Skip to main content
Erschienen in: Cancer Cell International 1/2019

Open Access 01.12.2019 | Primary research

RETRACTED ARTICLE: Silencing of TRIM11 suppresses the tumorigenicity of chordoma cells through improving the activity of PHLPP1/AKT

verfasst von: Bin Wang, Gang Wang, Qingfeng Wang, Ziqiang Zhu, Yunqing Wang, Kangwu Chen, Huilin Yang

Erschienen in: Cancer Cell International | Ausgabe 1/2019

Abstract

Background

Tripartite motif-containing protein 11 (TRIM11), a member of RING family of E3 ubiquitin ligases, is identified as an oncogene in certain human tumors. However, the detailed biological function of TRIM11 in chordoma is still unclear. The purpose of present research is to explore the role of TRIM11 in human chordoma cells.

Methods

TRIM11 was induced silencing and overexpression in human chordoma cells using RNA interference (RNAi) and lentiviral vector. qRT-PCR and western blot were used to determine gene expression in chordomas cells. Meanwhile, cell counting kit-8 (CCK-8) assay was used to examine the cell proliferation rate. Flow cytometry analysis was performed to quantify the cell apoptosis rate.

Results

We identified that TRIM11 was upregulated in chordomas tissues. Moreover, TRIM11 presented pro-proliferation and anti-apoptosis function in chordoma cells. Further, LY294002, a specific AKT inhibitor, was utilized to examine the connection between TRIM11 and AKT in human chordoma cells. Importantly, our findings elucidated that TRIM11 promoted the growth of chordoma cells and involved in AKT signaling. Much more importantly, knockdown of TRIM11 significantly upregulated the translation of PH domain leucine-rich repeats protein phosphatase 1 (PHLPP1), whereas did not affect its transcription. Results that obtained from co-immunoprecipitation (Co-IP) and ubiquitination assay demonstrated TRIM11 interacted with PHLPP1 and promoted its ubiquitination in chordoma cells. Moreover, overexpression of PHLPP1 inhibited the phosphorylation of AKT in human chordomas cells. These results suggested that TRIM11 mediated the post-translation modification of PHLPP1 and was a novel component in PHLPP1/AKT signaling pathway in human chordoma cells.

Conclusions

Taken together, the present research not only enhanced the understanding of TRIM11 but also indicated its potential target and signaling pathway in human chordoma cells.
Trial registration retrospectively registered.
Hinweise
Bin Wang and Gang Wang are co-first author
A Retraction Note to this article is available online at https://​doi.​org/​10.​1186/​s12935-023-03017-2.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12935-019-1007-7.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
TRIM11
tripartite motif-containing protein 11
PHLPP1
PH domain leucine-rich repeats protein phosphatase 1
Co-IP
co-immunoprecipitation
RNAi
RNA interference
cDNA
complementary DNA
CCK-8
cell counting kit-8
IHC
immunohistochemistry
H & E
hematoxylin and eosin
IACUC
Institutional Animal Care and Use Committee

Background

Chordomas are one of the aggressive malignancies, which arise from notochordal remnants along the spine and subsequently infiltrate the adjacent muscular, nervous and related joints [1, 2]. Due to the resistant of chordoma to conventional chemotherapies and radiotherapies, surgical resection is still the main option for its treatment [3]. However, the outcome is far from satisfactory. Worse more, the high recurrent of chordoma is associated with poor survival rate. The overall 5-year survival is less than 50% [4]. Therefore, the novel effective approaches for its treatment are urgently needed.
Tripartite motif-containing protein 11 (TRIM11), a member of E3 ubiquitin ligases, is reported as an oncogene in certain human tumors [5]. Overexpression of TRIM11 promotes cell proliferation and inhibits apoptosis in colon cancer cells [6]. Moreover, TRIM11 had improved the growth, migration and invasion of lung cancer cells [7]. Moreover, silencing of TRIM11 has suppressed the activation of the phosphatidylinositide 3-kinase/protein kinase B (PI3K/AKT) signaling pathway in hepatocellular and ovarian cancer cells, respectively [8, 9]. The PI3K/AKT signaling pathway plays a critical role in the pathogenies of human cancers, which is believed as a promising anti-cancer target [10, 11]. However, the detailed relationship between TRIM11 and AKT is still unclear in chordoma cells.
PH domain leucine-rich repeats protein phosphatase 1 (PHLPP1) belongs to a novel family of Ser/Thr protein phosphatases and serves as tumor suppressor in several cancers [12]. It has been confirmed that PHLPP1 suppresses the activity of PI3K/AKT signaling pathway through dephosphorylating [13]. By using UbiBrowser database, TRIM11 is predicted to interact with PHLPP1 and serves as its potential E3 ligase (http://​ubibrowser.​ncpsb.​org/​ubibrowser/​home/​index). However, the supporting evidence for this connection has not been explored in chordoma.
In the present research, we analyzed the biological function of TRIM11 in human chordoma cells. TRIM11 was induced knockdown and overexpression in chordoma cells using RNA interference (RNAi) and lentiviral vector. This research not only deepened the understanding of TRIM11 but also indicated its potential target and signaling pathway in human chordoma cells.

Methods

Human chordoma tissues

A total of 40 human chordoma and adjacent-matched noncancerous tissues were provided by the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China. All tissues were stored at − 80 °C after being snap-frozen in liquid nitrogen.

Cell culture

The cell lines used in this study (NPs; MUG-Chor1 and U-CH1) were purchased from cell bank of Shanghai biology institute (Shanghai, China). Cells were seeded in DMEM Medium (Trueline, USA) and were cultured in the incubator with the condition of 5% CO2, at 37 °C. The AKT inhibitor LY294002 (25 μmol/L; S1105, Selleck, USA) and MG132 (10 μmol/L; Selleck, USA) were dissolved in DMSO (D2650, Sigma, USA) and used to culture cells.

RNA extraction and qRT-PCR

TRIzol Reagent kit (Invitrogen, USA) was utilized to isolate total RNA from all samples. Then, the complementary DNA (cDNA) was synthesized by the cDNA synthesis kit (Fermentas, Canada) following the manufacturer’s introduction. The program of qRT-PCR was set as follows: 95 °C for 10 min followed by 40 cycles of 95 °C for 15 s, 60 °C for 45 s. 2−ΔΔCt method was used to calculate the gene relative expression and normalized to GAPDH. Each analysis was performed for three times. Primer sequences were listed in Additional file 1.

Western blot

Total proteins were isolated by using RIPA lysis buffer (JRDUN, Shanghai, China). Protein concentrations were determined by using BCA protein assay kit (Thermo Fisher, USA). The protein was fractionated through running on 10% SDS-PAGE and subsequently transferred the gel to a nitrocellulose membrane (Millipore, USA) for 12 h. Then, the membranes were probed with the primary antibodies at 4 °C overnight followed by secondary antibody anti-mouse IgG (Beyotime, China) at 37 °C for 60 min. The protein content was detected by chemiluminescence system (Tanon, China). The product information of primary antibodies was listed in Additional file 1: Table S1.

Knockdown and overexpression

Three short interfering RNAs (siRNA, 50 pmol) targeted to different regions of human TRIM11 (NM_145214.2) and a non-specific siRNA (siNC) were linked with Lentiviral vector (pLKO.1). Then, the recombined vectors (1 μg) transiently transfected in chordoma cells (2 × 105), respectively by Lipofectamine 2000 (Invitrogen, USA). The sequences information of siTRIM11 s was listed in Additional file 1: Table S2.
Meanwhile, pLVX-puro vector contained the full length of human TRIM11 or PHLPP1 (NM_194449.4) cDNA sequence (oeTRIM11 or oePHLPP1; 1 μg) was transiently transfected in chordoma cells (2 × 105), respectively. The mock plasmid (oeNC) was acted as negative control (oeNC). Analyses were established at 48 h after being transfected. Real-time PCR and Western blot analysis were performed at 48 h after transfection to assess knockdown efficiency.

Cell proliferation

The proliferation assay was performed by using the Cell Counting Kit-8 (CCK-8) assay kits (SAB, USA). In brief, Cells were cultured for 0, 24, 48 and 72 h and mixed with the CCK-8 solution (1:10) for each well. Then, cells were incubated for 1 h. The OD450 nm was measured by Microplate reader (Pulangxin, China). Triplicates independently assays were needed for each time point.

Flow cytometry

Briefly, Annexin V-fluorescein isothiocyanate (FITC) apoptosis detection kit (Beyotime, China) was utilized to examine the apoptosis rate of cells. All procedures were followed by the manufacturer’s instructions. Flow cytometer (BD, USA) were utilized to determine cells at 48 h after infection. Three replications were needed for each sample.

Immunohistochemistry (IHC)

The paraffin-embedded chordoma and adjacent matched tissues were cut into 3 μm slices. Then, the slices were deparaffinized in xylene and subsequently rehydrated in a series of graded alcohol dilutions. After that, antigen retrieval was performed through high pressure in sodium citrate solution (0.01 M) for 15 min. The slices were incubated in H2O2 (3%) solution to block the activity of endogenous peroxidase. Next, all samples were incubated with rabbit antibody to human TRIM11 antibody (Ab111694, Abcam, UK) overnight at 4 °C. Slides were then incubated with HRP at room temperature for 30 min. Then, all samples were stained by using DAB and re-stained by hematoxylin for 3 min. The images (magnification: 200×) were collected by using the microscope (ECLIPSE Ni, NIKON, Japan) and analyzed by the micro-image analysis system (DS-Ri2, NIKON, Japan).

Histology

The tumor were embedded in paraffin and sectioned. Serial 5-μm slices were stained with hematoxylin and eosin (H & E). The images (magnification: 200×) were collected by using the microscope (ECLIPSE Ni, NIKON, Japan) and analyzed by the micro-image analysis system (DS-Ri2, NIKON, Japan).

TUNEL

Tunel staining assay was conducted by using the commercial Tunel kit (11684817910, Roche, Switzerland). All procedures were followed by the instruction of manufacture. There replicates were needed for each sample.

Co-immunoprecipitation (Co-IP)

In brief, whole-cell extracts were isolated after transfection or stimulation with appropriate ligands. Then, all samples were incubated by the appropriate antibodies plus Protein A/G beads (Santa Cruz Biotechnology, USA) overnight. Beads were washed five times and separated by SDS-PAGE. Western blot was performed as indicated above.

Ubiquitination assay

U-CH1 cells that transfected with siNC or siTRIM11 were lysed by sonication in 1% SDS-containing radio immunoprecipitation assay buffer on ice. Then, Lysates were treated by Protein A/G PLUS-Agarose (Santa Cruz Biotechnology, USA) for 1 h. After that, each samples were incubated with the IgG (Proteintech, USA) overnight at 4 °C. Then, the nuclear pellet was collected by centrifugation at 3000 rpm for 5 min at 4 °C and subsequently washed by Protein A/G Plus-Agarose beads for four times. The purified proteins were run on 4–20% gradient SDS-PAGE. Anti-PHLPP1 antibody (Ab135957, Abcam, UK) and anti-Ubiquitin antibody (ab7780, Abcam, UK) antibody were used for immunoblotting.

Xenograft model

Experiment was performed in agreement with the independent ethics committee of the First Affiliated Hospital of Soochow University and was guided by the Institute’s guidelines for animal experiments. Moreover, this research was confirmed by the Institutional Animal Care and Use Committee (IACUC). U-CH1 cells that transfected with siNC or siTRIM11 (n = 5 × 106) were subcutaneously injected into the right flank of 4–6 week old nude mice (n = 6 for each group; Shanghai Laboratory Animal Company, China). Tumor length and width were determined every fourth day for a total of 40 days after injection. Tumor volume was calculated as length × (width2/2).

Statistical analysis

Statistical analyses were performed by using GraphPad Prism software Version 7.0 (CA, USA). All data represented as mean ± S.E.M from three independent experiments. For comparison between groups, statistical differences were tested with one-way analysis of variance (ANOVA), followed by a Sidak’s test for multiple comparisons. Student’s t test was used for unpaired comparison. Statistically significant was accepted as the p -value < 0.05.

Results

TRIM11 was upregulated in human chordoma tissues

In order to examine the expression profile of TRIM11, the location of TRIM11 was determined by performing immunohistochemical staining assay in human chordoma (Tumor, n = 20) and adjacent-matched bone tissues (Normal, n = 20). The normal tissues were functioned as negative control. As presented in Fig. 1, it was clearly identified that the positive area of TRIM11 in normal tissues was much lower than that of chordoma tissues. Interestingly, the content of TRIM11 was extremely overexpressed in certain chordoma tissues (n = 8). Therefore, our results indicated that the expression of TRIM11 showed heterogeneity in different human chordoma tissues.

Silencing and overexpression of TRIM11 in chordoma cells

Next, we examined the level of TRIM11 in human chordoma cells (MUG-Chor1, U-CH1) and nucleus pulposus cells (NPs). Clearly, the relative mRNA levels of TRIM11 showed no significant difference among all three cell lines (Fig. 2a). However, the protein contents of TRIM11 were upregulated in MUG-Chor1 or U-CH1 cells compared with that of NPs (Fig. 2b). Therefore, TRIM11 was induced silencing in MUG-Chor1 and U-CH1 cells.
Next, three short RNA interferences (siRNAs) targeting human gene TRIM11 (NM_145214.2) were synthesized (siTRIM11-1, siTRIM11-2 and siTRIM11-3) and subsequently transfected into MUG-Chor1 and U-CH1 cells respectively. Meanwhile, a non-specific siRNA served as negative control (siNC). Both the relative mRNA and protein contents of TRIM11 were remarkably inhibited by TRIM11 siRNAs (Fig. 2c, d).
Meanwhile, we also induced overexpression of TRIM11 in U-CH1 cells. The full length of TRIM11 cDNA was inserted into the lentiviral vector (pLVX-Puro; oeTRIM11). Then, the recombined vector was transfected into U-CH1 cells. The mock vector was treated as negative control (oeNC). Obviously, both the relative mRNA and protein level of TRIM11 were remarkably improved by oeTRIM11 in chordoma cells (Fig. 2e, f). Therefore, the oeTRIM11 transfected cells were chosen in the following analyses.

Silencing of TRIM11 suppressed the proliferation and promoted the apoptosis of chordoma cells

Next, we examined the proliferation rate of siTRIM11-1 or siTRIM11-2 transfected cells by performing CCK-8 assay. Clearly, the proliferation rate of MUG-Chor1 or U-CH1 cells was inhibited by siTRIM11-1 or siTRIM11-2 at 24 h after transfection and showed a stronger effect at 48 h in two cell lines (Fig. 3a, b). These results indicated that TRIM11 was a pro-proliferation factor in chordoma cells.
Moreover, we also analyzed the apoptosis profile of MUG-Chor1 or U-CH1 cells that transfected with siTRIM11-1 or siTRIM11-2. As presented in Fig. 3c, the apoptosis rates of siTRIM11 transfected cells (MUG-Chor1: 15.3% and 13.6%; U-CH1 10.6% and 10.8%) were extremely higher than that of siNC transfected cells (MUG-Chor1: 3.8%; U-CH1: 1.5%). Therefore, knockdown of TRIM11 promoted the apoptosis of chordoma cells. Taken together, all these results demonstrated TRIM11 was an anti-apoptosis factor in chordoma cells.
It has been confirmed that X‑linked inhibitor of apoptosis (XIAP) is a key apoptosis inhibitor [14]. Moreover, cleaved caspase-3 is well known as a biomarker for cell apoptosis [15]. In the present research, the protein content of XIAP was deeply reduced in siTRIM11-1 or siTRIM11-2 transfected cells. Meanwhile, the TRIM11 siRNAs significantly upregulated the protein content of cleaved caspase-3 in two chordoma cell lines. All these results indicated the similar results that knockdown of TRIM11 promoted the apoptosis of chordoma cells. Interestingly, the phosphorylation of AKT (p-AKT) was also deeply suppressed in siTRIM11-1 or siTRIM11-2 transfected cells.

LY294002 abolished the function of TRIM11 in chordoma cells

To further assess the relationship between TRIM11 and AKT in chordoma cells, the activity of endogenous AKT was inhibited by AKT inhibitor LY294002 in oeTRIM11 transfected cells.
Then, CCK-8 assay was used to examine the proliferation rate of oeTRIM11 transfected cells. These results illustrated that overexpression of TRIM11 accelerated the proliferation of chordoma cells. However, the proliferation rate of oeTRIM11 transfected cells was remarkably decreased in the presence of AKT inhibitor LY294002 (Fig. 4a).
As presented in Fig. 4b, overexpression of TRIM11 deeply suppressed the apoptosis of chordoma cells (rate: 0.6%), whereas this effect was significantly abolished in the presence of the AKT inhibitor LY294002 (rate: 9.2%). Moreover, the protein content of XIAP was promoted in oeTRIM11 transfected cells and deeply suppressed by the inhibitor LY294002. Meanwhile, the AKT inhibitor LY294002 was also significantly improved the content of cleaved caspase-3 in oeTRIM11 transfected cells (Fig. 4c).
Taken together, our findings demonstrated that the biological function of TRIM11 was abolished by the AKT inhibitor LY294002. Therefore, overexpression of TRIM11 promoted the phosphorylation of AKT in chordoma cells.

Silencing of TRIM11 promoted the translation of PHLPP1 in chordoma cells

We also examined the relative mRNA and protein levels of PHLPP1 in siTRIM11-1 or siTRIM11-2 transfected cells. Clearly, the relative mRNA level of PHLPP1 showed no significant difference between siNC and siTRIM11 transfected cells (Fig. 5a). Interestingly, the protein content of PHLPP1 was remarkably upregulated in siTRIM11 transfected cells (Fig. 5b). Therefore, these results suggested that silencing of TRIM11 contributed to the translation of PHLPP1, whereas didn’t affect its transcription in chordoma cells.

TRIM11 interacted with PHLPP1 in chordoma cells

To further examine the connection between TRIM11 and PHLPP1, co-immunoprecipitation (Co-IP) assay was performed to determine the interaction between TRIM11 and PHLPP1. As shown in Fig. 5c, it was easily identified that there was a strong interaction between TRIM11 and PHLPP1 in chordoma cells.

Silencing TRIM11 inhibited the ubiquitination of PHLPP1 in chordoma cells

Previous report has demonstrated that proteasome inhibitor MG132 induces the inhibition of the ubiquitin (Ub) -proteasome pathway in cancer cachexia [16]. At present research, we cultured the siNC or siTRIM11 transfected cells in the present of MG132. Interestingly, MG132 promoted the accumulation of PHLPP1 in siTRIM11 transfected cells (Fig. 5d). These findings elucidated that TRIM11 might involve in the ubiquitination of PHLPP1. Therefore, the ubiquitination assay was conducted to examine the level of Ub-PHLPP1 in siNC or siTRIM11 transfected cells respectively. Clearly, knockdown of TRIM11 deeply suppressed the content of Ub-PHLPP1 in siTRIM11 transfected cells (Fig. 5e). Therefore, TRIM11 might promote the ubiquitination of PHLPP1 to enhace its degradtion in chordoma cells.

Overexpression of PHLPP1 suppressed the function of TRIM11 in chordoma cells

To further analyze the function of PHLPP1 in chordoma cells, PHLPP1 was induced overexpression in chordoma cells. Clearly, both the relative mRNA and protein level of PHLPP1 were significantly upregulated in chordoma cells (Fig. 6a, b). Meanwhile, the oePHLPP1 vector was also transfected into oeTRIM11 cells. As presented in Fig. 6c, the apoptosis was significantly upregulated in oePHLPP1 transfected cells (rate: 10.7%). Interestingly, the apoptosis of double transfected cells (oeTRIM11 + oePHLPP1; rate: 5.9%) was lower than that of oePHLPP1 transfected cells. Moreover, overexpression of PHLPP1 deeply released the suppression of TRIM11 in the apoptosis of chordoma cells (rate: 0.3%). Importantly, the content of p-AKT was significantly inhibited in oePHLPP1 transfected cells. Moreover, overexpression of PHLPP1 deeply reduced the content of p-AKT in oeTRIM11 transfected cells (Fig. 6d).

Knockdown of TRIM11 reduced the tumorigenicity of chordoma cells in vivo

To examine the role of TRIM11 in tumor formation in vivo, we constructed chordoma model on nude mice. In this section, a total of 12 nude mice were randomly divided into two groups (siNC and siTRIM11). All mice were acclimated in separate cages for 1 week. Then, an equal number of U-CH1 cells (5 × 106) that transfected with siNC or siTRIM11 was subcutaneously inoculated in armpits of nude mice (n = 6 for each group).
Obviously, both of the siNC or siTRIM11 transfected cells were able to develop into tumor. However, our results indicated that the tumor volume and weight of siTRIM11 group was significantly reduced compared with that of siNC group (Fig. 7a, b). Moreover, histological examinations of the tumors were performed after resection. As presented in Fig. 7c, the tumor of siNC group identified a large number of dense and highly proliferative cancer cells. As for the tumor of siTRIM11 transfected cells, the tumor staining was significantly decreased. Moreover, the ratio of nuclear-to-cytoplasmic was lower than that of siNC group. All these results indicated a massive tumor cell death. Importantly, the results from TdT-mediated DUTP nick end labeling (TUNEL) staining also suggested the cell apoptosis rate of siTRIM11 tumor was much higher than that of siNC tumor (Fig. 7d).
We also examined the protein content of TRIM11 and PHLPP1 in different tumors. The content of TRIM11 was deeply downregulated in the tumor of siTRIM11 group. Meanwhile, PHLPP1 showed an opposite pattern as that of TRIM11 in siTRIM11 tumor. Taken together, all these results demonstrated that TRIM11 was the oncogene in the pathological process of chordoma in vivo.

Discussion

Chordoma is one of the deadly tumors that deeply threaten the quality of human life. Due to the limited outcome of the surgery resection, more attention has been focused on the molecular targeted therapy (MTT) in the treatment for chordoma [17]. In the present research, our results not only demonstrated that TRIM11 was an oncogene in the pathological process of chordoma but also indicated its potential value as a target in chordoma. These data deepened the understanding into the molecular basis of this deadly malignancy.
The PI3K/AKT signaling pathway plays an essential role in tumorigenesis. It has been confirmed that the phosphorylation of AKT is upregulated in chordoma tissues and cells [18]. Moreover, high content of p-AKT was associated with poor prognosis [19]. TRIM family members are closely related to the activity of PI3K/AKT signaling pathway in human tumor cells [20, 21]. In this research, our results indicated TRIM11 was positively correlated with p-AKT in chordoma cells. The AKT inhibitor LY294002 deeply abolished the function of TRIM11. Therefore, TRIM11 might promote the growth of chordoma cells through regulating AKT signaling pathway.
It has been reported that PHLPP inhibits the activity of PI3K/AKT pathway and served as the tumor suppressor [22]. Meanwhile, manipulation the expression of PHLPP1 is believed as a promising potential therapeutic approach in the treatment for chordoma [23]. In this study, overexpression of PHLPP1 promoted the apoptosis of chordoma cells and downregulated the level of p-AKT. Therefore, enhancing the activity of PHLPP1/AKT signaling pathway was promising approach in the treatment for chordoma.
E3 ubiquitin ligases have catalyzed the ubiquitination of many proteins, which plays an essential role in carcinogenesis [24]. At present research, it was the first time to provide evidences to confirm that TRIM11 interacted with PHLPP1 and mediated its ubiquitination in chordoma cells. Moreover, our results indicated that TRIM11 promoted the ubiquitination of PHLPP1, which might accelerate its degration in chordoma cells and subsequently release the suppression of PHLPP1 to p-AKT in chordoma cells. Hence, TRIM11 might be a novel component in PHLPP1/AKT signaling pathway. More importantly, our findings indicated the critical role of post-translational modification in the pathogenic process of chordoma.

Conclusions

In this study, we examined the biological function of TRIM11 in chordoma cells. Our research not only deepened the understanding of TRIM11 but also indicated its potential target and signaling pathway in chordoma cells.

Supplementary information

Supplementary information accompanies this paper at https://​doi.​org/​10.​1186/​s12935-019-1007-7.

Acknowledgements

We acknowledged the support given by the The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China.
This research was approved by the independent ethics committee of The First Affiliated Hospital of Soochow University and was conducted in accordance with the Declaration of Helsinki. All patients provided written informed consent.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
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.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge
Literatur
1.
Zurück zum Zitat Bergh P, Kindblom LG, Gunterberg B, Remotti F, Ryd W, Meis-Kindblom JM. Prognostic factors in chordoma of the sacrum and mobile spine: a study of 39 patients. Cancer. 2015;88(9):2122–34.CrossRef Bergh P, Kindblom LG, Gunterberg B, Remotti F, Ryd W, Meis-Kindblom JM. Prognostic factors in chordoma of the sacrum and mobile spine: a study of 39 patients. Cancer. 2015;88(9):2122–34.CrossRef
2.
Zurück zum Zitat Sun I, Guduk M, Gucyetmez B, Yapicier O, Pamir MN. Chordoma: immunohistochemical analysis of brachyury. Turk Neurosurg. 2018;28(2):174–8.PubMed Sun I, Guduk M, Gucyetmez B, Yapicier O, Pamir MN. Chordoma: immunohistochemical analysis of brachyury. Turk Neurosurg. 2018;28(2):174–8.PubMed
3.
Zurück zum Zitat Hug EB, Loredo LN, Slater JD, Devries A, Grove RI, Schaefer RA, Rosenberg AE, Slater JM. Proton radiation therapy for chordomas and chondrosarcomas of the skull base. J Neurosurg. 1999;91(3):432.CrossRefPubMed Hug EB, Loredo LN, Slater JD, Devries A, Grove RI, Schaefer RA, Rosenberg AE, Slater JM. Proton radiation therapy for chordomas and chondrosarcomas of the skull base. J Neurosurg. 1999;91(3):432.CrossRefPubMed
4.
Zurück zum Zitat Mcmaster ML, Goldstein AM, Bromley CM, Ishibe N, Parry DM. Chordoma: incidence and survival patterns in the United States, 1973–1995. Cancer Causes Control. 2001;12(1):1.CrossRefPubMed Mcmaster ML, Goldstein AM, Bromley CM, Ishibe N, Parry DM. Chordoma: incidence and survival patterns in the United States, 1973–1995. Cancer Causes Control. 2001;12(1):1.CrossRefPubMed
5.
Zurück zum Zitat Dhanendra T, Rajesh S. TRIM family proteins: emerging class of RING E3 ligases as regulator of NF-κB pathway. Biol Cell. 2015;107(1):22–40.CrossRef Dhanendra T, Rajesh S. TRIM family proteins: emerging class of RING E3 ligases as regulator of NF-κB pathway. Biol Cell. 2015;107(1):22–40.CrossRef
6.
Zurück zum Zitat Yin Y, Zhong J, Li SW, Li JZ, Zhou M, Chen Y, Sang Y, Liu L. TRIM11, a direct target of miR-24-3p, promotes cell proliferation and inhibits apoptosis in colon cancer. Oncotarget. 2016;7(52):86755.CrossRefPubMedPubMedCentral Yin Y, Zhong J, Li SW, Li JZ, Zhou M, Chen Y, Sang Y, Liu L. TRIM11, a direct target of miR-24-3p, promotes cell proliferation and inhibits apoptosis in colon cancer. Oncotarget. 2016;7(52):86755.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Wang X, Shi W, Shi H, Lu S, Wang K, Sun C, He J, Jin W, Lv X, Zou H. TRIM11 overexpression promotes proliferation, migration and invasion of lung cancer cells. J Exp Clin Cancer Res. 2016;35(1):100.CrossRefPubMedPubMedCentral Wang X, Shi W, Shi H, Lu S, Wang K, Sun C, He J, Jin W, Lv X, Zou H. TRIM11 overexpression promotes proliferation, migration and invasion of lung cancer cells. J Exp Clin Cancer Res. 2016;35(1):100.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Zhang Z, Xu C, Zhang X, Huang L, Zheng C, Chen H, Wang Y, Ju H, Yao Q. TRIM11 Upregulation contributes to proliferation, invasion, and EMT of hepatocellular carcinoma cells. Oncol Res. 2017;25(5):691.CrossRefPubMedPubMedCentral Zhang Z, Xu C, Zhang X, Huang L, Zheng C, Chen H, Wang Y, Ju H, Yao Q. TRIM11 Upregulation contributes to proliferation, invasion, and EMT of hepatocellular carcinoma cells. Oncol Res. 2017;25(5):691.CrossRefPubMedPubMedCentral
9.
11.
Zurück zum Zitat Wang Q, Chen X, Hay N. Akt as a target for cancer therapy: more is not always better (lessons from studies in mice). Br J Cancer. 2017;117(2):159.CrossRefPubMedPubMedCentral Wang Q, Chen X, Hay N. Akt as a target for cancer therapy: more is not always better (lessons from studies in mice). Br J Cancer. 2017;117(2):159.CrossRefPubMedPubMedCentral
12.
Zurück zum Zitat Dong L, Jin L, Tseng HY, Wang CY, Wilmott JS, Yosufi B, Yan XG, Jiang CC, Scolyer RA, Zhang XD. Oncogenic suppression of PHLPP1 in human melanoma. Oncogene. 2014;33(39):4756.CrossRefPubMed Dong L, Jin L, Tseng HY, Wang CY, Wilmott JS, Yosufi B, Yan XG, Jiang CC, Scolyer RA, Zhang XD. Oncogenic suppression of PHLPP1 in human melanoma. Oncogene. 2014;33(39):4756.CrossRefPubMed
13.
Zurück zum Zitat Yu Y, Dai M, Lu A, Yu E, Merlino G. PHLPP1 mediates melanoma metastasis suppression through repressing AKT2 activation. Oncogene. 2018;37(17):2225–36.CrossRefPubMedPubMedCentral Yu Y, Dai M, Lu A, Yu E, Merlino G. PHLPP1 mediates melanoma metastasis suppression through repressing AKT2 activation. Oncogene. 2018;37(17):2225–36.CrossRefPubMedPubMedCentral
14.
Zurück zum Zitat Yang WZ, Zhou H, Yan Y. XIAP underlies apoptosis resistance of renal cell carcinoma cells. Mol Med Rep. 2018;17(1):125–30.CrossRefPubMed Yang WZ, Zhou H, Yan Y. XIAP underlies apoptosis resistance of renal cell carcinoma cells. Mol Med Rep. 2018;17(1):125–30.CrossRefPubMed
15.
Zurück zum Zitat Liu PF, Hu YC, Kang BH, Tseng YK, Wu PC, Liang CC, Hou YY, Fu TY, Liou HH, Hsieh IC, et al. Expression levels of cleaved caspase-3 and caspase-3 in tumorigenesis and prognosis of oral tongue squamous cell carcinoma. PLoS ONE. 2017;12(7):e0180620.CrossRefPubMedPubMedCentral Liu PF, Hu YC, Kang BH, Tseng YK, Wu PC, Liang CC, Hou YY, Fu TY, Liou HH, Hsieh IC, et al. Expression levels of cleaved caspase-3 and caspase-3 in tumorigenesis and prognosis of oral tongue squamous cell carcinoma. PLoS ONE. 2017;12(7):e0180620.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Zhang L, Tang H, Kou Y, Li R, Zheng Y, Wang Q, Zhou X, Jin L. MG132-mediated inhibition of the ubiquitin-proteasome pathway ameliorates cancer cachexia. J Cancer Res Clin Oncol. 2013;139(7):1105–15.CrossRefPubMedPubMedCentral Zhang L, Tang H, Kou Y, Li R, Zheng Y, Wang Q, Zhou X, Jin L. MG132-mediated inhibition of the ubiquitin-proteasome pathway ameliorates cancer cachexia. J Cancer Res Clin Oncol. 2013;139(7):1105–15.CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Meng T, Jin J, Jiang C, Huang R, Yin H, Song D, Cheng L. Molecular targeted therapy in the treatment of chordoma: a systematic review. Front Oncol. 2019;9:30.CrossRefPubMedPubMedCentral Meng T, Jin J, Jiang C, Huang R, Yin H, Song D, Cheng L. Molecular targeted therapy in the treatment of chordoma: a systematic review. Front Oncol. 2019;9:30.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Liang C, Ma Y, Yong L, Yang C, Wang P, Liu X, Zhu B, Zhou H, Liu X, Liu Z. Y-box binding protein-1 promotes tumorigenesis and progression via the epidermal growth factor receptor/AKT pathway in spinal chordoma. Cancer Sci. 2019;110(1):166–79.CrossRefPubMed Liang C, Ma Y, Yong L, Yang C, Wang P, Liu X, Zhu B, Zhou H, Liu X, Liu Z. Y-box binding protein-1 promotes tumorigenesis and progression via the epidermal growth factor receptor/AKT pathway in spinal chordoma. Cancer Sci. 2019;110(1):166–79.CrossRefPubMed
19.
Zurück zum Zitat Jr AS. Tyrosine kinase receptor expression in chordomas: phosphorylated AKT correlates inversely with outcome. Hum Pathol. 2013;44(9):1747–55. Jr AS. Tyrosine kinase receptor expression in chordomas: phosphorylated AKT correlates inversely with outcome. Hum Pathol. 2013;44(9):1747–55.
20.
Zurück zum Zitat Tang SL, Gao YL, Wen-Zhong H. Knockdown of TRIM37 suppresses the proliferation, migration and invasion of glioma cells through the inactivation of PI3K/Akt signaling pathway. Biomed Pharmacother. 2018;99:59–64.CrossRefPubMed Tang SL, Gao YL, Wen-Zhong H. Knockdown of TRIM37 suppresses the proliferation, migration and invasion of glioma cells through the inactivation of PI3K/Akt signaling pathway. Biomed Pharmacother. 2018;99:59–64.CrossRefPubMed
21.
23.
Zurück zum Zitat Chen H, Zhang K, Wu G, Song D, Chen K, Yang H. Low expression of PHLPP1 in sacral chordoma and its association with poor prognosis. Int J Clin Exp Pathol. 2015;8(11):14741.PubMedPubMedCentral Chen H, Zhang K, Wu G, Song D, Chen K, Yang H. Low expression of PHLPP1 in sacral chordoma and its association with poor prognosis. Int J Clin Exp Pathol. 2015;8(11):14741.PubMedPubMedCentral
24.
Zurück zum Zitat Lucy G, Tim E, Maher ER. VHL, the story of a tumour suppressor gene. Nat Rev Cancer. 2015;15(1):55–64.CrossRef Lucy G, Tim E, Maher ER. VHL, the story of a tumour suppressor gene. Nat Rev Cancer. 2015;15(1):55–64.CrossRef
Metadaten
Titel
RETRACTED ARTICLE: Silencing of TRIM11 suppresses the tumorigenicity of chordoma cells through improving the activity of PHLPP1/AKT
verfasst von
Bin Wang
Gang Wang
Qingfeng Wang
Ziqiang Zhu
Yunqing Wang
Kangwu Chen
Huilin Yang
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
Cancer Cell International / Ausgabe 1/2019
Elektronische ISSN: 1475-2867
DOI
https://doi.org/10.1186/s12935-019-1007-7

Weitere Artikel der Ausgabe 1/2019

Cancer Cell International 1/2019 Zur Ausgabe

Update Onkologie

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