Open Access

FUBP1 mediates the growth and metastasis through TGFβ/Smad signaling in pancreatic adenocarcinoma

  • Authors:
    • Yue Zhang
    • Jinlian Chen
    • Nvshi Zhou
    • Yun Lu
    • Jingwen Lu
    • Xin Xing
    • Hua Chen
    • Xingxing Zhang
  • View Affiliations

  • Published online on: February 26, 2021     https://doi.org/10.3892/ijmm.2021.4899
  • Article Number: 66
  • Copyright: © Zhang 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

Recent studies have reported that the expression levels of far upstream element‑binding protein 1 (FUBP1) were upregulated and served a crucial role in several types of cancer. However, the underlying molecular mechanisms and clinical significance of FUBP1 in pancreatic adenocarcinoma (PAAD) remain unclear. The present study aimed to determine the expression levels of FUBP1 in patients with PAAD and subsequently investigated the biological functions and mechanisms of FUBP1 using in vitro assays. FUBP1 expression levels and survival outcomes in patients with PAAD were analyzed using The Cancer Genome Atlas and starBase databases. Reverse transcription‑quantitative PCR was used to analyze the mRNA expression levels of FUBP1 in PAAD and adjacent normal tissues. In addition, the expression of FUBP1 was knocked down with small interfering RNA and overexpressed using FUBP1‑overexpressed plasmids, and the effects on biological functions, including cell proliferation, migration and invasion, were investigated. Western blotting and immunofluorescence assays were used to determine the role of FUBP1 in epithelial‑mesenchymal transition (EMT). The results of the present study revealed that the expression levels of FUBP1 were upregulated in PAAD tissues compared with adjacent normal tissues and the upregulated expression was significantly associated with poor survival. The knockdown of FUBP1 expression significantly inhibited the proliferative, migratory and invasive abilities of the PAAD PaTu8988 cell line, while the overexpression of FUBP1 promoted cell proliferation, migration and invasion in the PAAD SW1990 cell line. Furthermore, the knockdown of FUBP1 downregulated the expression levels of EMT‑related markers, including N‑cadherin, β‑catenin and vimentin, while the expression levels of E‑cadherin were upregulated. The knockdown of FUBP1 was also revealed to regulate the TGFβ/Smad signaling cascade by downregulating phosphorylated‑Smad2/3 and TGFβ1 expression levels. Conversely, the overexpression of FUBP1 reversed these effects. In conclusion, the findings of the present study indicated that FUBP1 may be a potential oncogene that mediates the EMT of PAAD via TGFβ/Smad signaling. These data suggested that FUBP1 may represent a potential biomarker for the diagnosis of PAAD or a target for the treatment of patients with PAAD.

Introduction

Pancreatic adenocarcinoma (PAAD) is one of the most lethal types of malignancy and has an extremely poor prognosis, with an overall 5-year survival rate of <5% (1,2). The main reason for the poor prognosis is that it is difficult to diagnose PAAD at an early stage, because the cancer-specific symptoms usually only occur at an advanced stage (3). In addition, there are currently no effective treatments available for PAAD. Therefore, novel strategies to diagnose and prevent PAAD are required urgently.

Human far upstream element-binding protein 1 (FUBP1) was discovered to be an important regulator of transcription, mRNA splicing and translation by binding to far upstream element (FUSE), and is located in the reverse strand of chromosome 1p31.1 (4). Upon binding to FUSE, FUBP1 was reported to upregulate the expression levels of the oncogene Myc, which subsequently promoted cell growth and metastasis, and functioned as an oncogene by modulating the FUBP1/FUSE/Myc feedback loop (4-6). Notably, FUBP1 has also been reported to exert oncogenic roles in a variety of tumor types, including liver cancer (7,8), glioma (9), neuroblastoma (10), renal cell carcinoma (11), lung cancer (12), tongue squamous cell carcinoma (13), esophageal squamous cell carcinoma (14), gastric cancer (15,16) and colorectal cancer (17). A previous study demonstrated that FUBP1 served as an oncogene and was associated with a poor prognosis in patients with PAAD (18). In addition, FUBP1 regulated the immune response by upregulating the expression levels of programmed death-ligand 1 in PAAD cells (18). However, the biological functions and molecular mechanisms of FUBP1 in PAAD remain unknown and require further investigations.

The present study analyzed the expression levels of FUBP1 in PAAD and adjacent normal tissues and determined the association between FUBP1 and clinical prognosis of PAAD. Moreover, the role of FUBP1 on the biological functions of PAAD cells, in addition to the potential mechanisms, were investigated in vitro.

Materials and methods

Patient studies

From September 2017 to December 2019, a total of 7 patients including 5 males and 2 females, with a mean age of 49.5 years (range, 40-66 years), diagnosed with PAAD at the Shanghai Fengxian District Central Hospital (Shanghai, China) were enrolled in the present study. All patients provided written informed consent prior to participation. The 7 pairs of PAAD and adjacent normal tissues (at least 3 cm away from the margin of PAAD tissues) were resected and collected following surgery. All the patients had not received radiotherapy or chemotherapy before the operation and were diagnosed with PAAD according to the pathological results. The patients with other malignant tumors were excluded. The study protocol was approved by the Ethics Committee of the Shanghai Fengxian District Central Hospital (approval no. 2017-Ethical Review-KY-05) and was performed in accordance with the principles of the Declaration of Helsinki.

Expression dataset

Gene expression RNAseq data of FUBP1 were downloaded from the UCSC Xena database (http://xena.ucsc.edu/) (19), which included PAAD samples [The Cancer Genome Atlas (TCGA), n=183] and normal samples [Genotype Tissue Expression (GTEx), n=165]. The correlation between gene expression and overall survival was downloaded from the starBase database (http://starbase.sysu.edu.cn/index.php) (20), which contained 176 PAAD patients.

Cell lines and culture

Human PAAD cell lines (BxPC-3, PaTu8988, PANC-1 and SW1990) were conserved at the Central Laboratory of Shanghai Fengxian District Central Hospital, and were cultured in DMEM supplemented with 10% FBS (both from Gibco; Thermo Fisher Scientific, Inc.), and maintained in a humidified incubator with 5% CO2 at 37°C.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from tissues and cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). Total RNA was reverse transcribed into cDNA using a PrimeScript™ RT Reagent kit (Takara Bio, Inc.) with the conditions of 15 min at 37°C and 5 sec at 85°C. qPCR was subsequently performed on an ABI 7300 Real-Time PCR system (Applied Biosystems; Thermo Fisher Scientific, Inc.) using a SYBR® Premix Dimmer Eraser kit (Takara Bio, Inc.). Thermocycling conditions for the qPCR were as follows: Initial denaturation at 95°C for 30 sec; 40 cycles of denaturation at 95°C for 5 sec; annealing at 58°C for 30 sec, and extension at 72°C for 30 sec. The following primers pairs were used for the qPCR: FUBP1 forward, 5′-GGACAACACCCGAAAGGATA-3′ and reverse, 5′-ATGTTCCAGTTGCCTTGACC-3′; and GAPDH forward, 5′-TTGGTATCGTGGAAGGACTCA-3′ and reverse, 5′-TGTCATCATATTTGGCAGGTT-3′ (all purchased from Sangon Biotech Co., Ltd.). The relative mRNA expression levels were quantified using the 2−ΔΔCq method (21). GAPDH was used as the internal control.

Cell transfection

Cells were transfected with small interfering RNA (siRNA/si) targeting FUBP1 (si-FUBP1), sinegative control (NC), FUBP1 overexpression (OE) vector (FUBP1-OE) or control vector using Lipofectamine® 2000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.) and Opti-MEM (Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Briefly, PaTu8988 cells with a confluence of 50-60% in a 6-well plate were transfected with 100 pmol si-FUBP1 or an equal volume of si-NC. For the transfection of plasmids, the cell density of SW1990 was 70-80% before transfection, with 5 µg FUBP1-OE or VECTOR plasmids transfected in each well of a 6-well plate. Subsequently, the mixture of transfection reagents was replaced with 10% FBS-DMEM after 5 h at 37°C. Then, the following assays were performed after post-transfected cells were incubated at 37°C for 48 h. The sequences of the siRNAs (all from GenePharma, Co., Ltd.) were as follows: si-FUBP1 forward, 5′-GGUGCUGACAAACCUCUUATT-3′ and reverse, 5′-UAAGAGGUUUGUCAGCACCTT-3′; si-NC forward, 5′-UUCUCCGAACGUGUCACGUTT-3′ and reverse, 5′-ACGUGACACGUUCGGAGAATT-3′. The OE-FUBP1 plasmid contained full-length amplified sequences of FUBP1 cloned into a pCD513B plasmid (purchased from Biogot Biotechnology Co., Ltd).

Cell Counting Kit-8 (CCK-8) assay

Cell proliferation was analyzed using a CCK-8 assay (Dojindo Molecular Technologies, Inc.), according to the manufacturer's protocol. Briefly, following transfection, cells were seeded into 96-well plates at a density of 5×103 cells/well and cultured for 10, 24, 48 or 72 h. After the cells were washed twice with cold PBS, the mixture of 10 µl CCK-8 and 100 µl serum-free DMEM was added into each well and the plates were incubated at 37°C for 45 min. Subsequently, the absorbance was measured at a wavelength of 450 nm using a Wellscan MK3 microplate reader (Thermo Labsystems, Inc.).

Cell migration and invasion assays

Cell migration and invasion assays were performed in 8.0-µm pore insert Transwell chambers (EMD Millipore) and Transwell invasion Matrigel chambers (Corning, Inc.), respectively. For the invasion assay, the Transwell membrane of the Matrigel chambers was precoated for 1 h at 37°C using serum-free DMEM. Briefly, DMEM supplemented with 10% FBS was plated into the lower chambers and 5×104 cells in serum-free DMEM were plated into the upper chambers. Following incubation at 37°C for 24 h, the non-migratory and non-invasive cells remaining in the upper chambers were gently removed with a cotton bud. The invasive and migratory cells in the lower chambers of the Transwell plates were stained with 0.1% crystal violet for 30 min at room temperature. The migratory and invasive cells were counted under a bright-field fluorescence microscope (Olympus Corporation; magnification, ×200) in five randomly selected fields of view.

Wound healing assay

Upon transfected cells reaching 95% confluence, an artificial wound was made in the cell monolayer by creating a scratch with a 200-µl pipette tip. Since it was determined the pancreatic cells grow quickly, and, with the condition of 5% FBS medium it was revealed that the scratch wound could not be measured at 48 h. Thus, this assay was conducted by using 2% FBS medium to culture the cells (22). The cells were subsequently cultured in DMEM supplemented with 2% FBS at 37°C after washing with PBS three times. Images of the wound were captured at 0, 24 and 48 h using an OLYMPUS inverted microscope at a magnification of ×100, and measured by the ImageJ software (version 1.52) (National Institutes of Health), respectively, to determine the cell migratory ability.

Western blotting

Protein was extracted from transfected cells using RIPA buffer (Beyotime Institute of Biotechnology), and protein concentration was determined using a BCA kit (Beyotime Institute of Biotechnology). Briefly, 40-60 µg equal amounts of protein were loaded per lane and separated by 10% SDS-PAGE, and transferred to PVDF membranes (EMD Millipore). The PVDF membranes were blocked using 5% non-fat milk in TBST for 1 h at 37°C and then incubated with primary antibodies at 4°C overnight. Following several washes using TBST, the PVDF membranes were incubated with specific secondary antibodies [(HRP-labeled goat anti-mouse IgG; cat. no. S0002; 1:5,000) or (HRP-labeled goat anti-rabbit IgG, cat. no. S0001; 1:5,000; both from from Affinity Biosciences)] for 2 h at room temperature. Finally, after washing the membranes three times with TBST, the protein bands were visualized using an ECL kit (EMD Millipore) and the bands were quantified using ImageJ software (version 1.52) (National Institutes of Health). The following antibodies were used: Anti-FUBP1 (68 kDa; product code ab192867, 1:1,000; Abcam), anti-E-cadherin (135 kDa; product no. 3195; 1:1,000), anti-N-cadherin (140 kDa; product no. 13116; 1:1,000), anti-vimentin (57 kDa; product no. 5741; 1:1,000), anti-β-catenin (92 kDa; product no. 8480; 1:1,000), anti-Smad2/3 (52/60 kDa; product no. 3102; 1:1,000), anti-phosphorylated (p)-Smad2/3 (52/60 kDa; product no. 8828; 1:1,000), anti-TGFβ1 (12 kDa; product no. 3709; 1:1,000), and anti-β-tubulin (55 kDa; product no. 2128; 1:1,000; all from Cell Signaling Technology, Inc.) and anti-β-actin (42 kDa; cat. no. T0022, 1:5,000; Affinity Biosciences). β-actin and β-tubulin primary antibodies were used as the loading controls.

Immunofluorescence analysis

Following transfection with si-FUBP1 for 48 h, cells were cultured on glass coverslips for 48 h. The cells were subsequently fixed with 4% paraformaldehyde for 15 min at room temperature and blocked with 3% BSA (Beijing Solarbio Science & Technology Co., Ltd.) for 1 h at room temperature. The cells were then incubated with the primary antibody overnight at 4°C prior to incubation with a Cy3-conjugated Goat Anti-Rabbit IgG secondary antibody (1:500; cat. no. A0516; Beyotime Institute of Biotechnology) in a dark room for 1 h at room temperature. The cell nuclei were counterstained with DAPI (Beyotime Institute of Biotechnology) in a dark room for 15 min at room temperature. Stained cells were visualized with a fluorescence microscope (Olympus Corporation; magnification, ×200). The following primary antibodies were used: Anti-E-cadherin (cat. no. 3195; 1:200) and anti-vimentin (cat. no. 5741; 1:100; both from Cell Signaling Technology, Inc.).

Statistical analysis

Statistical analysis was performed using GraphPad Prism 5.0 software (GraphPad Software, Inc.). All data are presented as the mean ± SD of ≥3 independent experiments. Statistical differences were analyzed using a paired Student's t-test and one-way ANOVA followed by a Bonferroni's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

FUBP1 expression levels are upregulated in PAAD and positively associated with clinical prognosis

The expression levels of FUBP1 in PAAD and normal tissues were analyzed using data from TCGA database (19). The results revealed that the expression levels of FUBP1 in PAAD tissues were significantly upregulated compared with normal pancreatic tissues (Fig. 1A). Thus, the clinical significance of FUBP1 in PAAD was determined using the starBase database (20). The long-term overall survival was determined to be associated with the expression levels of FUBP1 (Fig. 1B). Thus, the patients with PAAD with upregulated FUBP1 expression levels were at a higher risk of prognosis compared with patients with lower expression levels of FUBP1. Similarly, RT-qPCR analysis of seven pairs of PAAD and adjacent normal tissues revealed that FUBP1 mRNA expression levels were significantly upregulated in PAAD tissues compared with the corresponding adjacent normal tissues (Fig. 1C). These findings indicated that FUBP1 may serve as an oncogene and may be associated with a poor prognosis in patients with PAAD.

FUBP1 promotes PAAD cell proliferation, migration and invasion

In human PAAD cell lines, PaTu8988, SW1990, BxPC-3 and Panc-1, the mRNA and protein expression levels of FUBP1 were analyzed using RT-qPCR and western blotting. Among the four human PAAD cell lines, the highest expression levels of FUBP1 were observed in PaTu8988 cells, whereas the lowest expression levels were recorded in SW1990 cells (Fig. 2A). Thus, PaTu8988 cells were selected to further investigate the effects of the knockdown of FUBP1 expression in PAAD, while SW1990 cells were selected to determine the effects of the overexpression of FUBP1. The transfection efficiencies of cells transfected with si-FUBP1 or FUBP1-OE were determined using RT-qPCR and western blotting (Fig. 2B and C).

To investigate whether FUBP1 was associated with biological functions in PAAD, a CCK-8 assay was used to determine the effect of FUBP1 on cell proliferation, while wound healing and Transwell assays were conducted to determine the role of FUBP1 on cell migration and invasion, respectively. The data demonstrated that the knockdown FUBP1 expression significantly inhibited the proliferative ability of PaTu8988 cells (Fig. 3A), whereas the overexpression of FUBP1 promoted proliferation in SW1990 cells (Fig. 3B). Following the knockdown of FUBP1 expression levels in PaTu8988 cells, the results of the Transwell assays revealed that the number of migratory and invasive cells was significantly decreased (Fig. 4A, C and D). Notably, following the upregulation of FUBP1, the number of migratory and invasive cells was significantly increased in SW1990 cells (Fig. 4B, E and F). Similarly, the results of the wound healing assay also illustrated that the knockdown of FUBP1 expression inhibited the migratory ability of PaTu8988 cells (Fig. 4G and I). By contrast, the overexpression of FUBP1 increased the migratory ability of SW1990 cells (Fig. 4H and J). These data suggested that FUBP1 may promote the proliferation, migration and invasion of PAAD cells, which implied that FUBP1 may facilitate the adhesion of tumor cells to the extracellular matrix.

FUBP1 activates epithelial-mesenchymal transition (EMT) via the TGFβ/Smad signaling pathway in PAAD cells

During tumor development, tumor cells constantly communicate with the surrounding microenvironment, which guides tumor cells undergoing EMT (23,24). EMT is regarded as a pivotal step for promoting tumor invasion and metastasis and has been discovered to serve a role in the progression of PAAD (22). Since FUBP1 was revealed to promote PAAD cell migration and invasion, it was hypothesized that FUBP1 may be involved in EMT and influence cancer metastasis. To further determine the effect of FUBP1 on the progression of PAAD, the expression levels of EMT-related genes and transcription factors were investigated using western blotting and immunofluorescence assays. Following the transfection of PaTu8988 cells with si-FUBP1, western blotting revealed that the expression levels of E-cadherin were significantly upregulated, while the expression levels of N-cadherin, vimentin and β-catenin were downregulated (Fig. 5A). In contrast, the overexpression of FUBP1 in SW1990 cells demonstrated the opposite trend (Fig. 5B). Similarly, the results of the immunofluorescence assay revealed that the knockdown of FUBP1 expression upregulated E-cadherin expression and downregulated vimentin expression (Fig. 5C). These observations suggested that FUBP1 may promote the transition of PAAD cells from an epithelial to mesenchymal phenotype.

Multiple previous studies have demonstrated that the TGFβ/Smad signaling pathway was a central regulator of cancer cell proliferation, metastasis and the EMT process (22,25,26). Thus, the present study subsequently aimed to determine the molecular mechanisms through which FUBP1 exerted its functions. In a previous study, Kyoto Encyclopedia of Genes and Genomes signaling pathway enrichment analysis identified that FUBP1 was associated with the TGFβ signaling pathway, and FUBP1 promoted the EMT of hepatocellular carcinoma cells by activating the TGFβ/Smad signaling pathway (8). Thus, whether FUBP1 could regulate EMT through the TGFβ/Smad signaling pathway in human PAAD cells was investigated. The expression levels of p-Smad2/3 and TGFβ1, which are pivotal signaling molecules in the TGFβ/Smad signaling pathway (27), were analyzed using western blotting. Notably, FUBP1 expression levels were found to be positively correlated with the expression levels of TGFβ/Smad signaling pathway target genes. The transfection of PaTu8988 cells with si-FUBP1 significantly downregulated p-Smad2/3 and TGFβ1 expression levels (Fig. 6A), while the overexpression of FUBP1 in SW1990 cells significantly upregulated the expression levels of p-Smad2/3 and TGFβ1 (Fig. 6B). Collectively, these data indicated that FUBP1 may activate EMT through TGFβ/Smad signaling in human PAAD cells.

Discussion

The results of the present study revealed that the expression levels of FUBP1 were upregulated in human PAAD tissues compared with adjacent normal tissues in both clinical tissues and data from TCGA database. Survival analysis using the starBase database also demonstrated that patients with PAAD with upregulated FUBP1 expression levels were at a higher risk of PAAD-related mortality compared with patients with lower expression levels of FUBP1. These data suggested that FUBP1 expression levels may be upregulated in patients with PAAD and may be associated with a poor prognosis, which is consistent with the findings of Fan et al (18). The results of the CCK-8 assay revealed that FUBP1 promoted cell proliferation. Moreover, wound healing and Transwell migration and invasion assays revealed that FUBP1 promoted PAAD cell migration and invasion in vitro. During EMT, epithelial cells transition from an epithelial to mesenchymal phenotype, which causes cells to lose their adhesive ability and acquire migratory and invasive properties. Characteristic changes during EMT include the downregulation of the expression levels of epithelial markers, such as E-cadherin, and the upregulation of mesenchymal markers, including N-cadherin, vimentin and EMT-related transcription factors, such as Snail, twist family bHLH transcription factor 1 and zinc finger E-box binding homeobox 1 (24). The results of the present study revealed that the knockdown of FUBP1 upregulated the expression levels of E-cadherin, downregulated vimentin, N-cadherin and β-catenin expression levels, and reversed the progression of EMT. The TGFβ/Smad signaling pathway is well established as a central regulator of cancer cell proliferation, metastasis and the EMT process (22-25). For example, a previous study demonstrated that FUBP1 promoted EMT by activating the TGFβ/Smad signaling pathway in hepatocellular carcinoma cells (8). However, to the best of our knowledge, the role of FUBP1 in TGFβ/Smad signaling pathway-mediated EMT has not been reported. Therefore, the present study hypothesized that FUBP1 may regulate the EMT process through the TGFβ/Smad signaling pathway. To address this hypothesis, the expression levels of proteins involved downstream of the TGFβ/Smad signaling pathway cascade were investigated in vitro. The results revealed that the knockdown of FUBP1 expression decreased p-Smad2/3 and TGFβ1 expression levels, while the overexpression of FUBP1 increased p-Smad2/3 and TGFβ1 expression levels. These findings suggested that FUBP1 may activate EMT via the TGFβ/Smad signaling pathway in human PAAD cells in vitro.

In conclusion, the results of the present study revealed that FUBP1 expression levels were upregulated in patients with PAAD and the upregulated expression levels predicted a poor clinical prognosis. Moreover, the findings indicated that FUBP1 may promote the proliferation, invasion and metastasis of PAAD cells. To the best of our knowledge, the present study was the first to identify that FUBP1 served as a pivotal regulator of EMT in PAAD cells by regulating the TGFβ/Smad signaling pathway. Thus, FUBP1 may serve as an oncogene that promotes PAAD cell proliferation and progression, and may serve as a clinically relevant prognostic biomarker or represent a novel therapeutic target for PAAD. Further preclinical studies and clinical trials will be required to determine whether FUBP1 can predict the benefit of prognosis in PAAD. In addition, further studies on tissues of PAAD patients are required to thoroughly understand the clinical features of FUBP1. In the future, the detailed mechanism of FUBP1, as well as the underlying effects of other genes regulated by FUBP1 in PAAD progression will be investigated.

Availability of data and materials

All data generated or analyzed during this study are included in this published article.

Authors' contributions

XZ, HC and JC conceived and designed the experiments and wrote the paper. YZ, XZ participated in all the experiments. HC and JC contributed to the design of the study and interpretation of experimental results. NZ and YL performed and analyzed the experiments of western blotting. JL performed and analyzed the experiments of immunofluorescence. XX analyzed the data and revised this manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

All patients provided written informed consent, and the study was approved by the Ethics Committee of Shanghai Fengxian District Central Hospital (Shanghai, China).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported by Shanghai Municipal Health Commission Foundation (grant no. 20204Y0181) and Shanghai University of Medicine and Health Sciences Seed Foundation (grant no. SFP-18-22-15-002).

Abbreviations:

FUBP1

far upstream element-binding protein 1;

PAAD

pancreatic adenocarcinoma;

RT-qPCR

reverse-transcription- quantitative PCR;

EMT

epithelial-mesenchymal transition;

DAB

diaminobenzidine;

FBS

fetal bovine serum;

DMEM

Dulbecco's modified Eagle's medium;

CCK-8

Cell Counting Kit-8

References

1 

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

2 

Miller KD, Nogueira L, Mariotto AB, Rowland JH, Yabroff KR, Alfano CM, Jemal A, Kramer JL and Siegel RL: Cancer treatment and survivorship statistics, 2019. CA Cancer J Clin. 69:363–385. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Stornello C, Archibugi L, Stigliano S, Vanella G, Graglia B, Capalbo C, Nigri G and Capurso G: Diagnostic delay does not influence survival of pancreatic cancer patients. United European Gastroenterol J. 8:81–90. 2020. View Article : Google Scholar : PubMed/NCBI

4 

Zhang J and Chen QM: Far upstream element binding protein 1: A commander of transcription, translation and beyond. Oncogene. 32:2907–2916. 2013. View Article : Google Scholar

5 

Debaize L and Troadec MB: The master regulator FUBP1: Its emerging role in normal cell function and malignant development. Cell Mol Life Sci. 76:259–281. 2019. View Article : Google Scholar

6 

Frost JR, Mendez M, Soriano AM, Crisostomo L, Olanubi O, Radko S and Pelka P: Adenovirus 5 E1A-Mediated Suppression of p53 via FUBP1. J Virol. 92:e00439–18. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Rabenhorst U, Thalheimer FB, Gerlach K, Kijonka M, Böhm S, Krause DS, Vauti F, Arnold HH, Schroeder T, Schnütgen F, et al: Single-stranded DNA-binding transcriptional regulator FUBP1 is essential for fetal and adult hematopoietic stem cell self-renewal. Cell Rep. 11:1847–1855. 2015. View Article : Google Scholar : PubMed/NCBI

8 

Fu PY, Hu B, Ma XL, Tang WG, Yang ZF, Sun HX, Yu MC, Huang A, Hu JW, Zhou CH, et al: Far upstream element-binding protein 1 facilitates hepatocellular carcinoma invasion and metastasis. Carcinogenesis. 41:950–960. 2020. View Article : Google Scholar

9 

Baumgarten P, Harter PN, Tonjes M, Capper D, Blank AE, Sahm F, von Deimling A, Kolluru V, Schwamb B, Rabenhorst U, et al: Loss of FUBP1 expression in gliomas predicts FUBP1 mutation and is associated with oligodendroglial differentiation, IDH1 mutation and 1p/19q loss of heterozygosity. Neuropathol Appl Neurobiol. 40:205–216. 2014. View Article : Google Scholar

10 

Jiang P, Huang M, Qi W, Wang F, Yang T, Gao T, Luo C, Deng J, Yang Z, Zhou T, et al: FUBP1 promotes neuroblastoma proliferation via enhancing glycolysis-a new possible marker of malignancy for neuroblastoma. J Exp Clin Cancer Res. 38:4002019. View Article : Google Scholar : PubMed/NCBI

11 

Duan J, Bao X, Ma X, Zhang Y, Ni D, Wang H, Zhang F, Du Q, Fan Y, Chen J, et al: Upregulation of far upstream element-binding protein 1 (FUBP1) promotes tumor proliferation and tumorigenesis of clear cell renal cell carcinoma. PLoS One. 12:e01698522017. View Article : Google Scholar : PubMed/NCBI

12 

Muller B, Bovet M, Yin Y, Stichel D, Malz M, González-Vallinas M, Middleton A, Ehemann V, Schmitt J, Muley T, et al: Concomitant expression of far upstream element (FUSE) binding protein (FBP) interacting repressor (FIR) and its splice variants induce migration and invasion of non-small cell lung cancer (NSCLC) cells. J Pathol. 237:390–401. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Chen Y, Liu J, Geng N and Feng C: Upregulation of far upstream element-binding protein 1 (FUBP1) promotes tumor proliferation and unfavorable prognosis in tongue squamous cell carcinoma. Int J Biol Markers. 35:56–65. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Yang L, Zhu JY, Zhang JG, Bao BJ, Guan CQ, Yang XJ, Liu YH, Huang YJ, Ni RZ and Ji LL: Far upstream element-binding protein 1 (FUBP1) is a potential c-Myc regulator in esophageal squamous cell carcinoma (ESCC) and its expression promotes ESCC progression. Tumour Biol. 37:4115–4126. 2016. View Article : Google Scholar

15 

Venturutti L, Cordo Russo RI, Rivas MA, Mercogliano MF, Izzo F, Oakley RH, Pereyra MG, De Martino M, Proietti CJ, Yankilevich P, et al: MiR-16 mediates trastuzumab and lapatinib response in ErbB-2-positive breast and gastric cancer via its novel targets CCNJ and FUBP1. Oncogene. 35:6189–6202. 2016. View Article : Google Scholar : PubMed/NCBI

16 

Zhang F, Tian Q and Wang Y: Far upstream element-binding protein 1 (FUBP1) is overexpressed in human gastric cancer tissue compared to non-cancerous tissue. Onkologie. 36:650–655. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Jia MY and Wang YJ: Far upstream element-binding protein 1 (FUBP1) expression differs between human colorectal cancer and non-cancerous tissue. Neoplasma. 61:533–540. 2014. View Article : Google Scholar

18 

Fan P, Ma J and Jin X: Far upstream element-binding protein 1 is up-regulated in pancreatic cancer and modulates immune response by increasing programmed death ligand 1. Biochem Biophys Res Commun. 505:830–836. 2018. View Article : Google Scholar : PubMed/NCBI

19 

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

20 

Goldman MJ, Craft B, Hastie M, Repečka K, McDade F, Kamath A, Banerjee A, Luo Y, Rogers D, Brooks AN, et al: Visualizing and interpreting cancer genomics data via the Xena platform. Nat Biotechnol. 38:675–678. 2020. View Article : Google Scholar : PubMed/NCBI

21 

Li JH, Liu S, Zhou H, Qu LH and Yang JH: StarBase v2.0: Decoding miRNA-ceRNA, miRNA-ncRNA and protein-RNA interaction networks from large-scale CLIP-Seq data. Nucleic Acids Res. 42:D92–97. 2014. View Article : Google Scholar

22 

van Staalduinen J, Baker D, Ten Dijke P and van Dam H: Epithelial-mesenchymal-transition-inducing transcription factors: New targets for tackling chemoresistance in cancer? Oncogene. 37:6195–6211. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Lamouille S, Xu J and Derynck R: Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 15:178–196. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Zhang X, Feng W, Zhang J, Ge L, Zhang Y, Jiang X, Peng W, Wang D, Gong A and Xu M: Long noncoding RNA PVT1 promotes epithelialmesenchymal transition via the TGFβ/Smad pathway in pancreatic cancer cells. Oncol Rep. 40:1093–1102. 2018.PubMed/NCBI

25 

Zhao L, Liu S, Che X, Hou K, Ma Y, Li C, Wen T, Fan Y, Hu X, Liu Y and Qu X: Bufalin inhibits TGF-β-induced epithelial-to-mesenchymal transition and migration in human lung cancer A549 cells by downregulating TGF-β receptors. Int J Mol Med. 36:645–652. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Park JH, Yoon J, Lee KY and Park B: Effects of geniposide on hepatocytes undergoing epithelial-mesenchymal transition in hepatic fibrosis by targeting TGFβ/Smad and ERK-MAPK signaling pathways. Biochimie. 113:26–34. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Valcourt U, Kowanetz M, Niimi H, Heldin CH and Moustakas A: TGF-beta and the Smad signaling pathway support transcriptomic reprogramming during epithelial-mesenchymal cell transition. Mol Biol Cell. 16:1987–2002. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2021
Volume 47 Issue 5

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang Y, Chen J, Zhou N, Lu Y, Lu J, Xing X, Chen H and Zhang X: FUBP1 mediates the growth and metastasis through TGFβ/Smad signaling in pancreatic adenocarcinoma. Int J Mol Med 47: 66, 2021
APA
Zhang, Y., Chen, J., Zhou, N., Lu, Y., Lu, J., Xing, X. ... Zhang, X. (2021). FUBP1 mediates the growth and metastasis through TGFβ/Smad signaling in pancreatic adenocarcinoma. International Journal of Molecular Medicine, 47, 66. https://doi.org/10.3892/ijmm.2021.4899
MLA
Zhang, Y., Chen, J., Zhou, N., Lu, Y., Lu, J., Xing, X., Chen, H., Zhang, X."FUBP1 mediates the growth and metastasis through TGFβ/Smad signaling in pancreatic adenocarcinoma". International Journal of Molecular Medicine 47.5 (2021): 66.
Chicago
Zhang, Y., Chen, J., Zhou, N., Lu, Y., Lu, J., Xing, X., Chen, H., Zhang, X."FUBP1 mediates the growth and metastasis through TGFβ/Smad signaling in pancreatic adenocarcinoma". International Journal of Molecular Medicine 47, no. 5 (2021): 66. https://doi.org/10.3892/ijmm.2021.4899