Skip to main content
Erschienen in: BMC Cancer 1/2011

Open Access 01.12.2011 | Research article

Small interfering RNA targeting mcl-1 enhances proteasome inhibitor-induced apoptosis in various solid malignant tumors

verfasst von: Wei Zhou, Jingzi Hu, Haimei Tang, Da Wang, Xuefeng Huang, Chao He, Hongbo Zhu

Erschienen in: BMC Cancer | Ausgabe 1/2011

Abstract

Background

Targeting the ubiquitin-proteasome pathway is a promising approach for anticancer strategies. Recently, we found Bik accumulation in cancer cell lines after they were treated with bortezomib. However, recent evidence indicates that proteasome inhibitors may also induce the accumulation of anti-apoptotic Bcl-2 family members. The current study was designed to analyze the levels of several anti-apoptotic members of Bcl-2 family in different human cancer cell lines after they were treated with proteasome inhibitors.

Methods

Different human cancer cell lines were treated with proteasome inhibitors. Western blot were used to investigate the expression of Mcl-1 and activation of mitochondrial apoptotic signaling. Cell viability was investigated using SRB assay, and induction of apoptosis was measured using flow cytometry.

Results

We found elevated Mcl-1 level in human colon cancer cell lines DLD1, LOVO, SW620, and HCT116; human ovarian cancer cell line SKOV3; and human lung cancer cell line H1299, but not in human breast cancer cell line MCF7 after they were treated with bortezomib. This dramatic Mcl-1 accumulation was also observed when cells were treated with other two proteasome inhibitors, MG132 and calpain inhibitor I (ALLN). Moreover, our results showed Mcl-1 accumulation was caused by stabilization of the protein against degradation. Reducing Mcl-1 accumulation by Mcl-1 siRNA reduced Mcl-1 accumulation and enhanced proteasome inhibitor-induced cell death and apoptosis, as evidenced by the increased cleavage of caspase-9, caspase-3, and poly (ADP-ribose) polymerase.

Conclusions

Our results showed that it was not only Bik but also Mcl-1 accumulation during the treatment of proteasome inhibitors, and combining proteasome inhibitors with Mcl-1 siRNA would enhance the ultimate anticancer effect suggesting this combination might be a more effective strategy for cancer therapy.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1471-2407-11-485) contains supplementary material, which is available to authorized users.

Competing interests

The authors declare that they have no competing interests.

Authors' contributions

WZ performed experimental and statistical analysis and drafted the manuscript. JZH and HMT participated in flow cytometry and SRB assay. DW participated in Western blot analysis. XFH and CH participated in manuscript proofreading. HBZ conceived the design, provided financial support, participated Western blot analysis and revised the manuscript. All authors read and approved the final manuscript.

Background

Proteasome inhibitors represent a new class of agents for cancer therapeutics [1, 2]. The 26S proteasome is a 2, 000-kDa multimeric cylindrical complex comprising a 20S catalytic core and a 19S regulatory subunit [3]. This structure is a promising target for cancer therapy because it regulates the crucial process of proteasome-mediated protein degradation, which involves many proteins such as cyclins, caspases, Bcl-2 and the nuclear factor of κB (NF-κB) [2, 4]. Inhibiting proteasome activity leads to the accumulation of these proteins, resulting in cell cycle arrest and apoptosis. Bortezomib, a specific and selective inhibitor of 26S proteasome, was approved for initial treatment of patients with Multiple Myeloma by the US Food and Drug Administration in 2008. Proteasome inhibitor-based combination therapies suggest that proteasome inhibitors could enhance chemosensitivity or reverse radiotherapy/chemotherapy resistance [5].
A growing body of evidence indicates that the intrinsic (or mitochondrial) apoptosis pathway represents a fundamental mechanism of apoptosis triggered by proteasome inhibition [6, 7]. Indeed, the Bcl-2 family proteins, key activators of mitochondrial apoptosis, play a fundamental role in mediating proteasome inhibition-induced toxicity [8]. However, proteasome inhibitors not only increase the pro-apoptotic Bcl-2 proteins [911], but they may also lead to the accumulation of anti-apoptotic Bcl-2 proteins [12]. These proteins include the Mcl-1 anti-apoptotic protein, originally identified as an early induction gene during the differentiation of myeloid leukemia cells [13], which could block cytochrome c release from mitochondria by forming heterodimers with BH3-only proteins Bim and NOXA, or with Bak [14, 15]. Thus, proteasome inhibitor-induced Mcl-1 accumulation may negatively affect their cytotoxic activity. Targeting Mcl-1 might be a strategy for enhancing the anticancer effect of proteasome inhibitors [16].
Our previous study demonstrated that proteasome inhibitors would induced a rapid Bik accumulation in various cancer cells [17]. Bik was also a member of BH3-only proteins, so the question of whether there were elevated anti-apoptotic members of Bcl-2 family existing in our system emerged inevitably. To clarify this question, we analyzed the levels of several anti-apoptotic members of Bcl-2 family in different human cancer cell lines after they were treated with proteasome inhibitors. Our results demonstrated that proteasome inhibitors induced a rapid accumulation of Mcl-1 but not others in our cell lines. The possible underlying mechanism of this accumulation might be the stabilization of proteins from degradation. We also showed that the knockdown of Mcl-1 levels by RNA interference enhanced the apoptosis induced by proteasome inhibitors. These findings suggested that treatment with proteasome inhibitors could induce Mcl-1 accumulation in various cancer cells and that combining these inhibitors with Mcl-1 siRNA might be a more effective strategy for cancer therapy.

Methods

Cells and cell culture

Human colon cancer cell lines DLD1, LOVO, SW620, and HCT116; human lung cancer cell lines H1299; human ovarian cancer cell line SKOV3 which were owned by our lab and human breast cancer cell line MCF7 that was purchased from ATCC, were maintained in RPMI 1640 or Dulbecco's modified Eagle's medium supplemented with 10% (v/v) heat-inactivated fetal bovine serum, 1% glutamine and 1 × antibiotics-antimycotics mixture (Invitrogen, Carlsbad, CA, USA). All cells were cultured at 37°C in a humidified incubator containing 5% CO2.

Chemicals

Bortezomib was obtained from the Pharmacy of Sir Run Run Shaw Hospital and dissolved in PBS at 5 mM as a stock solution. Proteasome inhibitor MG132 and ALLN were purchased from Calbiochem (La Jolla, CA, USA) and diluted in DMSO at stock concentrations of 10 and 20 mM, respectively. Cycloheximide and DMSO were purchased from Sigma (St Louis, MO, USA). Mcl-1 siRNA and negative control siRNA were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). The transfection of siRNA was performed using Oligofectamine (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's instructions.

Western blot analysis

Cells were lysed in Laemmli buffer after their respective treatments. Equal amounts of lysate were separated by 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis and evaluated by Western blot analysis as described previously [18]. Rabbit anti-human caspase-9, caspase-3, Bcl-2, Bcl-XL, and Mcl-1 antibodies were purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Mouse anti-human PARP antibodies were purchased from BD Pharmingen (San Diego, CA, USA). Mouse anti-human β-actin was obtained from Sigma.

Cell viability assay

The viability of the cell lines was determined by a sulforhodamine B colorimetric assay, as previously described [19]. Briefly, after fixation of adherent cells with trichloroacetic acid in a 96-well microplate, the protein was stained with sulforhodamine B, and the absorbance was determined at 570 nm to reflect the number of stained cells representing cell viability. The percentage of viable cells was determined relative to the cell viability of the PBS control, which was arbitrarily set as 1. Each experiment was performed in quadruplicate and repeated at least three times.

Protein stability assay

To determine protein stability, we treated cells with DMSO, MG132, bortezomib, or ALLN for up to 6 h and then added cycloheximide (25 μg/ml) to block protein synthesis [20]. Collected protein samples were subjected to Western blot analysis using anti-Mcl-1 antibody. Band densities were qualified using Optimas software (Media Cybernetics, Silver Spring, MD, USA), and the mean half-life of Mcl-1 was calculated.

Flow cytometry assay

Apoptosis was detected using an FITC Annexin-V Apoptosis Detection Kit (BD Pharmingen, San Diego, CA, USA) according to the manufacturer's instructions. The cells were digested with 0.25% trypsin, washed with cold phosphate-buffered saline (PBS) twice, and resuspended in binding buffer (1 × 106 cells/ml). Then 100 μl of the cell suspension (1 × 105 cells) was incubated with 5 μl of Annexin-V FITC and 5 μl of propidium iodide (PI) for 15 min at room temperature in the dark. The population of apoptosis cells was analyzed by flow cytometry (BD FACSCalibur, Becton Dickinson, San Jose, CA, USA).

Statistical analysis

The data were expressed as mean ± SD. Differences among the treatment groups were assessed via ANOVA using statistical software (Statsoft, Tulsa, OK). A P-value of ≤ 0.05 was considered significant. Survival was assessed using the Kaplan-Meier method.

Results

Rapid accumulation of Mcl-1 induced by proteasome inhibitors in various cancer cells

In this study, we evaluated the effect of proteasome inhibitors on Mcl-1 protein expression. Firstly, Cell lines which were owned by our lab including human colon cancer cell lines DLD1, LOVO, SW620, HCT116, human ovarian cancer cell line SKOV3 and human lung cancer cell line H1299 were treated with different concentrations of bortezomib (0.1-5.0 μM) for 6 h. Proteins were then collected and subjected to Western blot assay. The results showed that Mcl-1 expression was rapidly and dramatically upregulated by bortezomib in all six cell lines (Figure 1A), even at 0.1 μM. However, bortezomib did not substantially alter the expression levels of Bcl-2 and Bcl-XL (Figure 1A). We observed similar results about Mcl-1 when we used two other proteasome inhibitors, MG132 (Figure 1B) and calpain inhibitor I (ALLN) (Figure 1C). To evaluate whether the bortezomib could also induce Mcl-1 accumulation in other cancer cells besides the cell lines used as above, we chose the human breast cancer MCF7 cell line which was purchased from ATCC. However, Mcl-1 accumulation was not observed in this cell line (Figure 1D).
To investigate whether bortezomib treatment modifies Mcl-1 accumulation in cancer cells in a time-dependent manner, DLD1, LOVO and SKOV3 cells were treated with bortezomib (1 μM) for 2-6 h. Western blot results demonstrated that in these cells, the bortezomib-induced Mcl-1 accumulation was time-dependent. Its accumulation started within 2 h after treatment and became much stronger over time. We observed similar results when we used two other proteasome inhibitors, MG132 and ALLN (Figure 2).

Proteasome Inhibitors enhancement of Mcl-1 protein stability

Because proteasome inhibitors inhibit proteasome-mediated protein degradation, proteasome inhibitor-mediated Mcl-1 accumulation is likely to be caused by stabilization of the protein. To test this hypothesis, we treated DLD1 cells with dimethylsulfoxide (DMSO), 1 μM bortezomib, 5 μM MG132 or 20 μM ALLN for 6 h and then added cycloheximide to block protein synthesis in DLD1 cells [20]. Cells were then harvested over time and Mcl-1 levels were assessed by Western blot. Mcl-1 protein was rapidly degraded in cells treated with DMSO (Figure 3A) and had a mean half-life of less than 1 h. In contrast, in cells treated with bortezomib, MG132, or ALLN, the Mcl-1 protein level and mean half-life were stable, even after 6 h of cycloheximide treatment (Figure 3B). This result indicates that Mcl-1 degradation was blocked by treatment with proteasome Inhibitors.

Knockdown of Mcl-1 expression by siRNA enhanced MG132-induced cancer cell death

Next, we studied the relationship between Mcl-1 accumulation and cells' susceptibility to proteasome inhibitors. Although the endogenous expression of Mcl-1 varied, bortezomib induced the accumulation of Mcl-1 in various cancer cells. The folds of Mcl-1 accumulation in these cell lines varied from 1.59 to 11.09 (Figure 4). Simultaneously, we also determined the susceptibility of these cells to the treatment of bortezomib. The result demonstrated that the sensitivity of cancer cells to bortezomib differed (Table 1). These results demonstrate that the cells' susceptibility to bortezomib was not obviously associated with the amount of Mcl-1 accumulation (r = 0.781, P = 0.066).
Table 1
IC50 of Different Cell Lines after exposure to bortezomib
 
DLD1
LOVO
SKOV3
H1299
SW620
HCT116
bortezomib (nM)
22.5 ± 2.62
< 10
70.1 ± 14.4
25.6 ± 3.1
13.5 ± 1.9
17.7 ± 1.2
In order to further determine the role of Mcl-1 accumulation during proteasome inhibitor treatment, we used siRNA to knock down the expression of Mcl-1 protein. For this purpose, DLD1 cells were treated with 50 nM Mcl-1 or control siRNA for 24 h; then, cells were treated with 1 μM of MG132 for another 24 h. Levels of Mcl-1 protein were determined by Western blot assay. In comparison with DMSO or control siRNA, pretreatment with Mcl-1 siRNA dramatically reduced Mcl-1 protein levels. Moreover, although treatment with MG132 still resulted in obvious Mcl-1 accumulation in cells pretreated with Mcl-1 siRNA, the level of this accumulation was dramatically lower than in cells pretreated by control siRNA (Figure 5A and 5B, P < 0.01). This finding also suggests that MG132-mediated Mcl-1 accumulation was extremely efficient and could occur when the Mcl-1 level was very low. Cell viability analysis showed that treatment with DMSO, control or Mcl-1 siRNA alone did not lead to cell viability loss. Treatment with MG132 for 24 h led to significant viability loss. However, in comparison with cells pretreated with control siRNA, pretreatment with Mcl-1 siRNA significantly enhanced MG132-mediated cell death (P < 0.01, Figure 5C). A similar result was detected in the other colon cancer SW620 cell line (P < 0.01, Figure 5C). Thus, a reduced level of Mcl-1 accumulation correlates with increased cell death as a result of MG132.

Mcl-1 siRNA enhanced MG132-induced apoptotic signaling

Previous studies have shown that Mcl-1 is an anti-apoptotic protein that protects tumor cells against apoptosis. Thus, it is conceivable that the knockdown of Mcl-1 contributes to MG132-induced apoptosis. To confirm this hypothesis, DLD1 cells were treated with Mcl-1 siRNA and MG132 as described above. Levels of apoptosis were determined by Annexin-V FITC/PI assay (Figure 6A). The results showed that treatment with Mcl-1 siRNA plus MG132 resulted in a significantly higher apoptosis proportion (22.07 ± 3.44%) compared to control siRNA plus MG132 (12.22 ± 2.72%, P < 0.01); whereas Mcl-1 (4.37 ± 0.43%) or control siRNA alone (5.48 ± 0.38%) was no more effective than the DMSO (3.94 ± 0.45%, P > 0.05, Figure 6B).
Furthermore, we simultaneously evaluated the cleavage of several molecular markers of mitochondrial apoptotic signaling, including caspase-9, caspase-3, and poly (ADP-ribose) polymerase (PARP), by Western blot in DLD1 cells. The results showed that in cells with a knockdown of Mcl-1, cleavage of caspase-9, caspase-3, and PARP was dramatically enhanced after treatment with MG132 compared with control siRNA-treated cells (Figure 6C). Mcl-1 downregulation by siRNA alone did not exhibit any detectable effects compared with control siRNA or DMSO.

Discussion

Proteasomes play an essential role in degrading or processing intracellular proteins, some of which mediate cell cycle progression and apoptosis. Previous studies have shown that many types of actively proliferating malignant cells are more sensitive to proteasome blockade than non-cancerous cells [2]. Therefore, proteasome inhibitors are thought to be a novel class of anticancer drugs.
Proteasome inhibitors have a documented activity in a number of hematologic malignancies, especially in multiple myeloma and mantle cell lymphoma [21, 22]. However, despite encouraging preclinical data, studies in solid tumors have yielded disappointing results [2325]. Even in the treatment of multiple myeloma, the majority of patients do not respond, and resistance is common. The mechanism of proteasome inhibitor resistance is undefined.
Bcl-2 family members play a fundamental role in the regulation of apoptosis and are substrates of the proteasome. Previous studies implicated a role in the accumulation of pro-apoptotic Bcl-2 family members in proteasome inhibitor-induced apoptosis [16, 17]. Moreover, proteasome inhibitors may also upregulate the expression of antiapoptotic Bcl-2 family members [16]. We and others have reported that treatment with proteasome inhibitors does not affect the expression of Bcl-2 and Bcl-XL [10, 17, 26]. However, Mcl-1 differs from Bcl-2 and Bcl-XL because it is a short-lived molecule that is highly-regulated by ubiquitin proteasome pathway [16, 2729]. The ubiquitination of Mcl-1 is mediated by Mule-a BH3-only E3 ubiquitin ligase [30]. This process requires the association of Mcl-1 with Mule and is controlled by Noxa through the regulation of the Mcl/USP9X interaction [3032]. The level of Mcl-1/Mule complex would determine the sensitivity of cancer cells to apoptosis [33]. Therefore, Mcl-1 is likely an important survival molecule for regulating proteasome inhibitor-induced apoptosis.
In this study, we report a significant upregulation of Mcl-1 in lung cancer cell line H1299, the ovarian cancer cell line SKOV3, and the colon cancer cell lines DLD-1, LOVO, SW620 and HCT116 after treatment with different proteasome inhibitors. This effect is likely due to prolong half-life of Mcl-1. These results are similar with other previous studies, which showed that proteasome inhibitors upregulated Mcl-1 protein expression in melanoma and myeloma [16, 26, 34]. Previously, we had reported that proteasome inhibitors could induce Bik accumulation in various cancer cells [17]. Here we further reported that proteasome inhibitors could also induce Mcl-1 accumulation in these cells. Although both Bik and Mcl-1 protein were accumulated in these cells, they should play distinct role for cell survival. We had demonstrated that Bik accumulation induced by proteasome inhibitors might play a pro-apoptotic role in these cells [17]. Meanwhile, it had been reported that overexpressed Mcl-1 help malignant cells resistance to proteasome inhibitors [16]. Therefore, proteasome inhibitors-induced Mcl-1 in our cells may also interfere with its therapeutic effect [11, 35].
To further explore the role of Mcl-1 after treatment with proteasome inhibitor, we used RNA interference to knockdown Mcl-1 levels in DLD1 cells. Our results demonstrated that although the absolute value of difference between control siRNA+MG132 group and Mcl-1siRNA+MG132 group is not so large, Mcl-1 siRNA significantly increased the cytotoxicity of proteasome inhibitors (P < 0.01). Our data were consistent with studies on other tumor types, such as melanoma, myeloma and malignant pleural mesothelioma, in which the specific downregulation of Mcl-1 has been shown to sensitize cancer cells to proteasome inhibitor-induced apoptosis [16, 35, 36]. These data suggested that Mcl-1 could partial prevent cells from death. Base on these, we don't think that Mcl-1 increase following proteasome inhibitors treatment is an epiphenomenon without a functional meaning. These results also provide a molecular basis for a rational combination of proteasome inhibitors with a Mcl-1 antagonist, such as siRNA, UV light, or fludarabine [12, 16]. In the case of a potent cytotoxic with a restrictive side-effect profile [37], such as bortezomib, this combination strategy may also be effective using lower drug concentrations to avoid or minimize toxicities. Previous reports have shown that the knockdown of Mcl-1 significantly induced spontaneous apoptosis by its own [38, 39]. However, we did not find obvious cell death or apoptosis after the specific downregulation of Mcl-1 in DLD1 cells, suggesting that merely losing Mcl-1 expression may not be enough to induce apoptosis. The explanation for the differential effects of Mcl-1 knockdown on the survival of different cells is not entirely clear but might reflect different expression levels of other Bcl-2 family proteins related to Mcl-1 [14].

Conclusions

Our data showed that proteasome inhibitors induced not only Bik but also Mcl-1 accumulation in several cancer cell lines, particularly human colon cancer cell lines, and that this accumulation was mainly due to the stabilization of the Mcl-1 protein by proteasome inhibitors. The downregulation of Mcl-1 by Mcl-1 siRNA enhanced the apoptosis induced by proteasome inhibitors. Thus, using combined treatment with proteasome inhibitors and Mcl-1 antagonists may provide an effective and safe strategy for cancer therapy.

Acknowledgements

This work was supported by grants from the National Natural Science Foundation of China (30700970) and the Fundamental Research Funds for the Central Universities.
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

WZ performed experimental and statistical analysis and drafted the manuscript. JZH and HMT participated in flow cytometry and SRB assay. DW participated in Western blot analysis. XFH and CH participated in manuscript proofreading. HBZ conceived the design, provided financial support, participated Western blot analysis and revised the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Adams J: The development of proteasome inhibitors as anticancer drugs. Cancer Cell. 2004, 5 (5): 417-421. 10.1016/S1535-6108(04)00120-5.CrossRefPubMed Adams J: The development of proteasome inhibitors as anticancer drugs. Cancer Cell. 2004, 5 (5): 417-421. 10.1016/S1535-6108(04)00120-5.CrossRefPubMed
2.
Zurück zum Zitat Adams J: The proteasome: a suitable antineoplastic target. Nat Rev Cancer. 2004, 4 (5): 349-360. 10.1038/nrc1361.CrossRefPubMed Adams J: The proteasome: a suitable antineoplastic target. Nat Rev Cancer. 2004, 4 (5): 349-360. 10.1038/nrc1361.CrossRefPubMed
3.
Zurück zum Zitat Nussbaum AK, Dick TP, Keilholz W, Schirle M, Stevanovic S, Dietz K, Heinemeyer W, Groll M, Wolf DH, Huber R, et al: Cleavage motifs of the yeast 20S proteasome beta subunits deduced from digests of enolase 1. Proc Natl Acad Sci USA. 1998, 95 (21): 12504-12509. 10.1073/pnas.95.21.12504.CrossRefPubMedPubMedCentral Nussbaum AK, Dick TP, Keilholz W, Schirle M, Stevanovic S, Dietz K, Heinemeyer W, Groll M, Wolf DH, Huber R, et al: Cleavage motifs of the yeast 20S proteasome beta subunits deduced from digests of enolase 1. Proc Natl Acad Sci USA. 1998, 95 (21): 12504-12509. 10.1073/pnas.95.21.12504.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Karin M, Yamamoto Y, Wang QM: The IKK NF-kappa B system: a treasure trove for drug development. Nat Rev Drug Discov. 2004, 3 (1): 17-26. 10.1038/nrd1279.CrossRefPubMed Karin M, Yamamoto Y, Wang QM: The IKK NF-kappa B system: a treasure trove for drug development. Nat Rev Drug Discov. 2004, 3 (1): 17-26. 10.1038/nrd1279.CrossRefPubMed
5.
Zurück zum Zitat Orlowski RZ, Kuhn DJ: Proteasome inhibitors in cancer therapy: lessons from the first decade. Clin Cancer Res. 2008, 14 (6): 1649-1657. 10.1158/1078-0432.CCR-07-2218.CrossRefPubMed Orlowski RZ, Kuhn DJ: Proteasome inhibitors in cancer therapy: lessons from the first decade. Clin Cancer Res. 2008, 14 (6): 1649-1657. 10.1158/1078-0432.CCR-07-2218.CrossRefPubMed
6.
Zurück zum Zitat Voortman J, Checinska A, Giaccone G, Rodriguez JA, Kruyt FA: Bortezomib, but not cisplatin, induces mitochondria-dependent apoptosis accompanied by up-regulation of noxa in the non-small cell lung cancer cell line NCI-H460. Mol Cancer Ther. 2007, 6 (3): 1046-1053. 10.1158/1535-7163.MCT-06-0577.CrossRefPubMed Voortman J, Checinska A, Giaccone G, Rodriguez JA, Kruyt FA: Bortezomib, but not cisplatin, induces mitochondria-dependent apoptosis accompanied by up-regulation of noxa in the non-small cell lung cancer cell line NCI-H460. Mol Cancer Ther. 2007, 6 (3): 1046-1053. 10.1158/1535-7163.MCT-06-0577.CrossRefPubMed
7.
Zurück zum Zitat Yuan BZ, Chapman J, Reynolds SH: Proteasome inhibitors induce apoptosis in human lung cancer cells through a positive feedback mechanism and the subsequent Mcl-1 protein cleavage. Oncogene. 2009, 28 (43): 3775-3786. 10.1038/onc.2009.240.CrossRefPubMed Yuan BZ, Chapman J, Reynolds SH: Proteasome inhibitors induce apoptosis in human lung cancer cells through a positive feedback mechanism and the subsequent Mcl-1 protein cleavage. Oncogene. 2009, 28 (43): 3775-3786. 10.1038/onc.2009.240.CrossRefPubMed
8.
Zurück zum Zitat Fennell DA, Chacko A, Mutti L: BCL-2 family regulation by the 20S proteasome inhibitor bortezomib. Oncogene. 2008, 27 (9): 1189-1197. 10.1038/sj.onc.1210744.CrossRefPubMed Fennell DA, Chacko A, Mutti L: BCL-2 family regulation by the 20S proteasome inhibitor bortezomib. Oncogene. 2008, 27 (9): 1189-1197. 10.1038/sj.onc.1210744.CrossRefPubMed
9.
Zurück zum Zitat Yu J, Tiwari S, Steiner P, Zhang L: Differential apoptotic response to the proteasome inhibitor Bortezomib [VELCADE, PS-341] in Bax-deficient and p21-deficient colon cancer cells. Cancer Biol Ther. 2003, 2 (6): 694-699.CrossRefPubMed Yu J, Tiwari S, Steiner P, Zhang L: Differential apoptotic response to the proteasome inhibitor Bortezomib [VELCADE, PS-341] in Bax-deficient and p21-deficient colon cancer cells. Cancer Biol Ther. 2003, 2 (6): 694-699.CrossRefPubMed
10.
Zurück zum Zitat Nikrad M, Johnson T, Puthalalath H, Coultas L, Adams J, Kraft AS: The proteasome inhibitor bortezomib sensitizes cells to killing by death receptor ligand TRAIL via BH3-only proteins Bik and Bim. Mol Cancer Ther. 2005, 4 (3): 443-449.PubMed Nikrad M, Johnson T, Puthalalath H, Coultas L, Adams J, Kraft AS: The proteasome inhibitor bortezomib sensitizes cells to killing by death receptor ligand TRAIL via BH3-only proteins Bik and Bim. Mol Cancer Ther. 2005, 4 (3): 443-449.PubMed
11.
Zurück zum Zitat Perez-Galan P, Roue G, Villamor N, Montserrat E, Campo E, Colomer D: The proteasome inhibitor bortezomib induces apoptosis in mantle-cell lymphoma through generation of ROS and Noxa activation independent of p53 status. Blood. 2006, 107 (1): 257-264. 10.1182/blood-2005-05-2091.CrossRefPubMed Perez-Galan P, Roue G, Villamor N, Montserrat E, Campo E, Colomer D: The proteasome inhibitor bortezomib induces apoptosis in mantle-cell lymphoma through generation of ROS and Noxa activation independent of p53 status. Blood. 2006, 107 (1): 257-264. 10.1182/blood-2005-05-2091.CrossRefPubMed
12.
Zurück zum Zitat Nijhawan D, Fang M, Traer E, Zhong Q, Gao W, Du F, Wang X: Elimination of Mcl-1 is required for the initiation of apoptosis following ultraviolet irradiation. Genes Dev. 2003, 17 (12): 1475-1486. 10.1101/gad.1093903.CrossRefPubMedPubMedCentral Nijhawan D, Fang M, Traer E, Zhong Q, Gao W, Du F, Wang X: Elimination of Mcl-1 is required for the initiation of apoptosis following ultraviolet irradiation. Genes Dev. 2003, 17 (12): 1475-1486. 10.1101/gad.1093903.CrossRefPubMedPubMedCentral
13.
Zurück zum Zitat Kozopas KM, Yang T, Buchan HL, Zhou P, Craig RW: MCL1, a gene expressed in programmed myeloid cell differentiation, has sequence similarity to BCL2. Proc Natl Acad Sci USA. 1993, 90 (8): 3516-3520. 10.1073/pnas.90.8.3516.CrossRefPubMedPubMedCentral Kozopas KM, Yang T, Buchan HL, Zhou P, Craig RW: MCL1, a gene expressed in programmed myeloid cell differentiation, has sequence similarity to BCL2. Proc Natl Acad Sci USA. 1993, 90 (8): 3516-3520. 10.1073/pnas.90.8.3516.CrossRefPubMedPubMedCentral
14.
15.
Zurück zum Zitat Chen L, Willis SN, Wei A, Smith BJ, Fletcher JI, Hinds MG, Colman PM, Day CL, Adams JM, Huang DC: Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function. Mol Cell. 2005, 17 (3): 393-403. 10.1016/j.molcel.2004.12.030.CrossRefPubMed Chen L, Willis SN, Wei A, Smith BJ, Fletcher JI, Hinds MG, Colman PM, Day CL, Adams JM, Huang DC: Differential targeting of prosurvival Bcl-2 proteins by their BH3-only ligands allows complementary apoptotic function. Mol Cell. 2005, 17 (3): 393-403. 10.1016/j.molcel.2004.12.030.CrossRefPubMed
16.
Zurück zum Zitat Qin JZ, Xin H, Sitailo LA, Denning MF, Nickoloff BJ: Enhanced killing of melanoma cells by simultaneously targeting Mcl-1 and NOXA. Cancer Res. 2006, 66 (19): 9636-9645. 10.1158/0008-5472.CAN-06-0747.CrossRefPubMed Qin JZ, Xin H, Sitailo LA, Denning MF, Nickoloff BJ: Enhanced killing of melanoma cells by simultaneously targeting Mcl-1 and NOXA. Cancer Res. 2006, 66 (19): 9636-9645. 10.1158/0008-5472.CAN-06-0747.CrossRefPubMed
17.
Zurück zum Zitat Zhu H, Zhang L, Dong F, Guo W, Wu S, Teraishi F, Davis JJ, Chiao PJ, Fang B: Bik/NBK accumulation correlates with apoptosis-induction by bortezomib (PS-341, Velcade) and other proteasome inhibitors. Oncogene. 2005, 24 (31): 4993-4999. 10.1038/sj.onc.1208683.CrossRefPubMedPubMedCentral Zhu H, Zhang L, Dong F, Guo W, Wu S, Teraishi F, Davis JJ, Chiao PJ, Fang B: Bik/NBK accumulation correlates with apoptosis-induction by bortezomib (PS-341, Velcade) and other proteasome inhibitors. Oncogene. 2005, 24 (31): 4993-4999. 10.1038/sj.onc.1208683.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Wu S, Zhu H, Gu J, Zhang L, Teraishi F, Davis JJ, Jacob DA, Fang B: Induction of apoptosis and down-regulation of Bcl-XL in cancer cells by a novel small molecule, 2[[3-(2, 3-dichlorophenoxy)propyl]amino]ethanol. Cancer Res. 2004, 64 (3): 1110-1113. 10.1158/0008-5472.CAN-03-2790.CrossRefPubMed Wu S, Zhu H, Gu J, Zhang L, Teraishi F, Davis JJ, Jacob DA, Fang B: Induction of apoptosis and down-regulation of Bcl-XL in cancer cells by a novel small molecule, 2[[3-(2, 3-dichlorophenoxy)propyl]amino]ethanol. Cancer Res. 2004, 64 (3): 1110-1113. 10.1158/0008-5472.CAN-03-2790.CrossRefPubMed
19.
Zurück zum Zitat Pauwels B, Korst AE, de Pooter CM, Pattyn GG, Lambrechts HA, Baay MF, Lardon F, Vermorken JB: Comparison of the sulforhodamine B assay and the clonogenic assay for in vitro chemoradiation studies. Cancer Chemother Pharmacol. 2003, 51 (3): 221-226.PubMed Pauwels B, Korst AE, de Pooter CM, Pattyn GG, Lambrechts HA, Baay MF, Lardon F, Vermorken JB: Comparison of the sulforhodamine B assay and the clonogenic assay for in vitro chemoradiation studies. Cancer Chemother Pharmacol. 2003, 51 (3): 221-226.PubMed
20.
Zurück zum Zitat Kim GY, Mercer SE, Ewton DZ, Yan Z, Jin K, Friedman E: The stress-activated protein kinases p38 alpha and JNK1 stabilize p21(Cip1) by phosphorylation. J Biol Chem. 2002, 277 (33): 29792-29802. 10.1074/jbc.M201299200.CrossRefPubMed Kim GY, Mercer SE, Ewton DZ, Yan Z, Jin K, Friedman E: The stress-activated protein kinases p38 alpha and JNK1 stabilize p21(Cip1) by phosphorylation. J Biol Chem. 2002, 277 (33): 29792-29802. 10.1074/jbc.M201299200.CrossRefPubMed
21.
Zurück zum Zitat San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, Spicka I, Petrucci MT, Palumbo A, Samoilova OS, et al: Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med. 2008, 359 (9): 906-917. 10.1056/NEJMoa0801479.CrossRefPubMed San Miguel JF, Schlag R, Khuageva NK, Dimopoulos MA, Shpilberg O, Kropff M, Spicka I, Petrucci MT, Palumbo A, Samoilova OS, et al: Bortezomib plus melphalan and prednisone for initial treatment of multiple myeloma. N Engl J Med. 2008, 359 (9): 906-917. 10.1056/NEJMoa0801479.CrossRefPubMed
22.
Zurück zum Zitat Kane RC, Dagher R, Farrell A, Ko CW, Sridhara R, Justice R, Pazdur R: Bortezomib for the treatment of mantle cell lymphoma. Clin Cancer Res. 2007, 13 (18 Pt 1): 5291-5294.CrossRefPubMed Kane RC, Dagher R, Farrell A, Ko CW, Sridhara R, Justice R, Pazdur R: Bortezomib for the treatment of mantle cell lymphoma. Clin Cancer Res. 2007, 13 (18 Pt 1): 5291-5294.CrossRefPubMed
23.
Zurück zum Zitat Markovic SN, Geyer SM, Dawkins F, Sharfman W, Albertini M, Maples W, Fracasso PM, Fitch T, Lorusso P, Adjei AA, et al: A phase II study of bortezomib in the treatment of metastatic malignant melanoma. Cancer. 2005, 103 (12): 2584-2589. 10.1002/cncr.21108.CrossRefPubMed Markovic SN, Geyer SM, Dawkins F, Sharfman W, Albertini M, Maples W, Fracasso PM, Fitch T, Lorusso P, Adjei AA, et al: A phase II study of bortezomib in the treatment of metastatic malignant melanoma. Cancer. 2005, 103 (12): 2584-2589. 10.1002/cncr.21108.CrossRefPubMed
24.
Zurück zum Zitat Kondagunta GV, Drucker B, Schwartz L, Bacik J, Marion S, Russo P, Mazumdar M, Motzer RJ: Phase II trial of bortezomib for patients with advanced renal cell carcinoma. J Clin Oncol. 2004, 22 (18): 3720-3725. 10.1200/JCO.2004.10.155.CrossRefPubMed Kondagunta GV, Drucker B, Schwartz L, Bacik J, Marion S, Russo P, Mazumdar M, Motzer RJ: Phase II trial of bortezomib for patients with advanced renal cell carcinoma. J Clin Oncol. 2004, 22 (18): 3720-3725. 10.1200/JCO.2004.10.155.CrossRefPubMed
25.
Zurück zum Zitat Kozuch PS, Rocha-Lima CM, Dragovich T, Hochster H, O'Neil BH, Atiq OT, Pipas JM, Ryan DP, Lenz HJ: Bortezomib with or without irinotecan in relapsed or refractory colorectal cancer: results from a randomized phase II study. J Clin Oncol. 2008, 26 (14): 2320-2326. 10.1200/JCO.2007.14.0152.CrossRefPubMed Kozuch PS, Rocha-Lima CM, Dragovich T, Hochster H, O'Neil BH, Atiq OT, Pipas JM, Ryan DP, Lenz HJ: Bortezomib with or without irinotecan in relapsed or refractory colorectal cancer: results from a randomized phase II study. J Clin Oncol. 2008, 26 (14): 2320-2326. 10.1200/JCO.2007.14.0152.CrossRefPubMed
26.
Zurück zum Zitat Nencioni A, Hua F, Dillon CP, Yokoo R, Scheiermann C, Cardone MH, Barbieri E, Rocco I, Garuti A, Wesselborg S, et al: Evidence for a protective role of Mcl-1 in proteasome inhibitor-induced apoptosis. Blood. 2005, 105 (8): 3255-3262. 10.1182/blood-2004-10-3984.CrossRefPubMed Nencioni A, Hua F, Dillon CP, Yokoo R, Scheiermann C, Cardone MH, Barbieri E, Rocco I, Garuti A, Wesselborg S, et al: Evidence for a protective role of Mcl-1 in proteasome inhibitor-induced apoptosis. Blood. 2005, 105 (8): 3255-3262. 10.1182/blood-2004-10-3984.CrossRefPubMed
27.
Zurück zum Zitat Opferman JT, Letai A, Beard C, Sorcinelli MD, Ong CC, Korsmeyer SJ: Development and maintenance of B and T lymphocytes requires antiapoptotic MCL-1. Nature. 2003, 426 (6967): 671-676. 10.1038/nature02067.CrossRefPubMed Opferman JT, Letai A, Beard C, Sorcinelli MD, Ong CC, Korsmeyer SJ: Development and maintenance of B and T lymphocytes requires antiapoptotic MCL-1. Nature. 2003, 426 (6967): 671-676. 10.1038/nature02067.CrossRefPubMed
28.
Zurück zum Zitat Herrant M, Jacquel A, Marchetti S, Belhacene N, Colosetti P, Luciano F, Auberger P: Cleavage of Mcl-1 by caspases impaired its ability to counteract Bim-induced apoptosis. Oncogene. 2004, 23 (47): 7863-7873. 10.1038/sj.onc.1208069.CrossRefPubMed Herrant M, Jacquel A, Marchetti S, Belhacene N, Colosetti P, Luciano F, Auberger P: Cleavage of Mcl-1 by caspases impaired its ability to counteract Bim-induced apoptosis. Oncogene. 2004, 23 (47): 7863-7873. 10.1038/sj.onc.1208069.CrossRefPubMed
29.
Zurück zum Zitat Derouet M, Thomas L, Cross A, Moots RJ, Edwards SW: Granulocyte macrophage colony-stimulating factor signaling and proteasome inhibition delay neutrophil apoptosis by increasing the stability of Mcl-1. J Biol Chem. 2004, 279 (26): 26915-26921. 10.1074/jbc.M313875200.CrossRefPubMed Derouet M, Thomas L, Cross A, Moots RJ, Edwards SW: Granulocyte macrophage colony-stimulating factor signaling and proteasome inhibition delay neutrophil apoptosis by increasing the stability of Mcl-1. J Biol Chem. 2004, 279 (26): 26915-26921. 10.1074/jbc.M313875200.CrossRefPubMed
30.
Zurück zum Zitat Zhong Q, Gao W, Du F, Wang X: Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis. Cell. 2005, 121 (7): 1085-95. 10.1016/j.cell.2005.06.009.CrossRefPubMed Zhong Q, Gao W, Du F, Wang X: Mule/ARF-BP1, a BH3-only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl-1 and regulates apoptosis. Cell. 2005, 121 (7): 1085-95. 10.1016/j.cell.2005.06.009.CrossRefPubMed
31.
Zurück zum Zitat Warr MR, Acoca S, Liu Z, Germain M, Watson M, Blanchette M, Wing SS, Shore GC: BH3-ligand regulates access of MCL-1 to its E3 ligase. FEBS Lett. 2005, 579 (25): 5603-8.CrossRefPubMed Warr MR, Acoca S, Liu Z, Germain M, Watson M, Blanchette M, Wing SS, Shore GC: BH3-ligand regulates access of MCL-1 to its E3 ligase. FEBS Lett. 2005, 579 (25): 5603-8.CrossRefPubMed
32.
Zurück zum Zitat Gomez-Bougie P, Ménoret E, Juin P, Dousset C, Pellat-Deceunynck C, Amiot M: Noxa controls Mule-dependent Mcl-1 ubiquitination through the regulation of the Mcl-1/USP9X interaction. Biochem Biophys Res Commun. 2011 Gomez-Bougie P, Ménoret E, Juin P, Dousset C, Pellat-Deceunynck C, Amiot M: Noxa controls Mule-dependent Mcl-1 ubiquitination through the regulation of the Mcl-1/USP9X interaction. Biochem Biophys Res Commun. 2011
33.
Zurück zum Zitat Pervin S, Tran A, Tran L, Urman R, Braga M, Chaudhuri G, Singh R: Reduced association of anti-apoptotic protein Mcl-1 with E3 ligase Mule increases the stability of Mcl-1 in breast cancer cells. Br J Cancer. 2011, 105 (3): 428-37. 10.1038/bjc.2011.242.CrossRefPubMedPubMedCentral Pervin S, Tran A, Tran L, Urman R, Braga M, Chaudhuri G, Singh R: Reduced association of anti-apoptotic protein Mcl-1 with E3 ligase Mule increases the stability of Mcl-1 in breast cancer cells. Br J Cancer. 2011, 105 (3): 428-37. 10.1038/bjc.2011.242.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Qin JZ, Ziffra J, Stennett L, Bodner B, Bonish BK, Chaturvedi V, Bennett F, Pollock PM, Trent JM, Hendrix MJ, et al: Proteasome inhibitors trigger NOXA-mediated apoptosis in melanoma and myeloma cells. Cancer Res. 2005, 65 (14): 6282-6293. 10.1158/0008-5472.CAN-05-0676.CrossRefPubMed Qin JZ, Ziffra J, Stennett L, Bodner B, Bonish BK, Chaturvedi V, Bennett F, Pollock PM, Trent JM, Hendrix MJ, et al: Proteasome inhibitors trigger NOXA-mediated apoptosis in melanoma and myeloma cells. Cancer Res. 2005, 65 (14): 6282-6293. 10.1158/0008-5472.CAN-05-0676.CrossRefPubMed
35.
Zurück zum Zitat Gomez-Bougie P, Wuilleme-Toumi S, Menoret E, Trichet V, Robillard N, Philippe M, Bataille R, Amiot M: Noxa up-regulation and Mcl-1 cleavage are associated to apoptosis induction by bortezomib in multiple myeloma. Cancer Res. 2007, 67 (11): 5418-5424. 10.1158/0008-5472.CAN-06-4322.CrossRefPubMed Gomez-Bougie P, Wuilleme-Toumi S, Menoret E, Trichet V, Robillard N, Philippe M, Bataille R, Amiot M: Noxa up-regulation and Mcl-1 cleavage are associated to apoptosis induction by bortezomib in multiple myeloma. Cancer Res. 2007, 67 (11): 5418-5424. 10.1158/0008-5472.CAN-06-4322.CrossRefPubMed
36.
Zurück zum Zitat Yuan BZ, Chapman JA, Reynolds SH: Proteasome Inhibitor MG132 Induces Apoptosis and Inhibits Invasion of Human Malignant Pleural Mesothelioma Cells. Transl Oncol. 2008, 1 (3): 129-140.CrossRefPubMedPubMedCentral Yuan BZ, Chapman JA, Reynolds SH: Proteasome Inhibitor MG132 Induces Apoptosis and Inhibits Invasion of Human Malignant Pleural Mesothelioma Cells. Transl Oncol. 2008, 1 (3): 129-140.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Caravita T, de FP, Palumbo A, Amadori S, Boccadoro M: Bortezomib: efficacy comparisons in solid tumors and hematologic malignancies. Nat Clin Pract Oncol. 2006, 3 (7): 374-387.CrossRefPubMed Caravita T, de FP, Palumbo A, Amadori S, Boccadoro M: Bortezomib: efficacy comparisons in solid tumors and hematologic malignancies. Nat Clin Pract Oncol. 2006, 3 (7): 374-387.CrossRefPubMed
38.
Zurück zum Zitat Chetoui N, Sylla K, Gagnon-Houde JV, caide-Loridan C, Charron D, Al-Daccak R, Aoudjit F: Down-regulation of mcl-1 by small interfering RNA sensitizes resistant melanoma cells to fas-mediated apoptosis. Mol Cancer Res. 2008, 6 (1): 42-52. 10.1158/1541-7786.MCR-07-0080.CrossRefPubMed Chetoui N, Sylla K, Gagnon-Houde JV, caide-Loridan C, Charron D, Al-Daccak R, Aoudjit F: Down-regulation of mcl-1 by small interfering RNA sensitizes resistant melanoma cells to fas-mediated apoptosis. Mol Cancer Res. 2008, 6 (1): 42-52. 10.1158/1541-7786.MCR-07-0080.CrossRefPubMed
39.
Zurück zum Zitat Zhang B, Gojo I, Fenton RG: Myeloid cell factor-1 is a critical survival factor for multiple myeloma. Blood. 2002, 99 (6): 1885-1893. 10.1182/blood.V99.6.1885.CrossRefPubMed Zhang B, Gojo I, Fenton RG: Myeloid cell factor-1 is a critical survival factor for multiple myeloma. Blood. 2002, 99 (6): 1885-1893. 10.1182/blood.V99.6.1885.CrossRefPubMed
Metadaten
Titel
Small interfering RNA targeting mcl-1 enhances proteasome inhibitor-induced apoptosis in various solid malignant tumors
verfasst von
Wei Zhou
Jingzi Hu
Haimei Tang
Da Wang
Xuefeng Huang
Chao He
Hongbo Zhu
Publikationsdatum
01.12.2011
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2011
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/1471-2407-11-485

Weitere Artikel der Ausgabe 1/2011

BMC Cancer 1/2011 Zur Ausgabe

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

CUP-Syndrom: Künstliche Intelligenz kann Primärtumor finden

30.04.2024 Künstliche Intelligenz Nachrichten

Krebserkrankungen unbekannten Ursprungs (CUP) sind eine diagnostische Herausforderung. KI-Systeme können Pathologen dabei unterstützen, zytologische Bilder zu interpretieren, um den Primärtumor zu lokalisieren.

Sind Frauen die fähigeren Ärzte?

30.04.2024 Gendermedizin Nachrichten

Patienten, die von Ärztinnen behandelt werden, dürfen offenbar auf bessere Therapieergebnisse hoffen als Patienten von Ärzten. Besonders gilt das offenbar für weibliche Kranke, wie eine Studie zeigt.

Adjuvante Immuntherapie verlängert Leben bei RCC

25.04.2024 Nierenkarzinom Nachrichten

Nun gibt es auch Resultate zum Gesamtüberleben: Eine adjuvante Pembrolizumab-Therapie konnte in einer Phase-3-Studie das Leben von Menschen mit Nierenzellkarzinom deutlich verlängern. Die Sterberate war im Vergleich zu Placebo um 38% geringer.

Update Onkologie

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