Open Access

SPTBN2 regulates endometroid ovarian cancer cell proliferation, invasion and migration via ITGB4‑mediated focal adhesion and ECM receptor signalling pathway

  • Authors:
    • La Yang
    • Yuanyuan Gu
  • View Affiliations

  • Published online on: April 24, 2023     https://doi.org/10.3892/etm.2023.11977
  • Article Number: 277
  • Copyright: © Yang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Ovarian cancer is as a major contributor to gynaecologic death globally. The present study aimed to investigate the regulatory role of spectrin β non‑erythrocytic 2 gene (SPTBN2) in endometroid ovarian cancer and its mechanism of action. According to the Gene Expression Profiling Interactive Analysis (GEPIA) database, SPTBN2 expression is elevated in ovarian cancer tissues and higher SPTBN2 expression indicated a worse prognosis. The present study assessed SPTBN2 mRNA and protein expression levels by reverse transcription‑quantitative PCR and western blotting, respectively. Cell viability, proliferation, migration and invasion were assessed with Cell Counting Kit‑8, 5‑ethynyl‑2'‑deoxyuridine incorporation, wound healing and Transwell assays, respectively. SPTBN2 expression was notably enhanced in ovarian cancer cell lines, especially in A2780 cells compared with HOSEPiC cells (P<0.001). Following transfection with small interfering (si)RNA targeting SPTBN2, the viability, proliferation, migration and invasion of A2780 cells were decreased compared with those of A2780 cells transfected with siRNA‑NC (P<0.001). Gene Set Enrichment Analysis database revealed that SPTBN2 was primarily enriched in ‘focal adhesion’ and ‘extracellular matrix (ECM)‑receptor interaction’, whereas SPTBN2 was significantly associated with integrin β4 (ITGB4) in the GEPIA database. In addition, rescue experiments were performed to determine the mechanism of SPTBN2 in endometroid ovarian cancer. ITGB4 overexpression reversed the inhibitory effects of the SPTBN2 knockdown on viability, proliferation, migration and invasion of A2780 cells (P<0.05). The impacts of SPTBN2 on the expression of focal adhesion and downstream ECM receptor signalling‑related proteins, including Src and p‑FAK/FAK, were significantly reversed by ITGB4 overexpression (P<0.01). Collectively, SPTBN2 may regulate endometroid ovarian cancer cell proliferation, invasion and migration through the ITGB4‑mediated focal adhesion and ECM receptor signalling pathway.

Introduction

As a key contributor to cancer-related deaths among women, ovarian cancer has the highest mortality rate among gynaecological malignancies (1). It is estimated that there are ~14,000 mortalities due to this condition every year in the USA (2). Risk factors can result in ovarian cancer, such as BRCA mutation, smoking, obesity, nulliparity, early-onset menarche and late-onset menopause (3,4). The 5-year survival rate of patients with ovarian cancer is low and remained at 48% in the USA during 2009 through 2015, as most patients are diagnosed at advanced stages owing to the unspecific clinical manifestations of ovarian cancer (2,5,6). At present, the methods for the treatment of ovarian cancer include surgery, chemotherapy, immunotherapy and targeted therapy (7). Although great progress has been made in treatment methods, the 5-year survival rate of patients with ovarian cancer remains unsatisfactory (8).

Spectrin β non-erythrocytic 2 (SPTBN2) encodes β-III spectrin; mutation in this gene is the cause of spinocerebellar ataxia type 5(9). SPTBN2 is expressed in body cells including kidney, pancreatic and liver cells, especially in Purkinje cells (10). Studies have demonstrated that SPTBN2 is increased in endometrioid, endometrial and colorectal cancer, as well as lung adenocarcinoma; it is associated with several tumorigenesis-related biological processes, including proliferation, migration and invasion (11-13). Additionally, SPTBN2 has been recognized as a marker gene and serves as a crucial factor in the pathogenetic process of several types of cancer (14). Notably, SPTBN2 is shown to be correlated with integrin β4 (ITGB4) in Gene Expression Profiling Interactive Analysis (GEPIA) database.

As a heterodimeric transmembrane receptor, ITGB4 is located at the basal surface of airway epithelial cells in hemidesmosomal structures (15). Previous studies have reported that ITGB4 is aberrantly expressed in numerous types of cancer. For example, ITGB4 is upregulated in patients with colorectal cancer (CRC) and may be a potential serum biomarker for CRC (16). In addition, downregulation of ITGB4 may be a target for treatment of gastric cancer (17). ITGB4 is aberrantly expressed in ovarian cancer cells and ITGB4-mediated focal adhesion kinase (FAK) has been reported to regulate the metastatic potential of ovarian cancer cells by disrupting the basement membrane barrier and promoting cell motility (18).

The present study aimed to investigate the role of SPTBN2 in endometroid ovarian cancer as well as its mechanism of action to lay the foundation for the future exploration of targeted therapy for endometroid ovarian cancer.

Materials and methods

Cell culture

Human ovarian surface epithelial cell line HOSEPiC (cat. no. YS2089C) was purchased by Shanghai YaJi Biological Co., Ltd. Ovarian cancer cell lines SKOV3 (cat. no. BNCC310551) and A2780 (cat. no. BNCC351906) were from BeNa Culture Collection (Beijing Beina Chunglian Institute of Biotechnology). In addition, the ovarian cancer cell line HEY A8 (cat. no. CL-0671) was purchased from Procell Life Science & Technology Co., Ltd. All cells were cultured in Dulbecco's modified Eagle's medium (DMEM; Procell Life Science & Technology Co., Ltd.) supplemented with 10% foetal bovine serum (FBS; Beijing Solarbio Science & Technology Co., Ltd.) and 1% penicillin-streptomycin at 37˚C with 5% CO2.

Cell transfection

Two small interfering (si)RNAs specific to SPTBN2 (siRNA-SPTBN2-1, 5'-ACGTCAATGTACACAACTTCACC-3'; and siRNA-SPTBN2-2, 5'-ACCATTACTTCTCCAAGATGAAG-3'), a pcDNA3.1 expression vector carrying ITGB4 (Ov-ITGB4) as well as their corresponding negative controls (NC) si-NC (5'-AAGACAUUGUGUGUCCGCCTT-3') and Ov-NC (empty vector) were purchased from GeneChem, Inc. Using Lipofectamine® 2000 (Thermo Fisher Scientific, Inc.), 100 nM siRNA-SPTBN2-1/2, siRNA-NC and 4 µg Ov-ITGB4 and Ov-NC were transfected into A2780 cells at 37˚C for 24 h, according to the manufacturer's protocol. After 48 h, cells were washed with PBS and incubated in DMEM and reverse transcription-quantitative PCR (RT-qPCR) and western blotting were performed to test transfection efficacy.

RT-qPCR

Total RNA was extracted from ovarian cancer cells and human ovarian surface epithelial cell line (6x104 cells/well) with TRIzol® (Thermo Fisher Scientific, Inc.) and reverse-transcribed into cDNA using the PrimeScript™ RT Reagent kit (cat. no. RR047A; Takara Bio, Inc.) according to the manufacturer's instructions. qPCR was performed using SYBR Green Master Mix (Applied Biosystems; Thermo Fisher Scientific, Inc.) on an ABI PRISM 7900 Sequence Detection System (Applied Biosystems; Thermo Fisher Scientific, Inc.), according to the manufacturer's instructions. The thermocycling conditions were as follows: Initial denaturation at 95˚C for 8 min; 40 cycles of denaturation at 95˚C for 25 sec; annealing at 60˚C for 30 sec; extension at 72˚C for 30 sec and final extension at 72˚C for 10 min. The mRNA levels were quantified using the 2-ΔΔCq method and normalized to the internal reference gene GAPDH (19). The following primer pairs were used: SPTBN2 forward, 5'-GAGGTCTCGCATTAAGGCTCT-3' and reverse, 5'-CTTTGGCAGTATCTCTCCCGA-3'; ITGB4 forward, 5'-GCAGCTTCCAAATCACAGAGG-3' and reverse, 5'-CCAGATCATCGGACATGGAGTT-3' and GAPDH forward, 5'-TGTGGGCATCAATGGATTTGG-3' and reverse, 5'-ACACCATGTATTCCGGGTCAAT-3'.

Western blotting

Total protein was extracted from ovarian cancer cells and human ovarian surface epithelial cell line in six-well plates at 1x105 cells/well using RIPA lysis buffer (Beyotime Institute of Biotechnology). Total protein was quantified with a bicinchoninic acid protein assay kit (Beyotime Institute of Biotechnology) and separated by SDS-PAGE on an 8% gel (30 µg protein/lane). The separated proteins were transferred onto a PVDF membrane (Beijing Solarbio Science & Technology Co., Ltd.) and blocked with 5% skimmed milk or 5% BSA (MilliporeSigma) for 2 h at room temperature. Subsequently, membranes were incubated at 4˚C overnight with primary antibodies (all from Abcam) against SPTBN2 (cat. no. ab264178; 1:2,000), ITGB4 (cat. no. ab182120; 1:1,000), MMP2 (cat. no. ab92536; 1:1,000), MMP7 (cat. no. ab207299; 1:1,000), MMP9 (cat. no. ab76003; 1:1,000), proto-oncogene tyrosine-protein kinase Src (Src; cat. no. ab133283; 1:1,000), phosphorylated (p)-FAK (cat. no. ab81298; 1:1,000), FAK (cat. no. ab40794; 1:2,000) and GAPDH (cat. no. ab9485; 1:2,500). Membranes were subsequently incubated with HRP-labelled goat anti-rabbit secondary antibody (cat. no. ab6721; 1:2,000; Abcam) for 2 h at room temperature. Protein bands were visualized with Enhanced ECL Chemiluminescent Substrate Kit (Shanghai Yeasen Biotechnology Co., Ltd.) on a Bio-Rad Image Lab system (Shanghai Aiyan Biotechnology Co., Ltd.). Densitometric analysis was performed using Image J software (version 1.46; National Institutes of Health) with GAPDH as the loading control.

Cell Counting Kit-8 (CCK-8) assay

A2780 cells were plated into 96-well plates at a density of 5x103 cells/well and incubated at 37˚C for 24 h. Subsequently, 10 µl CCK-8 reagent (Beyotime Institute of Biotechnology) was added to each well and cells were incubated for another 3 h. Finally, the absorbance at 450 nm was measured with a microplate reader (Thermo Fisher Scientific, Inc.).

5-ethynyl-2'-deoxyuridine (EdU) incorporation cell proliferation assay

A2780 cells were plated into 6-well plates at a density of 4x105 cells/well and incubated overnight at room temperature. Subsequently, each well was filled with 50 µM EdU solution (Beyotime Institute of Biotechnology) and further incubated for 4 h at 37˚C. After removing the working solution, cells were fixed with 4% paraformaldehyde for 15 min at room temperature and permeabilized with 0.5% Triton X-100 for 10 min at room temperature. Following addition of 100 µl Apollo® Reaction Cocktail (Guangzhou RiboBio Co., Ltd.), cells were incubated in the dark for 30 min at room temperature. The cells were observed under a fluorescence microscope (Olympus Corporation).

Wound healing

The migration of A2780 cells was assessed by wound healing assay. Cells were plated into 6-well plates (2x105 cells/well) and incubated at 37˚C until 80-90% confluency was reached. A 10-µl pipette tip was used for creation of a wound in the cell monolayer. The cells were rinsed with PBS then cultured in serum-free DMEM medium at 37˚C with 5% CO2. Images were captured at 0 and 24 h under a light microscope (Olympus Corporation) and tracked with Image-J software (version 1.46; National Institutes of Health).

Transwell assay

The invasive ability of A2780 cells was assessed by Transwell assay. Cells (1x104 cells/well) were plated into the upper compartment of a Transwell plate (8 µm) that was pre-coated with Matrigel (BD Biosciences) at 37˚C for 30 min. The serum-free DMEM medium was placed in the upper chamber. In the lower chamber, the DMEM medium was supplemented with 10% FBS. Following 24 h incubation at 37˚C, the invaded cells were subjected to 4% paraformaldehyde fixation at room temperature for 25 min and 0.1% crystal violet staining at room temperature for 10 min. Finally, the invaded cells was observed under a light microscope (magnification, ⅹ100; Olympus Corporation) and the percentage invaded area was calculated with Image-J software (version 1.46; National Institutes of Health).

Immunofluorescence (IF) assay

Following transfection, A2780 cells were plated into 24-well plates (5x104 cells/well) and cultivated at 37˚C until 100% confluence was reached. Subsequently, A2780 cells were fixed with 4% paraformaldehyde for 20 min at room temperature and 0.5% Triton X-100 (MP Biomedicals, LLC) permeation for 20 min at room temperature. To block non-specific staining, A2780 cells were incubated with 5% BSA (MilliporeSigma) in PBS at room temperature for 2 h. Subsequently, cells were incubated overnight with anti-paxillin antibody (1:50; cat. no. ab32084; Abcam) at 4˚C. Cells were incubated with a FITC-conjugated anti-rabbit IgG secondary antibody (1:5,000; cat. no. 150077; Abcam) and phalloidin (1:10,000; Abcam) for 60 min at room temperature. DAPI (Beyotime Institute of Biotechnology) was used to counterstain the nuclei at room temperature for 5 min. Coverslips were placed on a microscope slide with a drop of anti-fading mounting medium. Finally, cells were observed under a fluorescence microscope (magnification, x400; Olympus Corporation).

Bioinformatics analysis

GEPIA database (gepia.cancer-pku.cn) (20) was used to determine the expression of SPTBN2 (accession no. ENSG00000173898) in 426 ovarian cancer (OV dataset) and 88 normal tissue samples obtained from The Cancer Genome Atlas and Genotype-Tissue Expression projects. GEPIA database also analysed the association of SPTBN2 expression with prognosis of patients with ovarian cancer (Cutoff-High, 50% and Cutoff-Low, 50%). Moreover, GEPIA database was used to explore the relationship between SPTBN2 and ITGB4 (accession no. ENSG00000132470) in the focal adhesion and ECM receptor signalling pathway. Gene Set Enrichment Analysis (GSEA; linkedomics.org/login.php) (21) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway database (kegg.jp/kegg/pathway.html) (22) detected the enrichment of SPTBN2 in ‘focal adhesion’ and ‘ECM-receptor interaction’. The false discovery rate (FDR) was calculated using the Benjamini-Hochberg method (23).

Statistical analysis

All experimental data are presented as the mean ± standard deviation of three replicates and were analysed using GraphPad Prism 8.0 software (GraphPad Software, Inc.; Dotmatics). One-way analysis of variance followed by Tukey's post hoc test was utilized for comparisons between multiple groups. P<0.05 was considered to indicate a statistically significant difference.

Results

SPTBN2 is upregulated in ovarian cancer tissue and is associated with a poor prognosis

The GEPIA database was used to examine SPTBN2 expression in ovarian cancer tissues as well as its association with prognosis of patients with ovarian cancer. Expression of SPTBN2 was increased in ovarian cancer compared with that in normal ovarian tissue (Fig. 1A). In addition, a higher SPTBN2 expression was associated with poorer prognosis (Fig. 1B).

SPTBN2 is upregulated in ovarian cancer cell lines and SPTBN2 silencing inhibits proliferation

RT-qPCR and western blotting results showed that mRNA and protein expression levels, respectively, of SPTBN2 were increased in ovarian cancer cell lines SKOV3, HEY A8 and A2780 relative to HOSEPiC normal ovarian surface epithelial cell line cells (Fig. 2A and B). The highest SPTBN2 expression was observed in A2780 cells, thus, this line was selected for further experiments. To knock down the SPTBN2 expression, two siRNAs specific for SPTBN2 were transfected into the A2780 cells; RT-qPCR and western blotting were used to test the transfection efficacy. Compared with the siRNA-NC group, siRNAs targeting SPTBN2 significantly decreased the expression of SPTBN2 (Fig. 2C and D). In particular, siRNA-SPTBN2-1 exhibited notably higher transfection efficacy compared with the siRNA-SPTBN2-2 group; therefore, siRNA-SPTBN2-1 was selected for subsequent experiments. CCK-8 and EdU incorporation assays were performed to assess the impact of SPTBN2 knockdown on the viability and proliferation of A2780 cells, respectively. The viability and proliferation of A2780 cells were significantly decreased after the transfection with siRNA-SPTBN2 compared with the siRNA-NC group (Fig. 2E and F).

SPTBN2 silencing inhibits the migration and invasion of ovarian cancer cells

The migration and invasion of A2780 cells, which were assessed by wound healing and Transwell assays, respectively, were decreased after SPTBN2 knockdown (Fig. 3A and B). In addition, SPTBN2 knockdown decreased the protein levels of MMP2, MMP7 and MMP9 compared with the siRNA-NC group (Fig. 3C).

SPTBN2 is mainly enriched in focal adhesion and ECM receptor signalling pathway

As displayed in Fig. 4A, GSEA analysed that SPTBN2 was mainly enriched in ‘focal adhesion’ and ‘ECM receptor interaction’ pathways. Additionally, KEGG pathway database revealed that SPTBN2 was enriched in ‘focal adhesion’ and ‘ECM-receptor interaction’ (Fig. 4B and C). In addition, IF demonstrated that SPTBN2 interference decreased the expression of focal adhesion adaptor protein paxillin compared with that in the siRNA-NC group (Fig. 4D).

SPTBN2 knockdown inhibits expression of ITGB4 and related proteins in focal adhesion and ECM receptor signalling pathway

As reported, ITGA and ITGB superfamily members can serve as prognostic markers for serous ovarian cancer (24). In the present study, KEGG pathway database showed the involvement of ITGA and ITGB receptors in ‘focal adhesion’ and ‘ECM-receptor interaction’ (Fig. 5). According to GEPIA, SPTBN2 was significantly correlated with ITGB4 in ovarian cancer tissues (Fig. 6A). RT-qPCR and western blotting were used to evaluate mRNA and protein levels, respectively, of ITGB4 in siRNA-transfected A2780 cells. Compared with that in the siRNA-NC group, SPTBN2 knockdown decreased expression of ITGB4 (Fig. 6B and C). Additionally, western blotting analysis showed that SPTBN2 knockdown decreased the protein expression levels of Src and p-FAK but had no notable effect on FAK compared with the siRNA-NC group (Fig. 6D). Collectively, SPTBN2 knockdown inhibited the expression of ITGB4 and focal adhesion and ECM receptor signalling pathway-associated proteins.

SPTBN2 inhibits proliferation, migration and invasion of ovarian cancer cells through ITGB4

To investigate the mechanism of SPTBN2 in ovarian cancer, rescue experiments were performed. Plasmids carrying ITGB4 were transfected into A2780 cells to overexpress ITGB4. RT-qPCR and western blotting analyses showed that, compared with that in the Ov-NC group, the mRNA and protein expression levels of ITGB4 were significantly increased following ITGB4 overexpression (Fig. 7A and B). Co-transfection experiments demonstrated that the decreased cell viability and proliferation caused by SPTBN2-knockdown A2780 cells were significantly reversed by ITGB4 overexpression (Fig. 7C and D). Similarly, the decreased migration and invasion of A2780 cells caused by SPTBN2 knockdown were rescued after ITGB4 overexpression compared with those in the siRNA-SPTBN2 + Ov-NC group (Fig. 7E and F). Moreover, the decreased levels of MMP2, MMP7 and MMP9 were reversed by ITGB4 overexpression compared with the siRNA-SPTBN2 + Ov-NC group (Fig. 7G). In conclusion, the data suggested that SPTBN2 may inhibit proliferation, migration and invasion of ovarian cancer cells through ITGB4.

SPTBN2 inhibits focal adhesion and the expression of downstream signalling pathway-related proteins in ovarian cancer cells through ITGB4

Compared with the siRNA-SPTBN2 + Ov-NC group, the decreased expression of focal adhesion adaptor protein paxillin caused by SPTBN2-knockdown A2780 cells was increased by ITGB4 overexpression (Fig. 8A). In addition, the SPTBN2-knockdown-induced decrease of Src and p-FAK expression levels were reversed in the siRNA-SPTBN2 + ITGB4 overexpression group (Fig. 8B). Notably, SPTBN2 knockdown combined with ITGB4 overexpression had no effect on FAK protein expression. In summary, SPTBN2 may inhibit focal adhesion and the expression of downstream signalling pathway-associated proteins in ovarian cancer cells through ITGB4.

Discussion

Ovarian cancer is one of the most common lethal epithelial malignancies and has poor outcome owing to late diagnosis (25). SPTBN2 is associated with tumorigenesis-associated biological activity (12). Previous studies have shown that SPTBN2 is involved with numerous types of malignant tumour. For example, SPTBN2 has been proposed as a potential driver gene in breast cancer (26). Huang et al (27) showed that SPTBN2 significantly affects the prognosis of patients with bladder cancer. Additionally, SPTBN2 is elevated in patients with colorectal cancer, and increased SPTBN2 expression is associated with worse prognosis (28). According to the GEPIA database, SPTBN2 is increased in ovarian cancer tissue, and its upregulation is associated with poor prognosis of patients with ovarian cancer (29). The aforementioned results revealed that SPTBN2 may influence the progression of ovarian cancer. Consistently, the present study used GEPIA to assess expression of SPTBN2 in clinical samples and it was also noticed that SPTBN2 was overexpressed in ovarian cancer tissues. In line with findings from the GEPIA database, mRNA and protein expression levels of SPTBN2 were increased in ovarian cancer cell lines (SKOV3, HEY A8 and A2780), particularly in A2780 cells.

A recent study demonstrated that SPTBN2 is highly expressed in lung adenocarcinoma and its downregulation is associated with suppressive effects on proliferative, migratory and invasive capabilities of lung cancer cells (11). In addition, Wang et al (12) suggested that SPTBN2 inhibition is implicated in halting proliferation, migration and invasion of endometrioid endometrial cancer cells. SPTBN2 upregulation promotes the proliferation, migration and invasion of colorectal cancer cells (30). Consistent with these findings, the present study demonstrated that SPTBN2 knockdown suppressed the proliferation, migration and invasion of A2780 cells, suggesting the oncogenic role of SPTBN2 in ovarian cancer. Furthermore, according to GSEA, SPTBN2 was primarily enriched in focal adhesion and ECM receptor signalling pathway, implying that SPTBN2 might contribute to ovarian cancer partially via modulation of focal adhesion and ECM receptor signalling pathway.

As a structural adhesion molecule, ITGB4 forms heterodimers with ITGA6 to achieve its biological functions (31). A previous study has demonstrated that ITGB4 expression is elevated in ovarian cancer tissue (24). In the present study, KEGG pathway analysis showed the involvement of ITGA and ITGB receptors in focal adhesion and ECM receptor signalling pathway. A member of ITGB, ITGB4 was associated with SPTBN2 in the GEPIA database. Additionally, further experiments elucidated that ITGB4 expression was decreased by SPTBN2 knockdown. Moreover, the expression levels of downstream signalling pathway-associated proteins Src and p-FAK were decreased by SPTBN2 knockdown. To assess the regulatory mechanism of SPTBN2 in ovarian cancer, rescue experiments were performed; results showed that the inhibitory effects of SPTBN2 knockdown on proliferation, migration and invasion, as well as the levels of Src and p-FAK, were rescued by ITGB4 overexpression, indicating that SPTBN2 knockdown suppressed the proliferation, migration and invasion, and inactivated focal adhesion and downstream signalling in ovarian cancer cells by reducing ITGB4 expression.

The present study demonstrated that SPTBN2 showed a protective effect against ovarian cancer through ITGB4, revealing that SPTBN2 may be a potential target for treatment of endometroid ovarian cancer. Nevertheless, the regulatory role of SPTBN2 in other ovarian cancer cell lines and animal models of ovarian cancer was not investigated by the present study. Therefore, SPTBN2 expression in ovarian cancer tissue and the association between SPTBN2 expression and prognosis should be confirmed in future studies.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by The National Natural Science Foundation of China (grant no. 81973769).

Availability of data and material

The datasets used and/or analysed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YL designed and conceived the study and wrote the manuscript. YL and GY conducted the experiments and analysed the data. YL and GY confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Torre LA, Islami F, Siegel RL, Ward EM and Jemal A: Global cancer in women: Burden and trends. Cancer Epidemiol Biomarkers Prev. 26:444–457. 2017.PubMed/NCBI View Article : Google Scholar

2 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2020. CA Cancer J Clin. 70:7–30. 2020.PubMed/NCBI View Article : Google Scholar

3 

Rooth C: Ovarian cancer: Risk factors, treatment and management. Br J Nurs. 22:S23–S30. 2013.PubMed/NCBI View Article : Google Scholar

4 

Jelovac D and Armstrong DK: Recent progress in the diagnosis and treatment of ovarian cancer. CA Cancer J Clin. 61:183–203. 2011.PubMed/NCBI View Article : Google Scholar

5 

Moufarrij S, Dandapani M, Arthofer E, Gomez S, Srivastava A, Lopez-Acevedo M, Villagra A and Chiappinelli KB: Epigenetic therapy for ovarian cancer: Promise and progress. Clin Epigenetics. 11(7)2019.PubMed/NCBI View Article : Google Scholar

6 

Siegel RL, Miller KD and Jemal A: Cancer Statistics, 2017. CA Cancer J Clin. 67:7–30. 2017.PubMed/NCBI View Article : Google Scholar

7 

Kim SI and Kim JW: Role of surgery and hyperthermic intraperitoneal chemotherapy in ovarian cancer. ESMO Open. 6(100149)2021.PubMed/NCBI View Article : Google Scholar

8 

Liu HD, Xia BR, Jin MZ and Lou G: Organoid of ovarian cancer: Genomic analysis and drug screening. Clin Transl Oncol. 22:1240–1251. 2020.PubMed/NCBI View Article : Google Scholar

9 

Jackson M, Song W, Liu MY, Jin L, Dykes-Hoberg M, Lin CI, Bowers WJ, Federoff HJ, Sternweis PC and Rothstein JD: Modulation of the neuronal glutamate transporter EAAT4 by two interacting proteins. Nature. 410:89–93. 2001.PubMed/NCBI View Article : Google Scholar

10 

Bian X, Wang S, Jin S, Xu S, Zhang H, Wang D, Shang W and Wang P: Two novel missense variants in SPTBN2 likely associated with spinocerebellar ataxia type 5. Neurol Sci. 42:5195–5203. 2021.PubMed/NCBI View Article : Google Scholar

11 

Wu C, Dong B, Huang L, Liu Y, Ye G, Li S and Qi Y: SPTBN2, a new biomarker of lung adenocarcinoma. Front Oncol. 11(754290)2021.PubMed/NCBI View Article : Google Scholar

12 

Wang P, Liu T, Zhao Z, Wang Z, Liu S and Yang X: SPTBN2 regulated by miR-424-5p promotes endometrial cancer progression via CLDN4/PI3K/AKT axis. Cell Death Discov. 7(382)2021.PubMed/NCBI View Article : Google Scholar

13 

Yue M, Liu T, Yan G, Luo X and Wang L: LINC01605, regulated by the EP300-SMYD2 complex, potentiates the binding between METTL3 and SPTBN2 in colorectal cancer. Cancer Cell Int. 21(504)2021.PubMed/NCBI View Article : Google Scholar

14 

Forman OP, De Risio L, Stewart J, Mellersh CS and Beltran E: Genome-wide mRNA sequencing of a single canine cerebellar cortical degeneration case leads to the identification of a disease associated SPTBN2 mutation. BMC Genet. 13(55)2012.PubMed/NCBI View Article : Google Scholar

15 

Han L, Wang L, Tang S, Yuan L, Wu S, Du X, Xiang Y, Qu X, Liu H, Luo H, et al: ITGB4 deficiency in bronchial epithelial cells directs airway inflammation and bipolar disorder-related behavior. J Neuroinflammation. 15(246)2018.PubMed/NCBI View Article : Google Scholar

16 

Jiang X, Wang J, Wang M, Xuan M, Han S, Li C, Li M, Sun XF, Yu W and Zhao Z: ITGB4 as a novel serum diagnosis biomarker and potential therapeutic target for colorectal cancer. Cancer Med. 10:6823–6834. 2021.PubMed/NCBI View Article : Google Scholar

17 

Hong D, Zhang X, Li R, Yu J, Lou Y, He Q, Li X, Xu D, Lv P, Lin J and Chen Y: Deletion of TMEM268 inhibits growth of gastric cancer cells by downregulating the ITGB4 signaling pathway. Cell Death Differ. 26:1453–1466. 2019.PubMed/NCBI View Article : Google Scholar

18 

Skubitz AP, Bast RC Jr, Wayner EA, Letourneau PC and Wilke MS: Expression of alpha 6 and beta 4 integrins in serous ovarian carcinoma correlates with expression of the basement membrane protein laminin. Am J Pathol. 148:1445–1461. 1996.PubMed/NCBI

19 

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

20 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45:W98–W102. 2017.PubMed/NCBI View Article : Google Scholar

21 

Vasaikar SV, Straub P, Wang J and Zhang B: LinkedOmics: Analyzing multi-omics data within and across 32 cancer types. Nucleic Acids Res. 46:D956–D963. 2018.PubMed/NCBI View Article : Google Scholar

22 

Kanehisa M, Sato Y, Kawashima M, Furumichi M and Tanabe M: KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res. 44:D457–D462. 2016.PubMed/NCBI View Article : Google Scholar

23 

Ferreira JA: The Benjamini-Hochberg method in the case of discrete test statistics. Int J Biostat. 3(Article 11)2007.PubMed/NCBI View Article : Google Scholar

24 

Zhu T, Chen R, Wang J, Yue H, Lu X and Li J: The prognostic value of ITGA and ITGB superfamily members in patients with high grade serous ovarian cancer. Cancer Cell Int. 20(257)2020.PubMed/NCBI View Article : Google Scholar

25 

Gong G, Lin T and Yuan Y: Integrated analysis of gene expression and DNA methylation profiles in ovarian cancer. J Ovarian Res. 13(30)2020.PubMed/NCBI View Article : Google Scholar

26 

Yang Z, Yu G, Guo M, Yu J, Zhang X and Wang J: CDPath: Cooperative driver pathways discovery using integer linear programming and markov clustering. IEEE/ACM Trans Comput Biol Bioinform. 18:1384–1395. 2021.PubMed/NCBI View Article : Google Scholar

27 

Huang M, Long Y, Jin Y, Ya W, Meng D, Qin T, Su L, Zhou W, Wu J, Huang C and Huang Q: Comprehensive analysis of the lncRNA-miRNA-mRNA regulatory network for bladder cancer. Transl Androl Urol. 10:1286–1301. 2021.PubMed/NCBI View Article : Google Scholar

28 

Zhang Z, Wang Q, Zhang M, Zhang W, Zhao L, Yang C, Wang B, Jiang K, Ye Y, Shen Z and Wang S: Comprehensive analysis of the transcriptome-wide m6A methylome in colorectal cancer by MeRIP sequencing. Epigenetics. 16:425–435. 2021.PubMed/NCBI View Article : Google Scholar

29 

Feng P, Ge Z, Guo Z, Lin L and Yu Q: A comprehensive analysis of the downregulation of miRNA-1827 and its prognostic significance by targeting SPTBN2 and BCL2L1 in ovarian cancer. Front Mol Biosci. 8(687576)2021.PubMed/NCBI View Article : Google Scholar

30 

Zhao SY, Wang Z, Wu XB, Zhang S, Chen Q, Wang DD and Tan QF: CERS6-AS1 contributes to the malignant phenotypes of colorectal cancer cells by interacting with miR-15b-5p to regulate SPTBN2. Kaohsiung J Med Sci. 38:403–414. 2022.PubMed/NCBI View Article : Google Scholar

31 

Meng X, Liu P, Wu Y, Liu X, Huang Y, Yu B, Han J, Jin H and Tan X: Integrin beta 4 (ITGB4) and its tyrosine-1510 phosphorylation promote pancreatic tumorigenesis and regulate the MEK1-ERK1/2 signaling pathway. Bosn J Basic Med Sci. 20:106–116. 2020.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

June-2023
Volume 25 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Yang L and Yang L: SPTBN2 regulates endometroid ovarian cancer cell proliferation, invasion and migration via ITGB4‑mediated focal adhesion and ECM receptor signalling pathway. Exp Ther Med 25: 277, 2023
APA
Yang, L., & Yang, L. (2023). SPTBN2 regulates endometroid ovarian cancer cell proliferation, invasion and migration via ITGB4‑mediated focal adhesion and ECM receptor signalling pathway. Experimental and Therapeutic Medicine, 25, 277. https://doi.org/10.3892/etm.2023.11977
MLA
Yang, L., Gu, Y."SPTBN2 regulates endometroid ovarian cancer cell proliferation, invasion and migration via ITGB4‑mediated focal adhesion and ECM receptor signalling pathway". Experimental and Therapeutic Medicine 25.6 (2023): 277.
Chicago
Yang, L., Gu, Y."SPTBN2 regulates endometroid ovarian cancer cell proliferation, invasion and migration via ITGB4‑mediated focal adhesion and ECM receptor signalling pathway". Experimental and Therapeutic Medicine 25, no. 6 (2023): 277. https://doi.org/10.3892/etm.2023.11977