SMAD specific E3 ubiquitin protein ligase 1 promotes ovarian cancer cell migration and invasion via the activation of the RhoA/ROCK signaling pathway

  • Authors:
    • Wei Wang
    • Han Du
    • Huiping Liu
    • Fangfang Hu
    • Guangzhi Liu
  • View Affiliations

  • Published online on: October 31, 2018     https://doi.org/10.3892/or.2018.6836
  • Pages: 668-676
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

SMAD specific E3 ubiquitin protein ligase 1 (SMURF1) serves a pivotal role in a variety of pathological processes and in tumor cell migration and invasion; however, its functional mechanism in ovarian cancer (OC) remains unknown. Previously, we observed overexpression of SMURF1 in OC tissues. In the present study, the role of SMURF1 in OC metastasis was investigated. The results revealed that SMURF1 was upregulated in OC cell lines of greater aggression than less aggressive cells. Downregulation of SMURF1 significantly inhibited OC cell invasion and migration, whereas upregulation of SMURF1 promoted OC cell invasion and migration. Investigation of the mechanism underlying the effects of SMURF1 in OC revealed that SMURF1 induced OC cell migration and invasion via activation of the Ras homolog family member A/Rho‑associated protein kinase signaling pathway. Further analysis demonstrated that higher levels of SMURF1 expression were associated with shorter overall survival in patients with OC. The findings of the present study indicated that overexpression of SMURF1 may contribute to the malignancy and metastasis of OC. The inhibition of SMURF1 expression may be a promising strategy for the treatment of patients with OC.

Introduction

Ovarian cancer (OC) is the leading cause of mortality associated with gynecological malignancies worldwide (1). Frequent metastasis is responsible for the rapid recurrence and poor survival associated with OC (2,3). Therefore, identifying molecular markers involved in the progression of OC metastasis may provide potential targets for the treatment of OC.

SMAD specific E3 ubiquitin protein ligase 1 (SMURF1), a member of the homologous to the E6-AP carboxyl terminus (HECT) family of E3 ubiquitin ligases, participates in a variety of physiological and pathological processes, including cell motility, cell proliferation and inflammatory responses (4,5). Recently, aberrant SMURF1 expression has been associated with the development of numerous cancers (68). There is increasing interest in the prominent role that SMURF1 serves in promoting recurrent tumor metastasis (9,10). SMURF1 expression has been reported to be upregulated in breast cancer (7,11). Inhibition of SMURF1 expression decreased breast cancer cell invasion and migration (7,11). In prostate cancer, SMURF1-mediated regulation of the tumor suppressor ovarian carcinoma 2/disabled homolog 2 interacting protein (DAB2IP) was reported to control tumor cell proliferation and migration (12). Elevated expression levels of ubiquitin ligase E3 genes, including SMURF1, SMURF2 and WW domain containing E3 ubiquitin protein ligase may underlie the mechanisms of occurrence, development and metastasis of prostate cancer (13). In pancreatic cancer, SMURF1 amplification has been detected in primary human pancreatic cancer tissues (14). Knockdown of SMURF1 in a pancreatic cancer cell line (AsPC-1) with focal amplification did not alter cell growth, but led to reduced cell invasion (14). The results from these studies indicated that SMURF1 may serve a role in cancer cell migration and invasion; however, the underlying pathophysiological mechanisms contributing to the progression of OC metastasis remains unknown. Therefore, the present study aimed to determine whether SMURF1 was involved in the development of OC metastasis.

Materials and methods

Patient samples

A total of 80 ovarian serous cystadenocarcinoma samples were collected from patients with age from 49 to 71 years between May 2007 and December 2013 at the Department of Obstetrics and Gynecology, the People's Hospital of Zhengzhou University (Zhengzhou, China). None of the patients had received preoperative treatments, including chemotherapy or radiotherapy. Tumour tissues were analyzed via histopathological analysis by two experienced pathologists independently in a blinded manner. The International Federation of Gynecology and Obstetrics (FIGO) staging system were used to classify the ovarian serous cystadenocarcinoma samples (15). Clinical information of the patients is presented in Table I. The patients were followed-up for a median duration of 45.9 months (range, 6–60 months). The present study was approved by the Life Sciences Ethics Committee of Zhengzhou University and informed consent was obtained from each patient.

Table I.

SMURF1 expression and clinicopathological characteristics in OC.

Table I.

SMURF1 expression and clinicopathological characteristics in OC.

SMURF1 expression

Total casesHighLowP-value
Age (years)804238
  ≤603921180.431
  >60412120
TNM stage
  I–II268180.0043a
  III–IV543420
Differentiation
  Well177100.373
  Moderate19  118
  Poor44  2420
Lymph node metastasis
  Yes4630160.0032a
  No341222
Residual tumor size size (cm)
  ≤14222200.464
  >1382018

a P<0.01 indicated a significant difference. OC, ovarian cancer; TNM, tumor-node-metastasis.

Cell lines and cell culture

SKOV3 and OVCAR3 cell lines were purchased from the Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences (Shanghai, China). Cells were cultured at 37°C in an atmosphere of 5% CO2 in Dulbecco's modified Eagle's medium (DMEM)/F-12 (Biological Industries, Cromwell, CT, USA) supplemented with 10% fetal bovine serum (FBS; Biological Industries).

Lentivirus production and transfection

The SMURF1-expressing lentivirus vector, LV5-SMURF1, was constructed by the insertion of a full-length SMURF1 cDNA into the LV5 vector (Shanghai GenePharma Co., Ltd., Shanghai, China) via the NotI and BamHI cloning sites. The LV5-SMURF1 and LV5 control (LV5-NC) vectors were, respectively, co-transfected with shuttle plasmid A1104 and packaging vectors pGag/Pol, pRev and pVSV-G (Shanghai GenePharma Co., Ltd.) into 293T cells (Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China) using the Lipofectamine® 2000 transfection reagent (Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA). Following culturing for 72 h, the supernatants of the transfected cells were harvested. The lentiviral titers were determined by fluorescence microscopy analysis for green fluorescence protein (GFP) expressed via the viral vectors. A total of 1×105 OVCAR3 cells/well were transduced with LV5-SMURF1 and LV5-NC vectors, respectively, at a multiplicity of infection of 30. Following culturing for 48 h, western blot analysis was performed to detect SMURF1 expression in transduced OVCAR3 cells.

The lentiviral vector pGLV3/H1/GFP+Puro (Shanghai GenePharma Co., Ltd.) was used to construct the human SMURF1 short hairpin RNA (shRNA) plasmids; 3 different SMURF1 targeting and silencing constructs were ligated into the LV3 vector via the BamHI and EcoRI cloning sites. The purified LV3-shRNA plasmids and packaging plasmids pGag/Pol, pRev and pVSV-G were co-transfected into 293T cells using 300 µl RNAi-Mate (Shanghai GenePharma Co., Ltd.). Following incubation at 37°C for 72 h, the supernatant was collected and concentrated. For lentiviral infection, SKOV3 cells (1×105 cells/well) were cultured in 6-well plates. Concentrated viral particles were used to transduce cells in an adherent monolayer culture at ~50% confluence in 1 ml complete medium supplemented with 5 µg/ml Polybrene®. Following overnight incubation at 37°C, the viral particles were removed and the medium was changed to one for normal growth conditions. Cells expressing non-targeting control shRNA (NC) or SMURF1 targeting shRNA (shSMURF1) were monitored for GFP expression. To determine the transfection efficiency of LV3-shSMURF1, western blot analysis was performed to detect SMURF1 expression levels in transduced OVCAR3 cells.

RNA isolation and reverse transcription-quantitative polymerase chain (RT-qPCR)

Total RNA was extracted using TRIzol reagent (Thermo Fisher Scientific, Inc.) according to the manufacturer's protocols and reverse transcribed for quantification using a PrimeScript RT reagent kit with gDNA Eraser (Takara Biotechnology Co., Ltd., Dalian, China). qPCR reactions were performed using SYBR Premix Ex Taq II (cat. no. DRRO81A; Takara Bio, Inc., Otsu Japan) on an ABI 7500 Fast Real-Time System (Applied Biosystems; Thermo Fisher Scientific, Inc.) with a 50-µl reaction consisting of 5 ng of cDNA, 10 µM of each of the forward and reverse primers, and 25 µl of 2X SYBR Premix. The qPCR conditions were 95°C for 10 sec, followed by 40 cycles at 95°C for 5 sec and 60°C for 34 sec, and a final stage of dissociation analysis. The primers employed in the present study were as follows: SMURF1 forward, 5′-ACCAGTGCCAACTCAAGGAG-3′ and reverse, 5′-CGACAGTTCGTGTCTGAGGA-3′. The expression levels were normalized to the endogenous control, GAPDH forward, 5′-GGGAAACTGTGGCGTGATGG-3′ and reverse, 5′-GTGTGGAAGTGGGAGACTCAAC-3′ and the relative expression was calculated using the comparative 2−∆∆Cq method (16).

Immunohistochemistry (IHC)

Formalin-fixed paraffin embedded OC sections were incubated with an anti-SMURF1 rabbit polyclonal antibody (dilution 1:100; cat. no. ab38866; Abcam, Cambridge, UK) and anti-cluster of differentiation (CD) 34 rabbit polyclonal antibody (dilution 1:100; cat. no. ab185732; Abcam). Immunohistochemical staining was performed according to the manufacturer's protocol of the SPlink Detection kit (Biotin-Streptavidin-HRP Detection system; cat. no. SP-9001; ZSGB-BIO, Inc., Beijing, China). The deparaffinized sections were placed in a pressure cooker at 100°C for 20 min for antigen retrieval. A 3,3′-diaminobenzidine kit and hematoxylin were used for staining. The methods of IHC scoring were performed as described in the study by Wang et al (17).

Protein extraction and western blot analysis

Cells were harvested 48 h following transduction and total proteins were extracted using radioimmunoprecipitation assay buffer (cat. no. R0010; Beijing Solarbio Science and Technology Co., Ltd., Beijing China) according to the manufacturer's protocols. Antibodies for SMURF1 (1:1,000; cat. no. 2174), Ras-related C3 botulinum toxin substrate 1 (Rac1)/cell division control protein 42 (cdc42) and phosphorylated Rac1/cdc42 (1:1,000; cat. nos. 4651 and 2461, respectively) and β-actin (1:1,000; cat. no. 8475) were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). Antibodies for Ras homolog family member A (RhoA; 1:500; cat. no. ab86297), phosphorylated RhoA (1:1,000; cat. no. ab41435), Rho-associated protein kinase (ROCK1; 1:1,000; cat. no. ab97592) and phosphorylated ROCK1 (1:500; cat. no. ab203273) were obtained from Abcam and incubated overnight (at 4°C) at a 1:500 dilution. Proteins were separated via 12% SDS-PAGE and then transferred onto polyvinylidene difluoride (PVDF) membranes, which were blocked with 5% non-fat dry milk. The blocked membranes were incubated with primary antibodies in Tris-buffered saline with Tween-20. The protein-antibody complexes were detected on the membranes using an enhanced chemiluminescence detection system (Amersham™ ECL™ Prime Western Blotting Reagents; GE Healthcare, Chicago, IL, USA).

Transwell invasion and migration assays

Cell migration and invasion assays using ovarian cell lines were conducted as previously described (6,17). A total of 1×105 or 1×107 cells/ml that were serum-starved overnight were seeded in the upper chamber in 200 µl DMEM/F-12 medium without FBS; 500 µl DMEM/F-12 medium supplemented with 10% FBS was added to the lower chamber (pore size, 8-µm; 24-well plates; cat. no. 3422; Corning Inc., Corning, NY, USA). Cells were incubated at 37°C for 48 h (migration assay) or 72 h (invasion assay). For the invasion assay, the membrane inserts were coated with Matrigel (BD Biosciences, San Jose, CA, USA) on the upper side for 30 min. Cells on the upper membrane surface were removed with a cotton swab. Cells on the lower membrane surface were fixed with 4% paraformaldehyde for 20 min and stained with 0.1% crystal violet for 30 min. Cells were counted in five randomly selected fields under an inverted light microscope at an ×200 magnification (Olympus Corp., Tokyo, Japan). Each assay was performed 3 times.

Statistical analysis

Experimental data were presented as the mean ± standard error of the mean. The Student's two tailed unpaired t-test or one-way analysis of variance (ANOVA) followed by the Tukey's multiple comparison was used to determine statistical significance of in vitro experiments and analyze the association of clinical pathology features and SMURF1 expression. Statistical data analyses were conducted using GraphPad Prism software (version 5; GraphPad Software, Inc., La Jolla, CA, USA). Survival statistical analyses were performed using SPSS 17.0 software (SPSS, Inc., Chicago, IL, USA). Kaplan-Meier curves were constructed to determine patient overall survival (OS) and relapse-free survival (RFS) rates. Patients who were not located for follow-up or who died from causes unrelated to OC were treated as censored events. The statistical differences in survival among subgroups were compared using the log-rank test. P<0.05 was considered to indicate a statistically significant difference.

Results

Expression of SMURF1 in OC cell lines with different invasive abilities

Previous studies have demonstrated that the OC cell lines, SKOV3 and OVCAR3, possess varying invasive abilities. OVCAR3 cells demonstrated an elliptical, less invasive morphology, while SKOV3 cells exhibited an elongated and more invasive morphological phenotype, and have been reported to be significantly more metastatic than OVCAR3 cells in xenograft mouse models (18,19). Consistent with these studies, the results of the present study revealed that OVCAR3 cells exhibited reduced cell migration compared with SKOV3 cells (Fig. 1A and B). To investigate the potential role of SMURF1 in the regulation of OC metastasis, the mRNA and protein expression levels of SMURF1 in SKOV3 and OVCAR3 cell lines were determined. The results revealed that the mRNA and protein levels of SMURF1 were increased in SKOV3 cells compared with in OVCAR3 cells (Fig. 1C and D). To assess the function of SMURF1 in OC cells, we determined whether expression within these cells could be manipulated. The LV3-shSMURF1 and LV5-SMURF1 lentiviral vectors were transduced into SKOV3 and OVCAR3 cells, respectively. To select the cell line with the most effective and stable transfection, the expression levels of SMURF1 protein in stably transfected SKOV3 and OVCAR3 cells cultured with puromycin were analyzed by western blotting. The results indicated that the expression levels of SMURF1 were significantly decreased in all three SMURF1 shRNA-transduced SKOV3 cell lines (Fig. 1E). Furthermore, compared with the non-transduced control cells, ectopic expression of SMURF1 in OVCAR3 cells was significantly increased at the protein level (Fig. 1F).

Effects of SMURF1 in OC cell migration and invasion

To determine the role of SMURF1 in OC metastasis, the effects of SMURF1 on the migration and invasion of OC cells were analyzed in vitro. As presented in Fig. 2, inhibition of SMURF1 in SKOV3 cells significantly reduced cell migration as detected by the number of cells that had migrated via the Transwell membrane; reduced invasion was also reported as observed by the number of cells invading via the Matrigel-coated membrane (Fig. 2A). Conversely, the stable ectopic expression of SMURF1 in OVCAR3 cells significantly enhanced cell migration and invasive abilities (Fig. 2B).

Enhancement of RhoA/ROCK signaling in OC cells by SMURF1

The present study further investigated the molecular mechanism by which SMURF1 promotes OC metastasis. It has been demonstrated that SMURF1 promoted the degradation of the small GTP protein, RhoA, by increasing its ubiquitination (5). RhoA is one of the most extensively investigated members of the Rho GTPase family of proteins; the RhoA/ROCK signaling pathway has been associated with malignant transformation, as well as tumor invasion and metastasis (20,21). To determine whether SMURF1 affects OC cell migration and invasion via this particular signaling pathway, the effects of SMURF1 on the activation of several key downstream signaling molecules were investigated via western blotting. As presented in Fig. 2, compared with SKOV3 control cells, inhibition of SMURF1 within SKOV3 cells significantly decreased the phosphorylation of RhoA, ROCK1, Rac1 and cdc42 (Fig. 2C). Conversely, overexpression of SMURF1 in OVCAR3 cells was observed to enhance RhoA/ROCK signaling in these OC cells (Fig. 2D). The findings of the present study indicated that SMURF1 may promote metastasis by activating the RhoA/ROCK-associated signaling pathways in OC cells.

Overexpression of SMURF1 in OC tissues is associated with poor prognosis

Previously, we observed SMURF1 overexpression in human OC tissues (6). To investigate the biological significance of this finding, the association between the SMURF1 expression levels and the clinical features of patients with OC was investigated in the present study. Real-time qPCR results demonstrated that the expression levels of SMURF1 in OC tissue samples were greater than the levels in the normal ovarian tissue samples (median, 3.195±0.102 vs. 1.971±0.115; P<0.0001). Since the median SMURF1 mRNA expression level in OC tissues was 3.195±0.102, we chose a cut-off value of 3.0 to divide the patients into those with high and low expression levels of SMURF1. According to this cut-off value, 42 patients had a SMURF1 level ≥3.0 and 38 patients had a SMURF1 level <3.0. The associations between the SMURF1 mRNA expression levels and the clinicopathological features are shown in Table I. Statistical analyses revealed that SMURF1 expression in OC specimens was positively associated with advanced clinical FIGO stages (III and IV stage) (P=0.0043; Table I) and tumor lymph node metastasis (P=0.0032; Table I). Of note, higher SMURF1 expression levels were associated with increased microvessel density and the presence of metastasis (Fig. 3), indicating that SMURF1 overexpression may contribute to the progression of OC by promoting tumor metastasis.

In the present study, Kaplan-Meier survival analyses were performed to further investigate whether SMURF1 expression is associated with the 5-year overall survival (OS) and relapse-free survival (RFS) in patients with OC. The results revealed that patients with lower SMURF1 expression levels exhibited longer durations of survival, whereas the patients with higher SMURF1 expression levels had shorter duration of survival (P=0.041; Fig. 4A). Additionally, the present study reported an association between higher SMURF1 expression levels with shorter RFS; however, statistical significance was not observed (P=0.057; Fig. 4B).

Discussion

SMURF1, a member of the HECT family of E3 ubiquitin ligases, is involved in the regulation of numerous pathological processes (22,23). The findings of the present study indicated that SMURF1 may promote ovarian cancer (OC) cell migration and invasion, and that overexpression of SMURF1 in OC tissues may facilitate the progression of OC.

SMURF1 has also been suggested as a promoter of metastasis in numerous human malignancies. In breast cancer, overexpression of the E3 ubiquitin ligase SMURF1 led to RhoA ubiquitination and degradation (7). Degradation of the small GTPase RhoA in tumor cells disrupted F-actin cytoskeletal organization, reduced cell adhesion, increased cell migration and invasion and promoted breast cancer progression and metastasis (7). In prostate cancer, SMURF1 was associated with androgen-induced cell migration and invasion. Downregulating SMURF1 completely inhibited the invasive ability of C4-2 cells (24). In the cervical cancer cell line, HeLa and the breast cancer cell line MCF-7, SMURF1 was reported to function as an upstream oncogenic factor, which negatively regulated the antimetastatic factor, DAB2IP for the control of aberrant tumor cell growth and migration (12). In gastric cancer, SMURF1 promoted cancer cell migration and invasion by suppressing the expression of double C2 protein/DAB2IP (25). Our previous study reported SMURF1 as a direct target of microRNA (miR)-497, in which miR-497 inhibited OC cell migration and invasion (6). These findings indicated a potential role of SMURF1 in cancer metastasis. In the present study, it was observed that the expression of SMURF1 was significantly increased in OC cells, which exhibited high invasive potentials. The inhibition of SMURF1 expression suppressed the invasion and migration of OC cells. In human OC specimens, SMURF1 was significantly overexpressed in the present study. Furthermore, high expression levels of SMURF1 were associated with aggressive tumor characteristics and poor OS of patients with OC. These findings support the role of SMURF1 in promoting the progression of OC; however, the regulatory mechanism of constitutive activation of SMURF1 in OC remains unknown.

RhoA, a member of the Rho homolog family of small GTPases, promotes the reorganization of the actin cytoskeleton and regulates cell shape, attachment and motility (26). Increased activation of RhoA has been associated with tumor cell proliferation and metastasis (27,28). In OC, the activation of RhoA GTPase and downstream ROCK led to enhanced cell invasion and migration (29,30). The suppressive effects of ROCK inhibitors, as well as macitentan, have demonstrated that RhoA may be involved in the cell migration of epithelial OC (31). Via interactions with a variety of factors, RhoA can modulate the activities of various signaling pathways, including that of phosphoinositide 3-kinase (PI3K)/protein kinase B, Ras/Raf and mitogen-activated protein kinase/extracellular signal-regulated kinase; loss of RhoA can further dysregulate proliferation and metastasis-associated pathways, inducing tumor development (12,32,33). It was previously demonstrated that SMURF1-mediated ubiquitination of RhoA was responsible for the precise temporal and spatial regulation of RhoA, which is required for optimal cell migration (34). Thus, the present study investigated whether SMURF1 was involved in the regulation of OC cell migration and invasion via activation of the RhoA signaling pathway. Consistent with these previous studies, the present study reported that reductions in SMURF1 expression decreased OC cell migration and invasion, and attenuated RhoA-mediated activation of the RhoA/ROCK signaling pathway. Recent studies have demonstrated that SMURF1 regulated a variety of signaling networks, including transforming growth factor β family signaling and the Wnt signaling pathway (34); however, we only investigated the role of SMURF1 in the promotion of OC metastasis via the RhoA/ROCK oncogenic pathway in the present study. We anticipate that future studies will reveal that SMURF1 may regulate other signalling pathways within OC cells. The identification of these pathways may provide novel insight into the function of SMURF1 in the progression and metastasis of OC.

At present, a few oncogenes have been reported to possess pro-metastatic functions in OC. Yu et al (35) identified a functional kinase, spleen tyrosine kinase (SYK), which was associated with OC cell motility and invasiveness. In addition, the inhibition of SYK significantly decreased the invasive ability of OC cells. Dorayappan et al (36) reported that patient-derived exosomes from ascites-associated OC cells cultured under hypoxic conditions, exhibited an increased abundance of potent oncogenic proteins, including signal transducer and activator of transcription 3 and Fas, which are capable of significantly increasing cell migration/invasion. Vascular endothelial growth factor (VEGF)-C and VEGF-D were proposed to contribute to tumor-associated lymphatic vessel growth, enhancing the metastatic spread of tumor cells to lymph nodes. Secreted protein acidic and rich in cysteine functions as a tumor suppressor by inhibiting lymph node metastasis via the regulation of VEGF-C/D expression in OC cells (37). Formyl peptide receptor 2 (FPR2) expression levels were upregulated in OC cells. The inhibition of FPR2 was observed to reduce the migration and invasion of OC cells (38). Park et al (39) reported that Toll-like receptor 5/7-mediated PI3K activation induced epithelial-mesenchymal transition and the metastasis of OC cells via the expression of Wiskott-Aldrich syndrome protein family member 3-dependent mesothelin or POU domain class 5 transcription factor 1/SRY-box 2. The results of the present study revealed that SMURF1 promoted OC cell metastasis via the activation of the RhoA/ROCK signaling pathway. This finding indicated that SMURF1 may be a promising molecular target for the optimization of individual therapeutic approaches for patients with OC.

Acknowledgements

Not applicable.

Funding

The present study was supported by a grant from the Natural Science Foundation of Henan Province of China (grant no. 182300410358).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

WW, HD, HL and FH conducted the data acquisition and the patient follow-up. WW and GL contributed to the conception and design, the data analysis and the interpretation of the results. WW and GL wrote, reviewed and edited the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Ethics approval and consent to participate

The present study was approved by the Life Sciences Ethics Committee of Zhengzhou University (Zhengzhou, China) and informed consent was obtained from all patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chen W, Zheng R, Baade PD, Zhang S, Zeng H, Bray F, Jemal A, Yu XQ and He J: Cancer statistics in China, 2015. CA Cancer J Clin. 66:115–132. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Ng JS, Low JJ and Ilancheran A: Epithelial ovarian cancer. Best practice & research. Clin Obstet Gynaecol. 26:337–345. 2012.

3 

Jayson GC, Kohn EC, Kitchener HC and Ledermann JA: Ovarian cancer. Lancet. 384:1376–1388. 2014. View Article : Google Scholar : PubMed/NCBI

4 

Wen M, Ma X, Cheng H, Jiang W, Xu X, Zhang Y, Zhang Y, Guo Z, Yu Y, Xu H, et al: Stk38 protein kinase preferentially inhibits TLR9-activated inflammatory responses by promoting MEKK2 ubiquitination in macrophages. Nat Commun. 6:71672015. View Article : Google Scholar : PubMed/NCBI

5 

Lee MG, Jeong SI, Ko KP, Park SK, Ryu BK, Kim IY, Kim JK and Chi SG: RASSF1A directly antagonizes RhoA activity through the assembly of a smurf1-mediated destruction complex to suppress tumorigenesis. Cancer Res. 76:1847–1859. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Wang W, Ren F, Wu Q, Jiang D, Li H, Peng Z, Wang J and Shi H: MicroRNA-497 inhibition of ovarian cancer cell migration and invasion through targeting of SMAD specific E3 ubiquitin protein ligase 1. Biochem Biophys Res Commun. 449:432–437. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Yu L, Liu X, Cui K, Di Y, Xin L, Sun X, Zhang W, Yang X, Wei M, Yao Z, et al: SND1 acts downstream of TGFβ1 and upstream of Smurf1 to promote breast cancer metastasis. Cancer Res. 75:1275–1286. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Khammanivong A, Gopalakrishnan R and Dickerson EB: SMURF1 silencing diminishes a CD44-high cancer stem cell-like population in head and neck squamous cell carcinoma. Mol Cancer. 13:2602014. View Article : Google Scholar : PubMed/NCBI

9 

Huang C, Rajfur Z, Yousefi N, Chen Z, Jacobson K and Ginsberg MH: Talin phosphorylation by Cdk5 regulates Smurf1-mediated talin head ubiquitylation and cell migration. Nat Cell Biol. 11:624–630. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Liu C, Billadeau DD, Abdelhakim H, Leof E, Kaibuchi K, Bernabeu C, Bloom GS, Yang L, Boardman L, Shah VH, et al: IQGAP1 suppresses TβRII-mediated myofibroblastic activation and metastatic growth in liver. J Clin Invest. 123:1138–1156. 2013. View Article : Google Scholar : PubMed/NCBI

11 

Kwon A, Lee HL, Woo KM, Ryoo HM and Baek JH: SMURF1 plays a role in EGF-induced breast cancer cell migration and invasion. Mol Cells. 36:548–555. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Li X, Dai X, Wan L, Inuzuka H, Sun L and North BJ: Smurf1 regulation of DAB2IP controls cell proliferation and migration. Oncotarget. 7:26057–26069. 2016.PubMed/NCBI

13 

Wang Z, Wang J, Li X, Xing L, Ding Y, Shi P, Zhang Y, Guo S, Shu X and Shan B: Bortezomib prevents oncogenesis and bone metastasis of prostate cancer by inhibiting WWP1, Smurf1 and Smurf2. Int J Oncol. 45:1469–1478. 2014. View Article : Google Scholar : PubMed/NCBI

14 

Kwei KA, Shain AH, Bair R, Montgomery K, Karikari CA, van de Rijn M, Hidalgo M, Maitra A, Bashyam MD and Pollack JR: SMURF1 amplification promotes invasiveness in pancreatic cancer. PLoS One. 6:e239242011. View Article : Google Scholar : PubMed/NCBI

15 

Javadi S, Ganeshan DM, Qayyum A, Iyer RB and Bhosale P: Ovarian cancer, the revised FIGO staging system, and the role of imaging. AJR Am J Roentgenol. 206:1351–1360. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2-ΔΔCT method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

17 

Wang J, Liang WJ, Min GT, Wang HP, Chen W and Yao N: LTBP2 promotes the migration and invasion of gastric cancer cells and predicts poor outcome of patients with gastric cancer. Int J Oncol. 52:1886–1898. 2018.PubMed/NCBI

18 

Chen J, Wang L, Matyunina LV, Hill CG and McDonald JF: Overexpression of miR-429 induces mesenchymal-to-epithelial transition (MET) in metastatic ovarian cancer cells. Gynecol Oncol. 121:200–205. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Shaw TJ, Senterman MK, Dawson K, Crane CA and Vanderhyden BC: Characterization of intraperitoneal, orthotopic, and metastatic xenograft models of human ovarian cancer. Mol Ther. 10:1032–1042. 2004. View Article : Google Scholar : PubMed/NCBI

20 

Kataoka K and Ogawa S: Variegated RHOA mutations in human cancers. Exp Hematol. 44:1123–1129. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Liu Z, Chu S, Yao S, Li Y, Fan S, Sun X, Su L and Liu X: CD74 interacts with CD44 and enhances tumorigenesis and metastasis via RHOA-mediated cofilin phosphorylation in human breast cancer cells. Oncotarget. 18:68303–68313. 2016.

22 

David D, Nair SA and Pillai MR: Smurf E3 ubiquitin ligases at the cross roads of oncogenesis and tumor suppression. Biochim Biophys Acta. 1835:119–128. 2013.PubMed/NCBI

23 

Cao Y and Zhang L: A Smurf1 tale: function and regulation of an ubiquitin ligase in multiple cellular networks. Cell Mol Life Sci. 70:2305–2317. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Gang X, Wang G and Huang H: Androgens regulate SMAD ubiquitination regulatory factor-1 expression and prostate cancer cell invasion. Prostate. 75:561–572. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Tao Y, Sun C, Zhang T and Song Y: SMURF1 promotes the proliferation, migration and invasion of gastric cancer cells. Oncol Rep. 38:1806–1814. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Blangy A: Tensins are versatile regulators of Rho GTPase signaling and cell adhesion. Biol Cell. 109:115–126. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Chang HR, Nam S, Lee J, Kim JH, Jung HR, Park HS, Park S, Ahn YZ, Huh I, Balch C, et al: Systematic approach identifies RHOA as a potential biomarker therapeutic target for Asian gastric cancer. Oncotarget. 7:81435–81451. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Li Y, Xu T, Zou H, Chen X, Sun D and Yang M: Cell migration microfluidics for electrotaxis-based heterogeneity study of lung cancer cells. Biosens Bioelectron. 89:837–845. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Semprucci E, Tocci P, Cianfrocca R, Sestito R, Caprara V, Veglione M, Castro VD, Spadaro F, Ferrandina G, Bagnato A, et al: Endothelin A receptor drives invadopodia function and cell motility through the beta-arrestin/PDZ-RhoGEF pathway in ovarian carcinoma. Oncogene. 35:3432–3442. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Paul NR, Allen JL, Chapman A, Morlan-Mairal M, Zindy E, Jacquemet G, Fernandez del Ama L, Ferizovic N, Green DM, Howe JD, et al: α5β1 integrin recycling promotes Arp2/3-independent cancer cell invasion via the formin FHOD3. J Cell Biol. 210:1013–1031. 2015. View Article : Google Scholar : PubMed/NCBI

31 

Tocci P, Caprara V, Cianfrocca R, Sestito R, Di Castro V, Bagnato A and Rosanò L: Endothelin-1/endothelin A receptor axis activates RhoA GTPase in epithelial ovarian cancer. Life Sci. 159:49–54. 2016. View Article : Google Scholar : PubMed/NCBI

32 

Zandvakili I, Lin Y, Morris JC and Zheng Y: Rho GTPases: Anti- or pro-neoplastic targets? Oncogene. 36:3213–3222. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Jansen S, Gosens R, Wieland T and Schmidt M: Paving the Rho in cancer metastasis: Rho GTPases and beyond. Pharmacol Ther. 183:1–21. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Huang C: Roles of E3 ubiquitin ligases in cell adhesion and migration. Cell Adh Migr. 4:10–18. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Yu Y, Suryo Rahmanto Y, Lee MH, Wu PH, Phillip JM, Huang CH, Vitolo MI, Gaillard S, Martin SS, Wirtz D, et al: Inhibition of ovarian tumor cell invasiveness by targeting SYK in the tyrosine kinase signaling pathway. Oncogene. 37:3778–3789. 2018. View Article : Google Scholar : PubMed/NCBI

36 

Dorayappan KD, Wanner R, Wallbillich JJ, Saini U, Zingarelli R, Suarez AA, Cohn DE and Selvendiran K: Hypoxia-induced exosomes contribute to a more aggressive and chemoresistant ovarian cancer phenotype: A novel mechanism linking STAT3/Rab proteins. Oncogene. 37:3806–3821. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Peng F, Zhong Y, Liu Y, Zhang Y, Xie Y, Lu Y, Zhang X and Li D: SPARC suppresses lymph node metastasis by regulating the expression of VEGFs in ovarian carcinoma. Int J Oncol. 51:1920–1928. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Xie X, Yang M, Ding Y, Yu L and Chen J: Formyl peptide receptor 2 expression predicts poor prognosis and promotes invasion and metastasis in epithelial ovarian cancer. Oncol Rep. 38:3297–3308. 2017.PubMed/NCBI

39 

Park GB and Kim D: TLR5/7-mediated PI3K activation triggers epithelial-mesenchymal transition of ovarian cancer cells through WAVE3-dependent mesothelin or OCT4/SOX2 expression. Oncol Rep. 38:3167–3176. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2019
Volume 41 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Wang W, Du H, Liu H, Hu F and Liu G: SMAD specific E3 ubiquitin protein ligase 1 promotes ovarian cancer cell migration and invasion via the activation of the RhoA/ROCK signaling pathway. Oncol Rep 41: 668-676, 2019
APA
Wang, W., Du, H., Liu, H., Hu, F., & Liu, G. (2019). SMAD specific E3 ubiquitin protein ligase 1 promotes ovarian cancer cell migration and invasion via the activation of the RhoA/ROCK signaling pathway. Oncology Reports, 41, 668-676. https://doi.org/10.3892/or.2018.6836
MLA
Wang, W., Du, H., Liu, H., Hu, F., Liu, G."SMAD specific E3 ubiquitin protein ligase 1 promotes ovarian cancer cell migration and invasion via the activation of the RhoA/ROCK signaling pathway". Oncology Reports 41.1 (2019): 668-676.
Chicago
Wang, W., Du, H., Liu, H., Hu, F., Liu, G."SMAD specific E3 ubiquitin protein ligase 1 promotes ovarian cancer cell migration and invasion via the activation of the RhoA/ROCK signaling pathway". Oncology Reports 41, no. 1 (2019): 668-676. https://doi.org/10.3892/or.2018.6836