Skip to main content
Erschienen in: Journal of Experimental & Clinical Cancer Research 1/2018

Open Access 01.12.2018 | Research

ERRα suppression enhances the cytotoxicity of the MEK inhibitor trametinib against colon cancer cells

verfasst von: Sheng Zhou, Hongwei Xia, Huanji Xu, Qiulin Tang, Yongzhan Nie, Qi yong Gong, Feng Bi

Erschienen in: Journal of Experimental & Clinical Cancer Research | Ausgabe 1/2018

Abstract

Background

ERRα, a constitutive transcription factor that regulates energy metabolism, plays an important role in the progression of various tumours. However, its role in cell survival and proliferation and its implication in targeted therapy in colon cancer remains elusive.

Methods

The expression of ERRα in colon cancer tissues and cell lines was detected by using western blotting and immunohistochemistry. A wound healing assay and a transwell assay were performed to examine the migration and invasion of the colon cancer cells. A cell viability assay, clonogenic assay, western blot assay and the dual-luciferase reporter assay were employed to study the interaction between trametinib (inhibitor of MEK) and EGF treatment. Flow cytometry, western blotting, quantitative reverse-transcription polymerase chain reaction and xenograft studies were used to identify whether the combination of trametinib and simvastatin had a synergistic effect.

Results

ERRα positively regulated the cell proliferation, migration and invasion of colon cancer cells, and the suppression of ERRα completely reduced the EGF treatment-induced proliferation of colon cancer cells. Further investigation showed that trametinib partially restrained the up-regulation of ERRα induced by the EGF treatment, and ERRα inhibition increased the sensitivity of colon cancer cells to trametinib. At last, we combined trametinib with simvastatin, a common clinically used drug with a new reported function of transcriptional activity inhibition of ERRα, and found that this combination produced a synergistic effect in inhibiting the proliferation and survival of colon cancer cells in vitro as well as in vivo.

Conclusions

The present data indicated that ERRα acted as an oncogene in colon cancer cells, and the combined targeting of ERRα and MEK might be a promising therapeutic strategy for colon cancer treatment.
Begleitmaterial
Additional file 2: Figure S2. Suppression of ERRα completely reduces the EGF treatment-induced cell proliferation and enhances the cytotoxicity of trametinib. a WB for ERRα, c-Myc, cyclin D1, pERK and ERK in the SW1116 cells treated with EGF (20/μl) at the indicated times (0.5, 2, 4, 6 and 8 h) in serum-free medium. b CCK-8 assay for the HCT116 and SW480 cells cultured with si-NC or si-ERRα (or/and 20 ng/μl EGF) for 3 d (* P< 0.05; ** P< 0.01; *** P< 0.001). The data are presented as the mean±SD of the experiments performed in triplicate. c WB for ERRα, c-Myc and cyclin D1 in the HCT116 and SW480 cells treated with si-NC or si-ERRα (or/and 20 ng/μl EGF) in serum-free medium for 48 h. d WB for pERK and ERK in the HCT116, SW480 and SW1116 cells treated with the indicated concentrations of trametinib (0–100 nM) or DMSO for 48 h. e WB for ERRα, c-Myc and cyclin D1 in the SW1116 cells treated with DMSO or 10 nM trametinib (or/and 20 ng/μl EGF) for 48 h. f CCK-8 assay for the HCT116 and SW480 cells treated with si-ERRα (or/and 50 nM trametinib) for 3 d. g, h, i WB for ERRα, IDH3A, c-Myc and Cyclin D1 in the HCT116, SW480 and SW1116 cells treated with si-ERRα (or/and 50 nM trametinib) for 2 d. j WB for ERRα in the HCT116 and SW1116 cells treated with cycloheximide (10 μg/ml) or trametinib combined with cycloheximide in a time-course experiment. k WB for ERRα in the HCT116 and SW1116 treated with trametinib (50 nM, 48 h) or DMSO supplemented with or without MG132 (10 μM) for 8 h. (PDF 898 kb)
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13046-018-0862-8) contains supplementary material, which is available to authorized users.
Sheng Zhou, Hongwei Xia and Huanji Xu contributed equally to this work.
Abkürzungen
AKT
Protein kinase B
CytC
Cytochrome c
EGF
Epidermal growth factor
EGFR
Epidermal growth factor receptor
ERRα
Oestrogen-related receptor α
FDA
Food and Drug Administration
HMGCR
3-hydroxy-3-Methylglutaryl-coenzyme A
IDH3A
Isocitrate Dehydrogenase 3 (NAD(+)) Alpha
MAPK
Extracellular signal regulated kinase
MEK
Mitogen-activated protein kinase
MG132
Proteasome inhibitor MG132
OS
Overall survival
PFS
Progression-free survival
PI3K
Phosphoinositide 3-kinase
RAS
Rat sarcoma
siRNA
Short interfering ribonucleic acid

Background

Colon cancer is the third most common and second deadliest malignancy in the word [1]. Chemotherapy and targeted therapy remain the key strategies for the treatment of the metastatic colon cancers. However, due to the mutation and over-expression of EGFR/RAS/BRAF, the abnormal activation of EGFR/RAS pathway occurs frequently in colon cancers and is associated with a poor prognosis and drug resistance [2, 3].
EGFR plays a critical role in the process of proliferation and differentiation in colon cancer cells. Activated EGFR constitutively activates multiple downstream pathways, including the RAS/MEK/ERK (MAPK-extracellular signal regulated kinase) and AKT/PI3K/mTOR pathways [4]. Various target drugs, including cetuximab, bevacizumab and regorafinib, are widely used in colon cancer and are involved in targeting the EGFR signalling molecules. However, their effects remain limited. A number of preclinical therapeutic strategies have been developed by combining EGFR pathway inhibitors with other target drugs in BRAF/KRAS mutant colon cancers [57]. However, none of these have been approved for clinical use because of safety issues or a lack of objective responses. Thus, it is urgent for us to develop more robust therapeutic approaches for the treatment of colon cancers.
Trametinib, a highly specific and potent MEK1/2 inhibitor, is approved by the Food and Drug Administration (FDA) for the treatment of BRAF-mutated metastatic melanoma. The dual inhibition of BRAF and MEK was tested in patients with metastatic BRAFV600E colon cancers but showed little efficacy [8].
The orphan nuclear hormone receptor, oestrogen-related receptor A (ERRα, NR3B1), is a constitutive transcription factor that is structurally and functionally related to the classic oestrogen receptors [9]. It interacts with and is modulated by members of the SRC and PGC-1 families of co-activators [1013]. Moreover, ERRα’s target genes include its own gene ESRRA [12], and it participates in the regulation of mitochondrial biogenesis and energy metabolism [1316]. ERRα plays an important role in the carcinogenesis of various tumours. A high expression of ERRα is globally associated with a poor prognosis in colon, endometrium, ovary, breast and prostate cancers [1721]. Previous studies have shown that the expression of ERRα is significantly up-regulated in colon cancer patients [18]. Additionally, ERRα also promotes cell migration and invasion [22, 23] and controls proliferation and tumourigenic capacity with energy metabolism in colon cancer cells [24]. These findings suggest that ERRα may be a potential biomarker in the progression of colon cancer.
Previous reports reveal that there are some links between the EGFR pathways and ERRα signalling [9, 25, 26]. The MEK/MAPK and PI3K/Akt signalling pathways regulate ERRα transcriptional activity and promote the malignant behaviour of breast cancer cells through increasing ERRα [25], while the overexpression of ERRα also negatively regulates ERK activation [27]. This interaction between ERRα and EGFR suggests a potential novel function of ERRα in the EGF-mediated survival and proliferation of colon cancer cells. Thus, targeting ERRα may be a potential novel therapeutic strategy to enhance the efficiency of EGFR signalling inhibition in colon cancer cells.
In this report, we showed that the suppression of ERRα completely reduced the EGF treatment- induced cell proliferation and survival in colon cancer cells. Furthermore, we found that trametinib partially restrained the up-regulation of ERRα induced by EGF exposure, and the inhibition of ERRα increased the sensitivity of colon cancer cells to trametinib. At last, we combined trametinib with simvastatin, a drug commonly used in the clinic, which has a new reported function of suppressing the transcriptional activity of ERRα [28], and the results showed that this combination synergised to inhibit proliferation and colony formation in vitro as well as the in vivo tumourigenic capacity of colon cancer cells.

Methods

Cell lines and culture

The human colon cells that were obtained from the State Key Laboratory of Biotherapy, West China Hospital, Sichuan University included HCT 116 (KRAS G13D), SW480 (KRAS G12 V) and SW1116 (KRASG12A) were grown in Dulbecco’s modified Eagle medium supplemented with 10% foetal bovine serum (FBS, Gibco, USA), 100 mU/mL penicillin, and 100 μg/mL streptomycin in a 5% CO2 atmosphere at 37 °C. All the cell lines used were negative for mycoplasma. Trametinib (GSK1120212), XCT790 (HY-10426) and CCCP (HY-100941) were from Medchemexpress. Simvastatin was purchased from J&K Scientific Ltd. (Beijing, China). These agents were all dissolved in dimethyl sulfoxide (DMSO). ERRα luciferase reporter plasmid (pGMERRα-Lu) was purchased from YESEN biology (Shanghai, China). Moreover, the following primary antibodies were obtained from Abcam: UK:rabbit anti-human c-Myc mAb and rabbit anti-human cyclin D1 mAb. The following antibodies were obtained from Santa Cruz: rabbit anti-human Bax mAb, mouse anti-human ERRα mAb, mouse anti-human IDH3A mAb and mouse anti-human GAPDH mAb.

Tissue samples

The human colon cancer tissue microarrays used in this study were prepared by Shanghai Outdo Biotech Co., Ltd. (Shanghai, China). All the patients signed informed consent forms. This study was approved by the Ethics Committee of Taizhou Hospital of Zhejiang Province.

Cell viability assay and Clonogenic assay

For the cell proliferation assays, the cells were seeded in 96-well plates for 24 h and were allowed to adhere overnight in regular growth media. After treatment with the indicated drugs, the relative cell growth was measured using the Cell Counting Kit-8 (Dojingdo, Kumamoto, Japan). For the clonogenic assays, the cells were seeded into 35-mm dishes and were cultured in Dulbecco’s modified Eagle medium with 10% foetal bovine serum and 100 IUml-1 penicillin/streptomycin overnight. The cells were then treated with the drug, as indicated, in complete media for 5–6 days. The growth media with or without drug were replaced every 2 days. The remaining cells were fixed with methanol (1%) and formaldehyde (1%), stained with 0.5% crystal violet, and photographed using a digital scanner. All the experiments were performed at least three times. Representative experiments are shown.

Transfection

The siRNAs against ERRα and the negative controls, the lentiviral shRNA expression vector targeting hERRα and scrambled control (pGPU6/GFP/Neo-shNC) were synthesized by GenePharma (Shanghai, China). ERRα luciferase reporter plasmid (pGMERRα-Lu) was purchased from YESEN biology (Shanghai, China) http://​www.​yeasen.​com/​index.​htm; The sequence of the shRNA/siRNA/pGMERRα-Lu sense was as follows: pGPU6/GFP/Neo-shERRα#1: 5’-CACCGTGGTGGGCATTGAGCCTCTCTACATTTCAAGAGAATGTAGAGAGGCTCAATGCCCACCATTTTTTG-3′ and pGPU6/GFP/Neo-shERRα#2: 5’-CACCGAATGCACTGGTGTCACATCTGCTGTTCAAGAGACAGCAGATGTGACACCAGTGCATTCTTTTTTG-3′; pGPU6/GFP/Neo-shNC:5'-CACCGTTCTCCGAACGTGTCACGTTTCAAGAGAACGTGACACGTTCGGAGAATTTTTTG-3′; siERRα: 5’-GAAUGCACUGGUGUCACAUCUGCUG-3′. The sequence of ERRα response element (32–91): GGCCTAACTGGCCGGTACCGCTAGCCTCGATAGCTTGAAGAGGTCACTGTGACCTACAACGAGCTTGAAGAGGTCACTGTGACCTACAACGGCGCGTAGA [29]; And the siRNAs, shRNAs and pGMERRα-Lu were transfected into the cells using Lipofectamine 2000 (Invitrogen/Life Sciences) according to the manufacturer’s instructions.

Immunohistochemistry

Immunohistochemistry (IHC) was performed on all colon cancer samples and the tissues of xenograft tumour using biotin-streptavidin HRP detection systems. Paraffin-embedded tissue sections were collected. After deparaffinization with xylene and dehydration in a graded alcohol series, the tissue sections were subjected to antigen retrieval by microwaving in sodium citrate buffer for 10 min and then inhibiting endogenous peroxidase activity. After nonspecific binding was blocked, the slides were incubated with ERRα (1:100) and IDH3A (1:200) antibody (Santa Cruz Biotechnology, CA, USA); c-Myc and Cyclin D1 antibody (1:200; Abcam, Cambridge, UK,) in phosphate-buffered saline (PBS) overnight at 4 °C in a humidified container. Biotinylated secondary antibodies (Zhongshan Golden Bridge Biotechnology Co. Ltd., China) were then used according to the manufacturer’s recommendations. The sections were incubated with HRP-streptavidin conjugates appropriate for detecting ERRα; IDH3A; c-Myc and Cyclin D1. The brown color indicative of peroxidase activity was developed by incubation with 0.1% 3,3-diaminobenzidine (Zhongshan Golden Bridge Biotechnology Co. Ltd. China) in distilled water for 1–3 min at room temperature. The appropriate positive and negative controls were included in each IHC assay.

Dual luciferase reporter gene assay

A dual luciferase reporter gene assay was performed by using a multifunctional microplate reader (Synergy H1, BioTek, Vermont, USA) and Dual-Luciferase® Report Assay System kit (TransGen Biotech, China). The following procedures were used: Luciferase Reaction buffer II was mixed with thawed Luciferase Reaction substrate II, placed in a centrifuge tube pre-wrapped in foil, and stored at − 80 °C. Thawing took place at room temperature in a dark environment. Stop & Glo buffer was thawed at room temperature and added to 50 x Stop & Glo substrate to prepare a 1 x Stop & Glo Reagent. The cell culture medium was discarded and the cells were washed twice with PBS. Any residual liquid was also removed before 100 uL of 1 x CLB lysis buffer (5 x CLB was diluted to 1× CLB with sterile water) was added into each well. The cells were lysed by shaking on a shaker for 15 min after which 20 uL of the cell lysate was drawn and added to a 96-well opaque detection plate. A total of 100 uL of LARII was the added quickly to the wells containing lysate and gently mixed. Cell lysate was detected on the multi-function microplate reader. Parameters were 10s of reading and 2–3 s delays. The firefly luciferase activity value (F) was measured in relative luminometer units (RFUs). After F was measured, the 96-well plate was immediately taken out of the multifunctional microplate reader and a 100 uL of 1 x Stop & Glo Reagent was added to each well and mixed evenly. The multifunctional microplate reader was used to measure RLUs of renilla luciferase activity (R) over 10s read periods and 2–3 s delays. The relative transcriptional activity of the promoter region was determined by the F/R ratio.

Transwell chamber migration assay

The cell migration assay was performed using a BD BioCoat™ Matrigel™ Invasion Chamber (BD Biosciences, San Jose, CA. The cells were photographed and counted in three random microscopic fields under a 10× objective to calculate the number of cells that migrated. The graph was plotted for the number of cells that invaded per microscopic field.

Scratch wounding migration assay

The cell migration ability was assessed by a scratch wound assay. The transfected cells were cultured in 6-well plates. When the cells reached 90% confluence, a scratch wound was created using a pipette tip. The wound edges were photographed with a Nikon Eclipse TE 2000-U (Nikon, Japan), and the scratch widths were analysed using ImageJ software (NIH). Three trials were used for each condition.

Western blot

The cells were lysed in RIPA buffer (150 mM NaCl, 1% NP-40, 50 mM Tris-HCl, PH 7.4, 1 mM phenylmethylsulfonyl fluoride, 1 μg/ml leupeptin, 1 mM deoxycholic acid and 1 mM EDTA) with protease inhibitors and phosphatase inhibitors (Calbiochem, Darmstadt, Germany). The protein concentration was determined by the Bradford protein assay kit (BioRad). The proteins were separated by SDS-PAGE and were immunoblotted and transferred to polyvinyl difluoride (PVDF) membranes (Millipore) according to standard protocols. Finally, we used the BioRad semidry transfer system to analyse the expression of the proteins, including ERRa, c-Myc, cyclin D1, Bax, and GAPDH.

Real-time quantitative polymerase chain reaction

The cells were collected in Trizol (Invitrogen, USA) for total RNA extraction as the manufacturer’s protocol instructed. Retrotranscription was performed with the Reverse Transcriptase M-MLV (Takara, Japan). The RT-PCR reactions were performed with a SYBR Premix Ex Taq™ kit (Takara,  Japan) on the iQ5 Real-Time PCR detection system (BioRad, Hercules, USA). The primers used were as follows: ERRa, forward: CACTATGGTGTGGCATCCTGT, reverse: CGTCTCCGCTTGGTGATCTC; IDH3A, forward: AGCCGGTCACCCATCTATGAA, reverse: CytC, forward: CAGTGCCACACCGTTGAAAA reverse: TGCATCGGTTATTTCACACTCC; cyclin D1, forward: GCTTCTGGTGAACAAGCTC, reverse: GTGGGCGTGCAGGCCAGACC; and c-Myc, forward: CAGCTGCTTAGACGCTGGATT, reverse: GTAGAAATACGGCTGCACCGA. The data were analysed using the 2^−ΔΔCT method.

Cell apoptosis analysis with flow cytometry

A flow-based Annexin V assay was used to measure cell apoptosis after treatment with the drugs. Briefly, the cells were treated with DMSO, trametinib, simvastatin, and trametinib plus simvastatin for 24 h. We used the Annexin V, FITC Apoptosis detection kit (Dojindo Molecular Technologies, Japan) to assess cell apoptosis. The cells were washed in PBS, resuspended in 500 μl of ANX-V binding buffer and were then stained with 5 μl of Annexin-V-fluorescein isothiocyanate (FITC) for 15 min on ice in the dark, according to the manufacturer’s instructions. Subsequent to the staining, the cells were incubated with 10 μl of propidium iodide (PI) for 5 min on ice in the dark. The analyses were performed using a Navios flow cytometer (Beckman Coulter).

Combination index evaluation

The drug interaction between simvastatin and trametinib was determined by the combination index (CI) value. The CI was evaluated by CompuSyn software (ComboSyn, Inc., Paramus, NJ), using the method proposed by Chou et al. [30]. CI valued < 1, =1, and > 1 indicate synergism, additive and antagonism effects, respectively.

In vivo xenograft experiment

Female BALB/c nude mice, 4–6 weeks old, were obtained from Dashuo (Chengdu, China). The mice (n = 6 per cell line per treatment group) were implanted subcutaneously with HCT116 cells (1.0 × 10^6 cells) in a 100 ul volume using a 23-gauge needle. Each mouse received two subcutaneous injections in the bilateral flank for the development of one tumour. Two weeks after implantation, the mice (n = 6 mice per cell line per treatment group) were assigned to one of four groups including PBS only, trametinib, simvastatin, or a combination of trametinib and simvastatin. The mice were treated daily orally with 1.5 mg/kg trametinib in PBS and/or daily orally with 5 mg/kg simvastatin dissolved in PBS. The tumour diameters were serially measured with a digital calliper (Proinsa, Vitoria, Spain) every 2–3 days, and the tumour volumes were calculated using the following formula: V = (L*W^2)/2, where L and W represent the length and width, respectively.

Statistical analysis

The data are expressed as the mean ± s.e.m. or the mean ± s.d. Each experiment was conducted at least three times with consistent results. The data were analysed using a two-tailed Student’s t-test by GraphPad Prism 5 (GraphPad Software). Significance is presented as a P-value of < 0.05 (*), < 0.01 (**) and < 0.001 (***); non-significant differences are presented as NS.

Results

ERRα suppression inhibits the growth of colon cancer cells

To investigate the expression of ERRα in colon cancer tissues, we randomly selected 12 pairs of colon cancer tissue samples for western blot analysis. The results showed that the expression of ERRα was higher in the carcinomatous tissues than in the distal normal tissues (Fig. 1a). Next, we detected ERRα expression by IHC from the pathological tissues of 66 colon cancer patients who had undergone tumour resection. As indicated in Fig. 1b, the expression levels of ERRα were significantly higher in colon tumour tissues than that in distal normal tissues. Unlike in normal tissues, colon tumour tissues also showed positive staining for ERRα in the nucleus. In the normal and cancer tissues, the mean immunoreactivity scores were 0.363 and 4.867 respectively. For ERRα, most of the tumour tissues immunoreactivity scores were 4–7(moderately staining) and 8–12 (highly staining) and the percentages are 41 and 29%, respectively, while majority of the adjacent normal tissues had a score of 0–3 (lowly staining), and the percentage is 98% (Fig. 1c). Then, we also studied the effect of ERRα suppression on the malignant phenotypes of colon cancer cells. The results revealed that the cellular growth and colony information were strongly inhibited in the HCT116 and SW480 cells with shERRα#1 or shERRα#2 transfection compared with the cells transfected with the control shRNA (Figs. 1d-e, 2e-f). To verify whether shERRα performs its inhibition function properly, we constructed a luciferase assay reporter system by transfected the ERRα luciferase reporter plasmid into SW480 cells. The luciferase activity of pGMERRα-Lu significantly decreased in the cells transfected with the shERRα#1 and shERRα#2 (Fig. 1f). We also evaluated whether the inhibition of ERRα activity by a ligand modulated cell proliferation and colony formation in colon cancer cells. XCT790, a potent and specific inverse agonist of ERRα, was used in this further study. The CCK8 assay showed that XCT790 treatment dramatically inhibited colon cell growth (Fig. 1g) and colony formation (Fig. 1h-i). A western blot analysis was used to test the effect of XCT790 treatment on the protein level of ERRα. As expected, colon cancer cells treated with XCT790 showed a reduced level of ERRα compared to the vector control (Additional file 1: Figure S1a). Moreover, XCT790 treatment decreased the expression of genes encoding hyperplasia proteins, including c-Myc and cyclin D1 (Fig. 1j). Then, we also found that the colon cancer cells transfected with si-ERRα displayed less migrated cells compared to the vector control in the transwell assay and wound healing assay (Additional file 1: Figure S1d-g). Collectively, the results suggest that ERRα is involved in the regulation of proliferation and migration of colon cancer cells and plays a role as an oncogene in colon cancer.

Suppression of ERRα completely reduces the EGF-induced cell proliferation and survival of colon cancer cells

EGFR plays a critical role in the regulation of cell proliferation and differentiation, and EGF is a crucial ligand of EGFR [31]. Here, we found that EGF up-regulated the expressions of ERRα, p-ERK and c-Myc in the HCT116, SW480 and SW1116 cell lines by western blot analysis (Fig. 2a, Additional file 2: Figure S2a). The functional studies revealed that the activated EGFR signalling also promoted cell proliferation, as demonstrated by the Cell Counting Kit-8 assay and colony formation. Further studies indicated that the inhibition of ERRα by shERRα#2, si-ERRα or XCT790 completely reversed the EGF treatment-induced cell proliferation (Fig. 2b-c, e-f, h-i, Additional file 2: Figure S2b) and the expressions of ERRα and c-Myc (Fig. 2g, j, Additional file 2: Figure S2c). In addition, qPCR analysis indicated that shERRα entirely reversed the up-regulation of ERRα, IDH3A [28] and CytC (ERRα’s downstream target) [32] induced by EGF treatment (Additional file 3: Figure S6a), and the up-regulation of luciferase activity of pGMERRα-Lu induced by EGF treatment was also significantly reversed in SW480 cells transfected with the shERRα#2 (Fig. 2d). Together, the above-mentioned data indicated that activated EGFR signalling acts by increasing ERRα to promote the proliferation and survival of colon cancer cells.

Suppression of ERRα enhances the antitumour property of trametinib in colon cancer cells

RAF-MEK-ERK (mitogen-activated protein kinase (MAPK) pathway) signalling is frequently activated in human cancers, resulting in an increase in cellular proliferation [23]. Trametinib, a specific MEK inhibitor, is clinically used in melanoma (Additional file 2: Figure S2d). Here, we found that trametinib inhibited cell growth (Fig. 3a) and decreased the expression of ERRα and its downstream target IDH3A (Fig. 3b). Further investigations indicated that trametinib partially reversed the elevated cell number and colony formation induced by EGF stimulation (Fig. 3c-e). QPCR analysis indicated that trametinib did not entirely reverse the up-regulation of ERRα, IDH3A and CytC induced by EGF treatment compared with the trametinib treatment alone (Additional file 3: Figure S6b), and western blot analysis also demonstrated that trametinib did not entirely reverse the up-regulation of ERRα, c-Myc and cyclin D1 induced by EGF treatment (Fig. 3f, Additional file 2: Figure S2e).
Therefore, we combined trametinib with shERRα, si-ERRα and XCT790 to investigate whether ERRα suppression enhances the cytotoxicity of trametinib against colon cancer. Our results showed that the combination was more effective at restraining cell proliferation (Fig. 3g, l, Additional file 2: Figure S2f) and colony formation (Fig. 3h-i, m-n). And the luciferase activity of pGMERRα-Lu was more significantly decreased in SW480 cells treated by combined trametinib and shERRα#2 (Fig. 3k). Western blot analysis also demonstrated that the combination inhibited ERRα, IDH3A, c-Myc and cyclin D1 more thoroughly compared to the single treatment (Fig. 3j, o, Additional file 2: Figure S2g-i). Furthermore, qPCR analyses also showed a substantial reduction in the ERRα and its downstream target genes IDH3A and CytC in the combination group (Additional file 3: Figure S6c). Although trametinib is an effective drug that suppresses the growth of colon cancer cells, it did not achieve adequate cytotoxicity and inhibit the over-expression of ERRα induced by EGF. This implies that the co-inhibition of ERRα and MEK achieved more efficiency.

Simvastatin decreases the transcriptional activity of ERRα in colon cancer cells

To determine whether FDA-approved inhibitors exist that block the activity of ERRα, we performed a literature review and found that statins and bisphosphonates inhibit the activity of ERRα by blocking its cholesterol modification. Cholesterol is identified as the first functional endogenous ERRα ligand, and it increases the transcriptional activity of ERRα, while statins lower intracellular sterol levels, thus attenuating ERRα transactivation [28]. In addition, we found that simvastatin decreased the expression of its downstream target IDH3A and proliferation-related genes, such as c-Myc and cyclin D1, in the HCT116 and SW480 cell lines (Fig. 4b). The functional studies revealed that simvastatin inhibited the proliferation and colony formation of colon cancer cells (Fig. 4a, c-d, Additional file 4: Figure S3a-c). Moreover, consistent with the effect of trametinib, simvastatin partially reversed the EGF treatment-induced proliferation (Fig. 4c-d, Additional file 4: Figure S3b-c). Taken together, our results indicated that simvastatin decreased the transcriptional activity of ERRα and inhibited tumour growth in colon cancer.

Anti-tumour effect of a combination of trametinib and simvastatin

The above-mentioned data indicated that simvastatin might strengthen the antitumour efficiency of trametinib by inhibiting the activity of ERRα. Further CCK8 assays indicated that simvastatin significantly enhanced the cytotoxicity of trametinib in the HCT 116 and SW480 cells (Fig. 5a). The colony formation assays revealed that simvastatin, combined with trametinib, inhibited cell survival more significantly than simvastatin or trametinib alone in the two colon cancer cell lines (Fig. 5b-c). Moreover, the flow cytometry assays showed that this combination also produced a combined activity, with regard to cell apoptosis in colon cancer cells (Fig. 5g-h). Western blot analysis demonstrated that simvastatin synergised with trametinib and dramatically reduced the expression of the IDH3A, the proliferation-related genes c-Myc and cyclin D1, and incresesd the pro-apoptotic gene Bax (Fig. 5e). Next, quantitative real-time PCR showed that the combination therapy strongly decreased the mRNA expression of ERRα and its downstream targets IDH3A, c-Myc, and cyclin D1 compared with the single drug in the HCT116 cells (Fig. 5d), and the similar results were also found in SW480 cells (Additional file 3: Figure S6d). Furthermore, the luciferase activity of pGMERRα-Lu was more potently decreased in SW480 cells when combined trametinib and simvastatin (Fig. 5f). To investigate the combined effects, we calculated the combination index (CI) values and the Fa values using CompuSyn software (ComboSyn, Inc., Paramus, NJ, USA). According to the method proposed by Chou et al., the combination index (CI) values of < 1, =1, > 1, indicate synergistic, additive and antagonistic effects, respectively[30] . The combination index (CI) values were 0.03 and 0.19 in the HCT116 and sw480 cells, respectively, indicating that the combined therapy produced a synergistic effect in the two cell lines (Additional file 5: Figure S4a).
To investigate the combined effect in vivo, we implanted HCT116 tumours in nude mice, and they were assigned to the following four groups: untreated control, trametinib, simvastatin, or a combination of trametinib and simvastatin. The combination group showed a statistically significant reduction in tumour volume and weight compared with the vehicle-treated controls or the monotherapy groups in the HCT116 xenografts (Fig. 5i-j). Next, we detected ERRα, IDH3A, c-Myc and Cyclin D1 expression by immunostaining pathological tissue sections of xenograft tumour. As indicated in Fig. 5k-l, the overall protein expression levels of ERRα, IDH3A, c-Myc and Cyclin D1 were significantly weaker in combination group. Furthermore, a western blot was preformed to investigate the expression of proliferative proteins in the lysate from the xenografts. In contrast to the monotherapy groups, a combination of trametinib and simvastatin significantly down-regulated the expressions of c-Myc and cyclin D1 (Additional file 5: Figure S4b). Altogether, our findings unveiled that trametinib, combined with simvastatin, produced synthetic lethality in vitro and in vivo.

Discussion

ERRα regulates multiple biosynthetic pathways involved in energy metabolism [15, 33]. Recently, increasing evidence supports a critical role for ERRα as a pro-tumourigenic factor, and the vast majority of studies show that high ERRα expression is correlated with a poor clinical outcome in endocrine-related cancers [19, 34, 35]. In colon cancer, ERRα expression is significantly up-regulated compared with adjacent normal colon tissues [18]. Notably, we verified a new insight into the pro-tumourigenic function of ERRα in colon cancer. In our study, shERRα and XCT790 (which acts as a superagonist of ERRα) were used to suppress the expression of ERRα. The results showed that ERRα was required for colon cancer cell growth in vitro, and silencing ERRα decreased the migration ability of the HCT116, SW480 and SW1116 cell lines, which was consistent with a previous study [22, 24]. Otherwise, XCT 790 is also a potent, fast-acting, mitochondrial uncoupler independent of its inhibition function of ERRα [36]. To explore whether XCT790 inhibits the cell growth and proliferation mainly by inhibiting ERRα activity, but independent of its disruption on the mitochondrial transmembrane electrochemical gradients. We used CCCP, a chemical mitochondrial uncoupler that could inhibit the mitochondrial respiration in our study [36], and found CCCP could not effectively suppress cell growth when taken alone, and combined with trametinib also has no synergistic effect on cell growth (Fig. 1k, Additional file 1: Figure S1b). And under the suppression of the mitochondrial respiration by CCCP, XCT790 could still significantly inhibit colon cancer cells growth (Fig. 1l, Additional file 1: Figure S1c), suggesting that XCT790 mainly acts through inhibiting ERRα activity to suppress cell growth and proliferation. Importantly, these effects are completely independent of its function of disrupting mitochondrial transmembrane electrochemical gradients. Furthermore, our study first found that the suppression of ERRα completely reduced the survival of EGF-treated colon cancer cells, though it has been known for many years that ERRα expression is regulated, in part, via the EGF signalling pathway. Thus, our data suggested that ERRα was an oncogene and acted as a novel target for colon cancer therapy. However, all the ERRα antagonists (DES, XCT790 and SR16388) are still in pre-clinical study.
The presence of the oncogenic BRAF/KRAS mutation excludes metastatic colon cancer patients from targeted therapies, leaving them with only chemotherapy or no treatment if the disease is chemorefractory. Additional target drugs to prolong the PFS (progression-free survival) and OS (overall survival) are limited in metastatic colon cancers, suggesting the need to target other pathways. Trametinib is a highly specific and potent MEK1/2 inhibitor that suppresses the activity of RAS/ERK signalling, which is expected to inhibit the growth of cancers with the RAS/BRAF mutation. However, due to drug resistance, trametinib has only been approved by the FDA, in combination with dabrafenib, for the treatment of BRAF-mutated metastatic melanoma and advanced non-small cell lung cancer.
In this study, we found that trametinib down-regulated ERRα gene expression and inhibited its transcriptional activity probably through posttranscriptional regulation, because the immunoblot analysis showed that trametinib rapidly accelerated the degradation rate of ERRα, and it was reversed by MG132 (Additional file 2: Figure S2j-k). Although trametinib is an effective medicine to suppress the growth of colon cancer cells, the expression of the ERRα was not completely suppressed by trametinib in the presence of the EGF. Our data showed that ERRα played a central role in the EGF-mediated growth of colon cancer cells; thus, we hypothesized that inhibiting ERRα may increase the sensitivity of colon cancer cells to trametinib. We combined trametinib and XCT790 or shERRα and found that suppressing ERRα increased the antitumour effect of trametinib. Therefore, a combination of trametinib and XCT790 might be a good choice for colon cancer treatment. However, XCT790 is not approved in clinical applications; thus, we need to find a safe and effective drug combined with trametinib to inhibit ERRα activity entirely.
Simvastatin, an oral lipid-lowering drug, is approved by the FDA. Many studies demonstrate its antitumour activity in several cancer types [3739]. Recently, cholesterol was identified as an endogenous ERRa agonist, and ERRa transcriptional activity is significantly enhanced by cholesterol and suppressed by statins [28]. Thus, we replaced XCT790 with simvastatin and found that this combination decreased ERRα expression completely and had a synergistic effect, inhibiting proliferation and colony formation in vitro as well as the in vivo tumourigenic capacity of colon cancer cells.
Furthermore, we detected the expression of HMGCR (3-hydroxy-3-Methylglutaryl -coenzyme A) in the tissues, and the results showed that the expression of HMGCR was also higher in carcinomatous tissues than that of the distal normal tissues in 12 pairs of colon cancer tissues (Additional file 6: Figure S5). HMGCR is a key enzyme in the mevalonate pathway in tissues, and its high expression may suggest a high concentration of produced cholesterol and high activities of ERRα. Thus, simvastatin, a HMG-CoA reductase inhibitor, synergised with trametinib, might be a good choice to inhibit the tumourigenic capacity of colon cancer cells.
It is known that various preclinical and therapeutic strategies using trametinib combined with another target drug in BRAF/KRAS mutant colon cancers were developed [40, 41]. However, none of these strategies are approved for clinical use due to safety issues or a lack of objective responses during clinical trials.

Conclusions

In our study, the results of in vitro and in vivo experiments demonstrate that the suppression of ERRα by simvastatin enhances the antitumour properties of trametinib in colon cancer cells. In addition, we provide a novel therapeutic strategy for colon cancer by combining trametinib and simvastatin to inhibit the ERRα signaling axis (Fig. 6a-b).

Acknowledgements

The authors would like to thank the technician Cui Yiyuan (Neuromolecular Research Laboratory of West China Hospital) and the technician Zhang Yi (Core Facility of West China Hospital) for their assistance with experiments and equipment in the pathological and RT-qPCR analyses.

Funding

National Natural Science Foundation of China (81621003); National key R&D Program of China (2016YFC1303200/2016YFC1303203); National Natural Science Foundation of China (81572731).

Availability of data and materials

The datasets supporting the findings of this study are included within the article.
The animals were handled according to the guidelines of the China Animal Welfare Legislation, as provided by the Committee on Ethics in the Care and Use of Laboratory Animals of Sichuan University West China Hospital. The human colon cancer tissues as provided by the Ethics Committee of Taizhou Hospital of Zhejiang Province.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Anhänge

Additional files

Additional file 2: Figure S2. Suppression of ERRα completely reduces the EGF treatment-induced cell proliferation and enhances the cytotoxicity of trametinib. a WB for ERRα, c-Myc, cyclin D1, pERK and ERK in the SW1116 cells treated with EGF (20/μl) at the indicated times (0.5, 2, 4, 6 and 8 h) in serum-free medium. b CCK-8 assay for the HCT116 and SW480 cells cultured with si-NC or si-ERRα (or/and 20 ng/μl EGF) for 3 d (* P< 0.05; ** P< 0.01; *** P< 0.001). The data are presented as the mean±SD of the experiments performed in triplicate. c WB for ERRα, c-Myc and cyclin D1 in the HCT116 and SW480 cells treated with si-NC or si-ERRα (or/and 20 ng/μl EGF) in serum-free medium for 48 h. d WB for pERK and ERK in the HCT116, SW480 and SW1116 cells treated with the indicated concentrations of trametinib (0–100 nM) or DMSO for 48 h. e WB for ERRα, c-Myc and cyclin D1 in the SW1116 cells treated with DMSO or 10 nM trametinib (or/and 20 ng/μl EGF) for 48 h. f CCK-8 assay for the HCT116 and SW480 cells treated with si-ERRα (or/and 50 nM trametinib) for 3 d. g, h, i WB for ERRα, IDH3A, c-Myc and Cyclin D1 in the HCT116, SW480 and SW1116 cells treated with si-ERRα (or/and 50 nM trametinib) for 2 d. j WB for ERRα in the HCT116 and SW1116 cells treated with cycloheximide (10 μg/ml) or trametinib combined with cycloheximide in a time-course experiment. k WB for ERRα in the HCT116 and SW1116 treated with trametinib (50 nM, 48 h) or DMSO supplemented with or without MG132 (10 μM) for 8 h. (PDF 898 kb)
Literatur
1.
Zurück zum Zitat Lucena-Cacace A, Otero-Albiol D, Jimenez-Garcia MP, Munoz-Galvan S, Carnero A. NAMPT is a potent oncogene in Colon Cancer progression that modulates Cancer stem cell properties and resistance to therapy through Sirt1 and PARP. Clin Cancer Res. 2018;24(5):1202–15.CrossRefPubMed Lucena-Cacace A, Otero-Albiol D, Jimenez-Garcia MP, Munoz-Galvan S, Carnero A. NAMPT is a potent oncogene in Colon Cancer progression that modulates Cancer stem cell properties and resistance to therapy through Sirt1 and PARP. Clin Cancer Res. 2018;24(5):1202–15.CrossRefPubMed
2.
Zurück zum Zitat Kopetz S, Desai J, Chan E, et al. Phase II pilot study of Vemurafenib in patients with metastatic BRAF-mutated colorectal Cancer. J Clin Oncol. 2015;33(34):4032–8.CrossRefPubMedPubMedCentral Kopetz S, Desai J, Chan E, et al. Phase II pilot study of Vemurafenib in patients with metastatic BRAF-mutated colorectal Cancer. J Clin Oncol. 2015;33(34):4032–8.CrossRefPubMedPubMedCentral
3.
Zurück zum Zitat Roth AD, Tejpar S, Delorenzi M, et al. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. J Clin Oncol. 2010;28(3):466–74.CrossRefPubMed Roth AD, Tejpar S, Delorenzi M, et al. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. J Clin Oncol. 2010;28(3):466–74.CrossRefPubMed
4.
Zurück zum Zitat Pek M, Yatim S, Chen Y, et al. Oncogenic KRAS-associated gene signature defines co-targeting of CDK4/6 and MEK as a viable therapeutic strategy in colorectal cancer. Oncogene. 2017;36(35):4975–86.CrossRefPubMed Pek M, Yatim S, Chen Y, et al. Oncogenic KRAS-associated gene signature defines co-targeting of CDK4/6 and MEK as a viable therapeutic strategy in colorectal cancer. Oncogene. 2017;36(35):4975–86.CrossRefPubMed
5.
Zurück zum Zitat She QB, Halilovic E, Ye Q, et al. 4E-BP1 is a key effector of the oncogenic activation of the AKT and ERK signaling pathways that integrates their function in tumours. Cancer Cell. 2010;18(1):39–51.CrossRefPubMedPubMedCentral She QB, Halilovic E, Ye Q, et al. 4E-BP1 is a key effector of the oncogenic activation of the AKT and ERK signaling pathways that integrates their function in tumours. Cancer Cell. 2010;18(1):39–51.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Zhang P, Kawakami H, Liu W, et al. Targeting CDK1 and MEK/ERK Overcomes Apoptotic Resistance in BRAF-Mutant Human Colorectal Cancer. Mol Cancer Res. 2018;16(3):378–89.CrossRefPubMed Zhang P, Kawakami H, Liu W, et al. Targeting CDK1 and MEK/ERK Overcomes Apoptotic Resistance in BRAF-Mutant Human Colorectal Cancer. Mol Cancer Res. 2018;16(3):378–89.CrossRefPubMed
7.
Zurück zum Zitat Corcoran RB, Cheng KA, Hata AN, et al. Synthetic lethal interaction of combined BCL-XL and MEK inhibition promotes tumour regressions in KRAS mutant cancer models. Cancer Cell. 2013;23(1):121–8.CrossRefPubMed Corcoran RB, Cheng KA, Hata AN, et al. Synthetic lethal interaction of combined BCL-XL and MEK inhibition promotes tumour regressions in KRAS mutant cancer models. Cancer Cell. 2013;23(1):121–8.CrossRefPubMed
8.
Zurück zum Zitat Corcoran RB, Atreya CE, Falchook GS, et al. Combined BRAF and MEK inhibition with Dabrafenib and Trametinib in BRAF V600-mutant colorectal Cancer. J Clin Oncol. 2015;33(34):4023–31.CrossRefPubMedPubMedCentral Corcoran RB, Atreya CE, Falchook GS, et al. Combined BRAF and MEK inhibition with Dabrafenib and Trametinib in BRAF V600-mutant colorectal Cancer. J Clin Oncol. 2015;33(34):4023–31.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Barry JB, Giguere V. Epidermal growth factor-induced signaling in breast cancer cells results in selective target gene activation by orphan nuclear receptor estrogen-related receptor alpha. Cancer Res. 2005;65(14):6120–9.CrossRefPubMed Barry JB, Giguere V. Epidermal growth factor-induced signaling in breast cancer cells results in selective target gene activation by orphan nuclear receptor estrogen-related receptor alpha. Cancer Res. 2005;65(14):6120–9.CrossRefPubMed
10.
Zurück zum Zitat Tremblay GB, Kunath T, Bergeron D, et al. Diethylstilbestrol regulates trophoblast stem cell differentiation as a ligand of orphan nuclear receptor ERR beta. Genes Dev. 2001;15(7):833–8.CrossRefPubMedPubMedCentral Tremblay GB, Kunath T, Bergeron D, et al. Diethylstilbestrol regulates trophoblast stem cell differentiation as a ligand of orphan nuclear receptor ERR beta. Genes Dev. 2001;15(7):833–8.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Huss JM, Kopp RP, Kelly DP. Peroxisome proliferator-activated receptor coactivator-1alpha (PGC-1alpha) coactivates the cardiac-enriched nuclear receptors estrogen-related receptor-alpha and -gamma. Identification of novel leucine-rich interaction motif within PGC-1alpha. J Biol Chem. 2002;277(43):40265–74.CrossRefPubMed Huss JM, Kopp RP, Kelly DP. Peroxisome proliferator-activated receptor coactivator-1alpha (PGC-1alpha) coactivates the cardiac-enriched nuclear receptors estrogen-related receptor-alpha and -gamma. Identification of novel leucine-rich interaction motif within PGC-1alpha. J Biol Chem. 2002;277(43):40265–74.CrossRefPubMed
12.
Zurück zum Zitat Laganiere J, Tremblay GB, Dufour CR, Giroux S, Rousseau F, Giguere V. A polymorphic autoregulatory hormone response element in the human estrogen-related receptor alpha (ERRalpha) promoter dictates peroxisome proliferator-activated receptor gamma coactivator-1alpha control of ERRalpha expression. J Biol Chem. 2004;279(18):18504–10.CrossRefPubMed Laganiere J, Tremblay GB, Dufour CR, Giroux S, Rousseau F, Giguere V. A polymorphic autoregulatory hormone response element in the human estrogen-related receptor alpha (ERRalpha) promoter dictates peroxisome proliferator-activated receptor gamma coactivator-1alpha control of ERRalpha expression. J Biol Chem. 2004;279(18):18504–10.CrossRefPubMed
13.
Zurück zum Zitat Carrier JC, Deblois G, Champigny C, Levy E, Giguere V. Estrogen-related receptor alpha (ERRalpha) is a transcriptional regulator of apolipoprotein A-IV and controls lipid handling in the intestine. J Biol Chem. 2004;279(50):52052–8.CrossRefPubMed Carrier JC, Deblois G, Champigny C, Levy E, Giguere V. Estrogen-related receptor alpha (ERRalpha) is a transcriptional regulator of apolipoprotein A-IV and controls lipid handling in the intestine. J Biol Chem. 2004;279(50):52052–8.CrossRefPubMed
14.
Zurück zum Zitat Yan M, Audet-Walsh E, Manteghi S, et al. Chronic AMPK activation via loss of FLCN induces functional beige adipose tissue through PGC-1alpha/ERRalpha. Genes Dev. 2016;(30):1034–46.CrossRefPubMedPubMedCentral Yan M, Audet-Walsh E, Manteghi S, et al. Chronic AMPK activation via loss of FLCN induces functional beige adipose tissue through PGC-1alpha/ERRalpha. Genes Dev. 2016;(30):1034–46.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Luo J, Sladek R, Carrier J, Bader JA, Richard D, Giguere V. Reduced fat mass in mice lacking orphan nuclear receptor estrogen-related receptor alpha. Mol Cell Biol. 2003;23(22):7947–56.CrossRefPubMedPubMedCentral Luo J, Sladek R, Carrier J, Bader JA, Richard D, Giguere V. Reduced fat mass in mice lacking orphan nuclear receptor estrogen-related receptor alpha. Mol Cell Biol. 2003;23(22):7947–56.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Schreiber SN, Emter R, Hock MB, et al. The estrogen-related receptor alpha (ERRalpha) functions in PPARgamma coactivator 1alpha (PGC-1alpha)-induced mitochondrial biogenesis. Proc Natl Acad Sci U S A. 2004;101(17):6472–7.CrossRefPubMedPubMedCentral Schreiber SN, Emter R, Hock MB, et al. The estrogen-related receptor alpha (ERRalpha) functions in PPARgamma coactivator 1alpha (PGC-1alpha)-induced mitochondrial biogenesis. Proc Natl Acad Sci U S A. 2004;101(17):6472–7.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Fujimoto J, Sato E. Clinical implication of estrogen-related receptor (ERR) expression in uterine endometrial cancers. J Steroid Biochem Mol Biol. 2009;116(1–2):71–5.CrossRefPubMed Fujimoto J, Sato E. Clinical implication of estrogen-related receptor (ERR) expression in uterine endometrial cancers. J Steroid Biochem Mol Biol. 2009;116(1–2):71–5.CrossRefPubMed
18.
Zurück zum Zitat Cavallini A, Notarnicola M, Giannini R, et al. Oestrogen receptor-related receptor alpha (ERRalpha) and oestrogen receptors (ERalpha and ERbeta) exhibit different gene expression in human colorectal tumour progression. Eur J Cancer. 2005;41(10):1487–94.CrossRefPubMed Cavallini A, Notarnicola M, Giannini R, et al. Oestrogen receptor-related receptor alpha (ERRalpha) and oestrogen receptors (ERalpha and ERbeta) exhibit different gene expression in human colorectal tumour progression. Eur J Cancer. 2005;41(10):1487–94.CrossRefPubMed
19.
Zurück zum Zitat Ariazi EA, Clark GM, Mertz JE. Estrogen-related receptor alpha and estrogen-related receptor gamma associate with unfavorable and favorable biomarkers, respectively, in human breast cancer. Cancer Res. 2002;62(22):6510–8.PubMed Ariazi EA, Clark GM, Mertz JE. Estrogen-related receptor alpha and estrogen-related receptor gamma associate with unfavorable and favorable biomarkers, respectively, in human breast cancer. Cancer Res. 2002;62(22):6510–8.PubMed
20.
Zurück zum Zitat Sun P, Sehouli J, Denkert C, et al. Expression of estrogen receptor-related receptors, a subfamily of orphan nuclear receptors, as new tumour biomarkers in ovarian cancer cells. J Mol Med (Berl). 2005;83(6):457–67.CrossRef Sun P, Sehouli J, Denkert C, et al. Expression of estrogen receptor-related receptors, a subfamily of orphan nuclear receptors, as new tumour biomarkers in ovarian cancer cells. J Mol Med (Berl). 2005;83(6):457–67.CrossRef
21.
Zurück zum Zitat Fujimura T, Takahashi S, Urano T, et al. Increased expression of estrogen-related receptor alpha (ERRalpha) is a negative prognostic predictor in human prostate cancer. Int J Cancer. 2007;120(11):2325–30.CrossRefPubMed Fujimura T, Takahashi S, Urano T, et al. Increased expression of estrogen-related receptor alpha (ERRalpha) is a negative prognostic predictor in human prostate cancer. Int J Cancer. 2007;120(11):2325–30.CrossRefPubMed
22.
Zurück zum Zitat Dwyer MA, Joseph JD, Wade HE, et al. WNT11 expression is induced by estrogen-related receptor alpha and beta-catenin and acts in an autocrine manner to increase cancer cell migration. Cancer Res. 2010;70(22):9298–308.CrossRefPubMedPubMedCentral Dwyer MA, Joseph JD, Wade HE, et al. WNT11 expression is induced by estrogen-related receptor alpha and beta-catenin and acts in an autocrine manner to increase cancer cell migration. Cancer Res. 2010;70(22):9298–308.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Lin L, Sabnis AJ, Chan E, et al. The hippo effector YAP promotes resistance to RAF- and MEK-targeted cancer therapies. Nat Genet. 2015;47(3):250–6.CrossRefPubMedPubMedCentral Lin L, Sabnis AJ, Chan E, et al. The hippo effector YAP promotes resistance to RAF- and MEK-targeted cancer therapies. Nat Genet. 2015;47(3):250–6.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Bernatchez G, Giroux V, Lassalle T, Carpentier AC, Rivard N, Carrier JC. ERRalpha metabolic nuclear receptor controls growth of colon cancer cells. Carcinogenesis. 2013;34(10):2253–61.CrossRefPubMed Bernatchez G, Giroux V, Lassalle T, Carpentier AC, Rivard N, Carrier JC. ERRalpha metabolic nuclear receptor controls growth of colon cancer cells. Carcinogenesis. 2013;34(10):2253–61.CrossRefPubMed
25.
Zurück zum Zitat Ariazi EA, Kraus RJ, Farrell ML, Jordan VC, Mertz JE. Estrogen-related receptor alpha1 transcriptional activities are regulated in part via the ErbB2/HER2 signaling pathway. Mol Cancer Res. 2007;5(1):71–85.CrossRefPubMed Ariazi EA, Kraus RJ, Farrell ML, Jordan VC, Mertz JE. Estrogen-related receptor alpha1 transcriptional activities are regulated in part via the ErbB2/HER2 signaling pathway. Mol Cancer Res. 2007;5(1):71–85.CrossRefPubMed
26.
Zurück zum Zitat Chang CY, Kazmin D, Jasper JS, Kunder R, Zuercher WJ, McDonnell DP. The metabolic regulator ERRalpha, a downstream target of HER2/IGF-1R, as a therapeutic target in breast cancer. Cancer Cell. 2011;20(4):500–10.CrossRefPubMedPubMedCentral Chang CY, Kazmin D, Jasper JS, Kunder R, Zuercher WJ, McDonnell DP. The metabolic regulator ERRalpha, a downstream target of HER2/IGF-1R, as a therapeutic target in breast cancer. Cancer Cell. 2011;20(4):500–10.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Murray J, Huss JM. Estrogen-related receptor alpha regulates skeletal myocyte differentiation via modulation of the ERK MAP kinase pathway. Am J Physiol Cell Physiol. 2011;301(3):C630–45.CrossRefPubMedPubMedCentral Murray J, Huss JM. Estrogen-related receptor alpha regulates skeletal myocyte differentiation via modulation of the ERK MAP kinase pathway. Am J Physiol Cell Physiol. 2011;301(3):C630–45.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Wei W, Schwaid AG, Wang X, et al. Ligand activation of ERRalpha by cholesterol mediates statin and bisphosphonate effects. Cell Metab. 2016;23(3):479–91.CrossRefPubMedPubMedCentral Wei W, Schwaid AG, Wang X, et al. Ligand activation of ERRalpha by cholesterol mediates statin and bisphosphonate effects. Cell Metab. 2016;23(3):479–91.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Sladek R, Bader JA, Giguere V. The orphan nuclear receptor estrogen-related receptor alpha is a transcriptional regulator of the human medium-chain acyl coenzyme a dehydrogenase gene. Mol Cell Biol. 1997;17(9):5400–9.CrossRefPubMedPubMedCentral Sladek R, Bader JA, Giguere V. The orphan nuclear receptor estrogen-related receptor alpha is a transcriptional regulator of the human medium-chain acyl coenzyme a dehydrogenase gene. Mol Cell Biol. 1997;17(9):5400–9.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzym Regul. 1984;22:27–55.CrossRef Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined effects of multiple drugs or enzyme inhibitors. Adv Enzym Regul. 1984;22:27–55.CrossRef
31.
Zurück zum Zitat Bodnar RJ. Epidermal growth factor and epidermal growth factor receptor: the yin and Yang in the treatment of cutaneous wounds and Cancer. Adv Wound Care (New Rochelle). 2013;2(1):24–9.CrossRef Bodnar RJ. Epidermal growth factor and epidermal growth factor receptor: the yin and Yang in the treatment of cutaneous wounds and Cancer. Adv Wound Care (New Rochelle). 2013;2(1):24–9.CrossRef
32.
Zurück zum Zitat Ha BG, Jung SS, Shon YH. Effects of proton beam irradiation on mitochondrial biogenesis in a human colorectal adenocarcinoma cell line. Int J Oncol. 2017;51(3):859–66.CrossRefPubMed Ha BG, Jung SS, Shon YH. Effects of proton beam irradiation on mitochondrial biogenesis in a human colorectal adenocarcinoma cell line. Int J Oncol. 2017;51(3):859–66.CrossRefPubMed
33.
Zurück zum Zitat Huss JM, Torra IP, Staels B, Giguere V, Kelly DP. Estrogen-related receptor alpha directs peroxisome proliferator-activated receptor alpha signaling in the transcriptional control of energy metabolism in cardiac and skeletal muscle. Mol Cell Biol. 2004;24(20):9079–91.CrossRefPubMedPubMedCentral Huss JM, Torra IP, Staels B, Giguere V, Kelly DP. Estrogen-related receptor alpha directs peroxisome proliferator-activated receptor alpha signaling in the transcriptional control of energy metabolism in cardiac and skeletal muscle. Mol Cell Biol. 2004;24(20):9079–91.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Stein RA, Chang CY, Kazmin DA, et al. Estrogen-related receptor alpha is critical for the growth of estrogen receptor-negative breast cancer. Cancer Res. 2008;68(21):8805–12.CrossRefPubMedPubMedCentral Stein RA, Chang CY, Kazmin DA, et al. Estrogen-related receptor alpha is critical for the growth of estrogen receptor-negative breast cancer. Cancer Res. 2008;68(21):8805–12.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Suzuki T, Miki Y, Moriya T, et al. Estrogen-related receptor alpha in human breast carcinoma as a potent prognostic factor. Cancer Res. 2004;64(13):4670–6.CrossRefPubMed Suzuki T, Miki Y, Moriya T, et al. Estrogen-related receptor alpha in human breast carcinoma as a potent prognostic factor. Cancer Res. 2004;64(13):4670–6.CrossRefPubMed
36.
Zurück zum Zitat Eskiocak B, Ali A, White MA. The estrogen-related receptor alpha inverse agonist XCT 790 is a nanomolar mitochondrial uncoupler. Biochemistry. 2014;53(29):4839–46.CrossRefPubMedPubMedCentral Eskiocak B, Ali A, White MA. The estrogen-related receptor alpha inverse agonist XCT 790 is a nanomolar mitochondrial uncoupler. Biochemistry. 2014;53(29):4839–46.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Mantha AJ, Hanson JE, Goss G, Lagarde AE, Lorimer IA, Dimitroulakos J. Targeting the mevalonate pathway inhibits the function of the epidermal growth factor receptor. Clin Cancer Res. 2005;11(6):2398–407.CrossRefPubMed Mantha AJ, Hanson JE, Goss G, Lagarde AE, Lorimer IA, Dimitroulakos J. Targeting the mevalonate pathway inhibits the function of the epidermal growth factor receptor. Clin Cancer Res. 2005;11(6):2398–407.CrossRefPubMed
38.
Zurück zum Zitat Dai Y, Khanna P, Chen S, Pei XY, Dent P, Grant S. Statins synergistically potentiate 7-hydroxystaurosporine (UCN-01) lethality in human leukemia and myeloma cells by disrupting Ras farnesylation and activation. Blood. 2007;109(10):4415–23.CrossRefPubMedPubMedCentral Dai Y, Khanna P, Chen S, Pei XY, Dent P, Grant S. Statins synergistically potentiate 7-hydroxystaurosporine (UCN-01) lethality in human leukemia and myeloma cells by disrupting Ras farnesylation and activation. Blood. 2007;109(10):4415–23.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Bil J, Zapala L, Nowis D, Jakobisiak M, Golab J. Statins potentiate cytostatic/cytotoxic activity of sorafenib but not sunitinib against tumour cell lines in vitro. Cancer Lett. 2010;288(1):57–67.CrossRefPubMed Bil J, Zapala L, Nowis D, Jakobisiak M, Golab J. Statins potentiate cytostatic/cytotoxic activity of sorafenib but not sunitinib against tumour cell lines in vitro. Cancer Lett. 2010;288(1):57–67.CrossRefPubMed
40.
Zurück zum Zitat Pang X, Liu M. A combination therapy for KRAS-mutant lung cancer by targeting synthetic lethal partners of mutant KRAS. Chin J Cancer. 2016;35(1):92.CrossRefPubMedPubMedCentral Pang X, Liu M. A combination therapy for KRAS-mutant lung cancer by targeting synthetic lethal partners of mutant KRAS. Chin J Cancer. 2016;35(1):92.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Geukes FM, Boogerd W, Blank CU, van Thienen JV, Haanen JB, Brandsma D. Clinical and radiological response of BRAF inhibition and MEK inhibition in patients with brain metastases from BRAF-mutated melanoma. Melanoma Res. 2018;28(2):126–33. Geukes FM, Boogerd W, Blank CU, van Thienen JV, Haanen JB, Brandsma D. Clinical and radiological response of BRAF inhibition and MEK inhibition in patients with brain metastases from BRAF-mutated melanoma. Melanoma Res. 2018;28(2):126–33.
Metadaten
Titel
ERRα suppression enhances the cytotoxicity of the MEK inhibitor trametinib against colon cancer cells
verfasst von
Sheng Zhou
Hongwei Xia
Huanji Xu
Qiulin Tang
Yongzhan Nie
Qi yong Gong
Feng Bi
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Journal of Experimental & Clinical Cancer Research / Ausgabe 1/2018
Elektronische ISSN: 1756-9966
DOI
https://doi.org/10.1186/s13046-018-0862-8

Weitere Artikel der Ausgabe 1/2018

Journal of Experimental & Clinical Cancer Research 1/2018 Zur Ausgabe

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Erhöhtes Risiko fürs Herz unter Checkpointhemmer-Therapie

28.05.2024 Nebenwirkungen der Krebstherapie Nachrichten

Kardiotoxische Nebenwirkungen einer Therapie mit Immuncheckpointhemmern mögen selten sein – wenn sie aber auftreten, wird es für Patienten oft lebensgefährlich. Voruntersuchung und Monitoring sind daher obligat.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Onkologie

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