah
A
,
Shafarenko
M
,
Liebermann
DA
. 
The proto-oncogene c-myc in hematopoietic development and leukemogenesis
.
Oncogene
2002
;
21
:
3414
21
.
10.
Soucek
L
,
Whitfield
J
,
Martins
CP
,
Finch
AJ
,
Murphy
DJ
,
Sodir
NM
, et al
Modelling Myc inhibition as a cancer therapy
.
Nature
2008
;
455
:
679
83
.
11.
Senft
C
,
Polacin
M
,
Priester
M
,
Seifert
V
,
Kogel
D
,
Weissenberger
J
. 
The nontoxic natural compound Curcumin exerts anti-proliferative, anti-migratory, and anti-invasive properties against malignant gliomas
.
BMC Cancer
2010
;
10
:
491
.
12.
Jeong
JH
,
Kang
SS
,
Park
KK
,
Chang
HW
,
Magae
J
,
Chang
YC
. 
p53-independent induction of G1 arrest and p21WAF1/CIP1 expression by ascofuranone, an isoprenoid antibiotic, through downregulation of c-Myc
.
Mol Cancer Ther
2010
;
9
:
2102
13
.
13.
Fujita
E
,
Nagao
Y
,
Node
M
,
Kaneko
K
,
Nakazawa
S
,
Kuroda
H
. 
Antitumor activity of the Isodon diterpenoids: structural requirements for the activity
.
Experientia
1976
;
32
:
203
6
.
14.
Henan Medical Institute, Henan Medical College, Yunnan Institute of Botany
. 
Oridonin: a new antitumor subject
.
Chin Sci Bull
1978
;
23
:
53
6
.
15.
Ikezoe
T
,
Chen
SS
,
Tong
XJ
,
Heber
D
,
Taguchi
H
,
Koeffler
HP
. 
Oridonin induces growth inhibition and apoptosis of a variety of human cancer cells
.
Int J Oncol
2003
;
23
:
1187
93
.
16.
Zhou
GB
,
Kang
H
,
Wang
L
,
Gao
L
,
Liu
P
,
Xie
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
:
3441
50
.
17.
Li
CY
,
Wang
EQ
,
Cheng
Y
,
Bao
JK
. 
Oridonin: an active diterpenoid targeting cell cycle arrest, apoptotic and autophagic pathways for cancer therapeutics
.
Int J Biochem Cell Biol
2011
;
43
:
701
4
.
18.
Ikezoe
T
,
Yang
Y
,
Bandobashi
K
,
Saito
T
,
Takemoto
S
,
Machida
H
, et al
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
:
578
86
.
19.
Leung
CH
,
Grill
SP
,
Lam
W
,
Han
QB
,
Sun
HD
,
Cheng
YC
. 
Novel mechanism of inhibition of nuclear factor-kappa B DNA-binding activity by diterpenoids isolated from Isodon rubescens
.
Mol Pharmacol
2005
;
68
:
286
97
.
20.
Li
RF
,
Wang
QD
. 
[Regulation of telomerase activity and cell cycle of K562 cells by oridonin]
.
Yao Xue Xue Bao
2004
;
39
:
865
8
.
21.
Krichevsky
AM
,
King
KS
,
Donahue
CP
,
Khrapko
K
,
Kosik
KS
. 
A microRNA array reveals extensive regulation of microRNAs during brain development
.
RNA
2003
;
9
:
1274
81
.
22.
Malempati
S
,
Tibbitts
D
,
Cunningham
M
,
Akkari
Y
,
Olson
S
,
Fan
G
, et al
Aberrant stabilization of c-Myc protein in some lymphoblastic leukemias
.
Leukemia
2006
;
20
:
1572
81
.
23.
Cai
MY
,
Hou
JH
,
Rao
HL
,
Luo
RZ
,
Li
M
,
Pei
XQ
, et al
High expression of H3K27me3 in human hepatocellular carcinomas correlates closely with vascular invasion and predicts worse prognosis in patients
.
Mol Med
2011
;
17
:
12
20
.
24.
Iorio
MV
,
Casalini
P
,
Tagliabue
E
,
Menard
S
,
Croce
CM
. 
MicroRNA profiling as a tool to understand prognosis, therapy response and resistance in breast cancer
.
Eur J Cancer
2008
;
44
:
2753
9
.
25.
Blower
PE
,
Verducci
JS
,
Lin
S
,
Zhou
J
,
Chung
JH
,
Dai
Z
, et al
MicroRNA expression profiles for the NCI-60 cancer cell panel
.
Mol Cancer Ther
2007
;
6
:
1483
91
.
26.
Petrocca
F
,
Visone
R
,
Onelli
MR
,
Shah
MH
,
Nicoloso
MS
,
de Martino
I
, et al
E2F1-regulated microRNAs impair TGFbeta-dependent cell-cycle arrest and apoptosis in gastric cancer
.
Cancer Cell
2008
;
13
:
272
86
.
27.
Hoeller
D
,
Dikic
I
. 
Targeting the ubiquitin system in cancer therapy
.
Nature
2009
;
458
:
438
44
.
28.
Strohmaier
H
,
Spruck
CH
,
Kaiser
P
,
Won
KA
,
Sangfelt
O
,
Reed
SI
. 
Human F-box protein hCdc4 targets cyclin E for proteolysis and is mutated in a breast cancer cell line
.
Nature
2001
;
413
:
316
22
.
29.
Koepp
DM
,
Schaefer
LK
,
Ye
X
,
Keyomarsi
K
,
Chu
C
,
Harper
JW
, et al
Phosphorylation-dependent ubiquitination of cyclin E by the SCFFbw7 ubiquitin ligase
.
Science
2001
;
294
:
173
7
.
30.
Mao
JH
,
Kim
IJ
,
Wu
D
,
Climent
J
,
Kang
HC
,
DelRosario
R
, et al
FBXW7 targets mTOR for degradation and cooperates with PTEN in tumor suppression
.
Science
2008
;
321
:
1499
502
.
31.
Popov
N
,
Wanzel
M
,
Madiredjo
M
,
Zhang
D
,
Beijersbergen
R
,
Bernards
R
, et al
The ubiquitin-specific protease USP28 is required for MYC stability
.
Nat Cell Biol
2007
;
9
:
765
74
.
32.
O'Neil
J
,
Grim
J
,
Strack
P
,
Rao
S
,
Tibbitts
D
,
Winter
C
, et al
FBW7 mutations in leukemic cells mediate NOTCH pathway activation and resistance to gamma-secretase inhibitors
.
J Exp Med
2007
;
204
:
1813
24
.
33.
Malyukova
A
,
Dohda
T
,
von der Lehr
N
,
Akhoondi
S
,
Corcoran
M
,
Heyman
M
, et al
The tumor suppressor gene hCDC4 is frequently mutated in human T-cell acute lymphoblastic leukemia with functional consequences for Notch signaling
.
Cancer Res
2007
;
67
:
5611
6
.
34.
Sears
R
,
Nuckolls
F
,
Haura
E
,
Taya
Y
,
Tamai
K
,
Nevins
JR
. 
Multiple Ras-dependent phosphorylation pathways regulate Myc protein stability
.
Genes Dev
2000
;
14
:
2501
14
.
35.
Crusio
KM
,
King
B
,
Reavie
LB
,
Aifantis
I
. 
The ubiquitous nature of cancer: the role of the SCF(Fbw7) complex in development and transformation
.
Oncogene
2010
;
29
:
4865
73
.
36.
Askew
DS
,
Ashmun
RA
,
Simmons
BC
,
Cleveland
JL
. 
Constitutive c-myc expression in an IL-3-dependent myeloid cell line suppresses cell cycle arrest and accelerates apoptosis
.
Oncogene
1991
;
6
:
1915
22
.
37.
Evan
GI
,
Littlewood
TD
. 
The role of c-myc in cell growth
.
Curr Opin Genet Dev
1993
;
3
:
44
9
.
38.
Delgado
MD
,
Leon
J
. 
Myc roles in hematopoiesis and leukemia
.
Genes Cancer
2010
;
1
:
605
16
.
39.
Gao
FH
,
Hu
XH
,
Li
W
,
Liu
H
,
Zhang
YJ
,
Guo
ZY
, et al
Oridonin induces apoptosis and senescence in colorectal cancer cells by increasing histone hyperacetylation and regulation of p16, p21, p27 and c-myc
.
BMC Cancer
2010
;
10
:
610
.
40.
Sundberg
TB
,
Ney
GM
,
Subramanian
C
,
Opipari
AW
 Jr
,
Glick
GD
. 
The immunomodulatory benzodiazepine Bz-423 inhibits B-cell proliferation by targeting c-myc protein for rapid and specific degradation
.
Cancer Res
2006
;
66
:
1775
82
.
41.
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
:
261
8
.
42.
Welcker
M
,
Clurman
BE
. 
FBW7 ubiquitin ligase: a tumour suppressor at the crossroads of cell division, growth and differentiation
.
Nat Rev Cancer
2008
;
8
:
83
93
.
43.
Reavie
L
,
Della
Gatta G
,
Crusio
K
,
Aranda-Orgilles
B
,
Buckley
SM
,
Thompson
B
, et al
Regulation of hematopoietic stem cell differentiation by a single ubiquitin ligase-substrate complex
.
Nat Immunol
2010
;
11
:
207
15
.
44.
Onoyama
I
,
Tsunematsu
R
,
Matsumoto
A
,
Kimura
T
,
de Alboran
IM
,
Nakayama
K
, et al
Conditional inactivation of Fbxw7 impairs cell-cycle exit during T-cell differentiation and results in lymphomatogenesis
.
J Exp Med
2007
;
204
:
2875
88
.
45.
Inuzuka
H
,
Shaik
S
,
Onoyama
I
,
Gao
D
,
Tseng
A
,
Maser
RS
, et al
SCF(FBW7) regulates cellular apoptosis by targeting MCL1 for ubiquitylation and destruction
.
Nature
2011
;
471
:
104
9
.
46.
Zhao
D
,
Zheng
HQ
,
Zhou
Z
,
Chen
C
. 
The Fbw7 tumor suppressor targets KLF5 for ubiquitin-mediated degradation and suppresses breast cell proliferation
.
Cancer Res
2010
;
70
:
4728
38
.
47.
Wertz
IE
,
Kusam
S
,
Lam
C
,
Okamoto
T
,
Sandoval
W
,
Anderson
DJ
, et al
Sensitivity to antitubulin chemotherapeutics is regulated by MCL1 and FBW7
.
Nature
2011
;
471
:
110
4
.
48.
Li
Q
,
Kluz
T
,
Sun
H
,
Costa
M
. 
Mechanisms of c-myc degradation by nickel compounds and hypoxia
.
PLoS One
2009
;
4
:
e8531
.
49.
Popov
N
,
Herold
S
,
Llamazares
M
,
Schulein
C
,
Eilers
M
. 
Fbw7 and Usp28 regulate myc protein stability in response to DNA damage
.
Cell Cycle
2007
;
6
:
2327
31
.
50.
Kimura
T
,
Gotoh
M
,
Nakamura
Y
,
Arakawa
H
. 
hCDC4b, a regulator of cyclin E, as a direct transcriptional target of p53
.
Cancer Sci
2003
;
94
:
431
6
.