Open Access

STAT1 is a modulator of the expression of frequently rearranged in advanced T‑cell lymphomas 1 expression in U251 cells

  • Authors:
    • Geng Guo
    • Shule Wang
    • Yining Hao
    • Yeqing Ren
    • Yongqiang Wu
    • Jianping Zhang
    • Dong Liu
  • View Affiliations

  • Published online on: April 21, 2020     https://doi.org/10.3892/ol.2020.11555
  • Pages: 248-256
  • Copyright: © Guo 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

Aberrant expression of frequently rearranged in advanced T‑cell lymphomas 1 (FRAT1) contributes to poor prognosis in a number of carcinomas. However, its role in glioma remains controversial. In the present study, gene expression profiling was performed using Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO) functional enrichment and ingenuity pathway analysis (IPA) to evaluate the differential expression of genes and proteins in FRAT1 knockdown U251 glioma cells in comparison with the control. Western blot analysis was conducted to assess the expression levels of FRAT1 and STAT1. A total of 895 downregulated genes were identified in FRAT1‑silenced U251 cells. The most enriched processes determined by GO and KEGG analysis of the 895 differentially expressed genes were associated with proliferation, migration and invasion. According to IPA, significant canonical pathways, including the interferon, hepatic fibrosis and Wnt/β‑catenin signaling pathways, were identified to be the major enriched pathways. The elevated expression of STAT1 in U251 cells was validated. These results highlighted the regulatory role of FRAT1 in glioma cells with upregulated STAT1 expression.

Introduction

Glioma is the most common type of primary intracranial tumors in adults and is associated with a poor prognosis (16). The majority of clinical studies neglected the evaluation of survival time after the change in the World Health Organization definition that was put forward in 2000 (711). Although a limited number of patients (2-5%) survive >3 years (reported in 2007) (12,13), the median survival time of most patients is only 15 months (reported in 2012) (14). Currently, the standard treatment for patients with glioma involves surgical resection, followed by a combination of the chemotherapy drug temozolomide and radiotherapy (15,16). Despite the effective treatment strategy, the prognosis for glioma remains poor, with a median survival period of ~14.6 months and a 3-year survival rate of 10% (reported in 2009) (15). In contrast to therapies developed for other types of cancer, simple and small improvements have been made in the treatment of glioma over the recent decades; the pathophysiology of glioma remains to be clearly elucidated, and the discovery of novel molecular targets is imperative for the advanced therapy of glioma.

The frequently rearranged in advanced T-cell lymphomas 1 (FRAT1) gene is a protooncogene that was first cloned from T-cell lymphoma (17). FRAT1 acts as a positive regulator of the Wnt/β-catenin pathway (18,19) and is able to suppress glycogen synthase kinase-3 (GSK-3)-mediated phosphorylation (18,20). High expression of FRAT1 has been identified in breast, cervical, ovarian, esophageal and non-small cell lung cancer, suggesting its crucial role in malignant tumors (2126). In addition, FRAT1 knockdown has been demonstrated to inhibit the expression levels of β-catenin, cyclin D1 (CCND1) and c-myc in hepatocellular carcinoma cells under hypoxic conditions (27). A previous study has suggested that FRAT1 may be a useful molecular marker for diagnosis by acting as a prognostic indicator of glioma, and a promising candidate protein for glioma therapy (28). Although FRAT1 expression has been identified to be associated with glioma, further understanding of the detailed molecular mechanisms is required in order to improve the efficacy of conventional therapeutic regimens.

Research focusing on the genome level of diseases has become increasingly common due to the continuous advancements in biotechnology. Gene expression profiling provides an insight into the process of tumorigenesis and has been identified as an efficient method for the identification of pathogenic genes (29). Based on a recent study on the protumorigenic role of STAT1 in glioblastoma (30) and a previous study (28), FRAT1 was identified as a novel target biomarker in glioma. The aim of the present study was to elucidate the potential association between STAT1 and FRAT1 expression and to analyze the expression levels of STAT1 in glioma cells by gene expression profiling.

Materials and methods

Cell culture

Tumor cells were used to construct glioma samples as previously described (28). According to the same study (28), FRAT1 was highly expressed in U251 cells. Thus, in the current study, U251 cells were selected to observe the expression of STAT1 and investigate the mechanism of FRAT1 in glioma. U251 cell lines were purchased from the American Type Culture Collection, cultured in Dulbecco's modified Eagle's medium containing 10% fetal bovine serum (Gibco; Thermo Fisher Scientific, Inc.) and maintained in a humidified incubator (CO2 water-jacketed incubator; Thermo Electron Corporation) at 37°C in an atmosphere of 5% CO2 and 95% air.

Transfection

Transfection was performed according to a previously described method (28). To generate stable FRAT1 knockdown cell lines, the pRNAT-U6.1/Neo plasmid (GenScript), which carries the green fluorescence protein gene, was selected for the expression of short hairpin RNAs (shRNAs) targeting FRAT1. The DNA oligonucleotides (Biomics Co., Ltd) with the sense and the antisense shRNAs sequences separated by a 9 bp spacer and having BamHI and HindIII compatible overhanging ends were fused to linearized pRNAT-U6.1/Neo plasmid. All of the plasmids were confirmed by sequencing. After constructing the shRNA plasmid, the most potent plasmid for the knockdown of FRAT1 was identified, designated as pRNAT-FRAT1, which was used to knock- down FRAT1 expression in subsequent experiments.

In gene transfection, 2×105 U251 cells per well were plated onto 6-well plates and grown overnight to 60–70% confluence. Subsequently, these cells were transfected with pRNAT-FRAT1 and empty pRNAT-U6.1/Neo vector using Lipofectamine™ 2000 reagent (Invitrogen; Thermo Fisher Scientific, Inc.) under standard conditions. According to the multiplicity of infection, the amount of transfected plasmid was 0.7 µl of 6×108 TU/ml. Untransfected parental cells were used as control for stable selection. Fluorescence microscope (Olympus micropublisher 3.3RTV; Olympus Corporation) were used to observe the expression of green fluorescence protein, the cells with fluorescence ratio of ≥80% were used for subsequent experiments. And the time interval between transfection and subsequent experimentation was 168 h. The FRAT1 sequences were as follows: 5′-GAGCTGGCAAGCAGGGCAT-3′, 5′-AGCTAGTGCTCTCTGGAAA-3′ and 5′-GCAGTTACGTGCAAAGCTT-3′.

Gene expression profiling and hierarchical clustering analysis

Total RNA was extracted from U251 cell lines using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.), followed by purification on an RNeasy column (Qiagen, Inc.). The integrity of RNA was assessed using a bioanalyzer (Agilent Technologies, Inc.). The total extracted RNA (100 ng) was labeled and hybridized to Human Gene 1.0 ST microarrays (Affymetrix; Thermo Fisher Scientific, Inc.) according to the manufacturer's protocol. The differential expression of genes were calculated based on the log2-fold change between the normal U251 cells samples (4448-1, 4448-2, 4448-3) and FRAT1 knockdown (4449-1, 4449-2, 4449-3) U251 cells samples. Bidirectional hierarchical clustering was conducted for differentially expressed genes (DEGs) (31).

Enrichment analysis

Gene ontology (GO; http://www.geneontology.org/) terms were displayed as a significant network using the BiNGO plug-in of Cytoscape software (version 3.2.1) (32). The Kyoto Encyclopedia of Genes and Genomes (KEGG; http://www.genome.jp/kegg/) database was used to identify significantly enriched pathways with a false discovery rate (FDR) <0.05 (33). The Database for Annotation Visualization and Integrated Discovery was used to identify the enriched functions with FDR<0.05 set as the significance cut-off level (34).

Ingenuity pathway analysis (IPA)

The list of DEGs between FRAT1 knockdown and control FRAT1 U251 cells, which contained gene identifiers and corresponding expression values, was uploaded to the IPA software (Qiagen, Inc.) (35). IPA was used to evaluate the differentially expressed data associated with metabolic pathways, molecular networks and biological processes. Each gene identifier was matched to its corresponding gene object in the ingenuity pathway knowledge base (http://www.ingenuity.com).

Western blot analysis

Western blot analysis was performed as previously described (28). Briefly, cells were harvested and lysed, and the cleared lysates (30–50 µg/well) were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (12% gel) and transferred onto nitrocellulose membranes (EMD Millipore). After blocking for 2 h at room temperature in Tris-buffered saline (TBS; pH 7.4) with 0.1% Tween 20 (TBS-T) containing 5% non-fat dry milk, the membranes were incubated with primary mouse anti-FRAT1 (diluted 1;200; cat. no. ab108405; Abcam) and mouse anti-STAT1 (diluted 1;500; cat. no. ab3987; Abcam). Membranes were then washed in PBS-T and incubated with horseradish peroxidase-conjugated anti-mouse secondary antibody (diluted 1;5,000; cat. no. sc-2005; Santa Cruz Biotechnology, Inc.) at room temperature for 1 h. The signals were detected using enhanced chemiluminescence detection solution (Pierce; Thermo Fisher Scientific, Inc.). Protein expression was calculated by densitometry using the Scion Image software (Scion Corporation).

Results

DEGs between control and FRAT1 knockdown U251 cells

U251 cell lines were used to identify DEGs in glioma cells and to explore the changes in gene expression. A total of 1,388 genes were identified as DEGs between normal and FRAT1 knockdown cells. Of these DEGs, 493 were identified as upregulated and 895 as downregulated in FRAT1 knockdown cells compared with control U251 cells. Hierarchical cluster analysis revealed that the three clusters of FRAT1 knockdown cells were distributed within the FRAT1 knockdown cells and that the three glioma samples (4449-1, 4449-2 and 4449-3) were within the glioma sample cluster (Fig. 1). In addition, the generated scatter plot also demonstrated the differential genes expression in FRAT1 knockdown cells in comparison with the control (Fig. 2).

Functional enrichment of DEGs

To investigate the biological functions of DEGs in FRAT1 knockdown U251 cells, GO enrichment and KEGG pathway enrichment analyses were conducted. In GO enrichment analysis, the majority of enriched GO terms were biological processes (39% of the terms). DEGs were mainly enriched in the following biological processes: i) ‘Biopolymer metabolic process’; ii) ‘signal transduction’; and iii) ‘protein metabolic process’ (Fig. 3). The top 10 significantly enriched terms in biological processes were also identified (Fig. 4). In the cellular component group, DEGs were primarily enriched in the following terms: i) ‘Nucleus’; ii) ‘cytoplasm’; and ‘organelles’ (Fig. 5). In order to conduct a comprehensive analysis, the top 10 significantly enriched cellular component terms were also identified (Fig. 6). In the molecular function group, DEGs were significantly enriched in: i) ‘DNA binding’; ii) ‘transcription factor activity’; and iii) ‘receptor binding’ (Fig. 7). The top 10 significantly enriched terms in molecular function are presented in Fig. 8. The results of the KEGG pathway analysis revealed that differentially expressed genes mainly enriched in the prostate cancer and cytosolic DNA-sensing pathways following FRAT1 knockdown in glioma cells (Fig. 9).

IPA

To further identify the key genes and pathways involved in FRAT1 knockdown cells and establish the associations among these genes, IPA was performed. Based on IPA, several pathways that were associated with enriched genes were identified. ‘Interferon signaling’ was the most enriched pathway, and the activation of this pathway was significantly inhibited (Fig. 10). A diagram presenting the genes involved in the interferon (IFN) signaling pathway was generated (Fig. 11). The results demonstrated that the IFN signaling pathway may be a major pathway of FRAT1 activity in U251 cells, and demonstrated that STAT1 may act as a crucial downstream molecule of FRAT1. The expression level of STAT1 was downregulated in FRAT1 knockdown cells.

FRAT1 improves the expression of STAT1 in U251 cells

To further explore the role of FRAT1 in the expression of STAT1 in glioma cells, FRAT1 knockdown clones and control cells were used to analyze STAT1 protein expression by western blot analysis. As presented in Fig. 12, FRAT1 and STAT1 protein expression was significantly downregulated in FRAT1 knockdown clones compared with the levels in untreated cells. Thus, this experiment validated the study hypothesis that FRAT1 regulated the expression of STAT1 protein in U251 cells, suggesting the involvement of FRAT1-regulated STAT1 modulation in the occurrence and progression of glioma.

Discussion

FRAT1 is one of the main components of the Wnt signaling pathway and has been considered to positively regulate the Wnt/β-catenin signaling transduction pathway via the dissociation of GSK-3β from axin and inhibition of β-catenin phosphorylation (3639). As a result, unphosphorylated β-catenin allows nuclear translocation and transcription activation of Wnt target genes, including c-myc and CCND1, thus leading to subsequent abnormal cancer cell proliferation (40). Recently, upregulation of FRAT1 has been identified in several types of cancer and is believed to serve a role in cancer invasion, metastasis and other malignant phenotypes (41). A number of studies have demonstrated that FRAT1 serves a major role in tumor progression (2126). Previous studies revealed that low expression of FRAT1 resulted in the inhibition of human glioma cell proliferation, migration and invasion (28,42). In the current study, a total of 1,388 genes significantly differentially expressed between normal U251 and FRAT1 knockdown U251 cells were identified. Of these, 493 were upregulated and 895 were downregulated following FRAT1 knockdown. Next, GO, KEGG and IPA pathway analyses were conducted. In GO enrichment analysis, the majority of enriched GO terms were in the category of biological process. In KEGG pathway analysis, the most significant pathways were associated with cancer. The results of IPA pathway analysis revealed that 155 genes were associated with immune response, and the majority of these genes were involved in the IFN pathway. In addition, the interaction of these genes in the IFN pathway demonstrated that STAT1 was one of the main modules associated with the response to the knockdown of FRAT1, the module most significantly associated with immune response.

STATs, the downstream targets of IFN, are stimulated by tyrosine phosphorylation in the C-termini (43). Previous studies have indicated that STATs contribute to the upregulation of several genes associated with tumor cell proliferation, including c-myc, pim-1 proto-oncogene, serine/threonine kinase and CCND1 (4447). In addition, recent studies have demonstrated that pim-1 serves two distinct roles in cancer; it increases the rate of cancer cell proliferation and inhibits apoptosis (48,49). STAT1 overexpression has been reported to markedly increase the proliferation of glioma cells, whereas its suppression evidently inhibits cell proliferation (50). In addition, western blot analysis confirmed that STAT1 protein was highly expressed in U251 cells, which was reduced following the knockdown of FRAT1 in U251 cells.

The activated STAT proteins are present in various types of malignancies including leukemia, prostate cancer and neck tumors (51,52). Recent studies have indicated that STAT signaling was activated in cancer, such as in lymphoma, lung cancer and head and neck cancer, and dysregulation of this factor may contribute to oncogenesis (44,53,54). Moreover, a recent study has demonstrated that STAT1 serves a protumorigenic role in glioma (30). Furthermore, it was hypothesized that poor prognosis may be attributable to chemoresistance and/or radiation resistance of tumors that express STAT1 (30). Similarly, STATs have been identified to increase the expression of certain anti-apoptotic regulatory proteins, including the Bcl family proteins (55). STATs are considered important regulators of the development and differentiation of multicellular organisms; STAT proteins have been suggested to have primarily evolved to mediate cytokine signaling, particularly in cells of the immune system (56). Indeed, gene knockout experiments in mice indicated a pivotal role of STAT proteins in the development and regulation of the immune system (57). Thus, STAT1 may serve important roles in glioma via immune response, including: i) Proliferation; ii) inhibition of apoptosis; iii) chemoresistance; and/or iv) radiation resistance. Therefore, targeting STAT1 may be a novel therapeutic approach for the treatment of glioma. A recent study reported that oncolytic virotherapy using herpes simplex virus type I promoted glioma regression by inhibiting STAT1/3 activity; STAT1/3-induced therapeutic resistance was inhibited and, as a result, oncolytic action was promoted (58). These findings are consistent with the results of the current study.

To the best of our knowledge, the present study suggested for the first time that FRAT1 may positively regulate the Wnt/β-catenin pathway, which in turn activates target genes, including c-myc and CCND1. STAT1 mediated the upregulation of c-myc, pim-1, CCND1 and Bcl family genes, thus enhancing the proliferation of glioma cells. In addition, low expression levels of STAT1 were identified in FRAT1 knockdown U251 cells, indicating that STAT1 expression was positively regulated by FRAT1. Therefore, it was concluded that FRAT1 acted as a positive regulator of STAT1, which led to increased glioma cell proliferation, and that the protumorigenic effect of STAT1 was mediated by FRAT1. Based on a previous study on the effects of FRAT1, more experiments analyzing the role of STAT1 in glioma should be conducted in the future to validate the results of the present study. In addition, the Wnt/β-catenin and IFN/STAT1 pathways were identified to be associated in glioma through FRAT1. Investigation of the effect of IFN on FRAT1in glioma would be of great benefit in future studies.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Natural Science Foundation of China (grant. no. 81201991), the China Postdoctoral Science Foundation (grant. nos. 2015M571068 and 2016T90115) and the Beijing Postdoctoral Research Foundation (grant. nos. 2015ZZ-56 and 2016ZZ-43). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Availability of data and materials

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

Author's contributions

GG, SW and YW performed data analyses and wrote the manuscript. YH, YR, JZ and DL contributed significantly to data analyses and manuscript revision. GG, SW and YW conceived and designed the study. All authors 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 

Kleihues P, Louis DN, Scheithauer BW, Rorke LB, Reifenberger G, Burger PC and Cavenee WK: The WHO classification of tumors of the nervous system. J Neuropathol Exp Neurol. 61:215–225; discussion 226-229. 2002. View Article : Google Scholar : PubMed/NCBI

2 

Schiffer D: Classification and biology of astrocytic gliomas. Forum (Genova). 8:244–255. 1998.PubMed/NCBI

3 

Kleihues P, Soylemezoglu F, Schauble B, Scheithauer BW and Burger PC: Histopathology, classification, and grading of gliomas. Glia. 15:211–221. 1995. View Article : Google Scholar : PubMed/NCBI

4 

Wiestler OD and Wolf HK: Revised WHO classification and new developments in diagnosis of central nervous system tumors. Pathologe. 16:245–255. 1995.(In German). View Article : Google Scholar : PubMed/NCBI

5 

Louis DN, Ohgaki H, Wiestler OD, Cavenee WK, Burger PC, Jouvet A, Scheithauer BW and Kleihues P: The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol. 114:97–109. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Kleihues P and Ohgaki H: Primary and secondary glioblastomas: From concept to clinical diagnosis. Neuro Oncol. 1:44–51. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Durmaz R, Erken S, Arslantas A, Atasoy MA, Bal C and Tel E: Management of glioblastoma multiforme: With special reference to recurrence. Clin Neurol Neurosurg. 99:117–123. 1997. View Article : Google Scholar : PubMed/NCBI

8 

Hsu E, Keene D, Ventureyra E, Matzinger MA, Jimenez C, Wang HS and Grimard L: Bone marrow metastasis in astrocytic gliomata. J Neurooncol. 37:285–293. 1998. View Article : Google Scholar : PubMed/NCBI

9 

Jubelirer SJ: A review of the treatment and survival rates of 138 patients with glioblastoma multiforme. W V Med J. 92:186–190. 1996.PubMed/NCBI

10 

Park CC, Hartmann C, Folkerth R, Folkerth R, Loeffler JS, Wen PY, Fine HA, Black PM, Shafman T and Louis DN: Systemic metastasis in glioblastoma may represent the emergence of neoplastic subclones. J Neuropathol Exp Neurol. 59:1044–1050. 2000. View Article : Google Scholar : PubMed/NCBI

11 

Scott JN, Rewcastle NB, Brasher PM, Fulton D, Hagen NA, MacKinnon JA, Sutherland G, Cairncross JG and Forsyth P: Long-term glioblastoma multiforme survivors: A population-based study. Can J Neurol Sci. 25:197–201. 1998. View Article : Google Scholar : PubMed/NCBI

12 

Burton EC, Lamborn KR, Feuerstein BG, Prados M, Scott J, Forsyth P, Passe S, Jenkins RB and Aldape KD: Genetic aberrations defined by comparative genomic hybridization distinguish long-term from typical survivors of glioblastoma. Cancer Res. 62:6205–6210. 2002.PubMed/NCBI

13 

Krex D, Klink B, Hartmann C, von Deimling A, Pietsch T, Simon M, Sabel M, Steinbach JP, Heese O, Reifenberger G, et al: Long-term survival with glioblastoma multiforme. Brain. 130:2596–2606. 2007. View Article : Google Scholar : PubMed/NCBI

14 

Bleeker FE, Molenaar RJ and Leenstra S: Recent advances in the molecular understanding of glioblastoma. J Neurooncol. 108:11–27. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Stupp R, Hegi ME, Mason WP, van den Bent MJ, Taphoorn MJ, Janzer RC, Ludwin SK, Allgeier A, Fisher B, Belanger K, et al: Effects of radiotherapy with concomitant and adjuvant temozolomide versus radiotherapy alone on survival in glioblastoma in a randomised phase III study: 5-year analysis of the EORTC-NCIC trial. Lancet Oncol. 10:459–466. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B, Taphoorn MJ, Belanger K, Brandes AA, Marosi C, Bogdahn U, et al: Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 352:987–996. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Freemantle SJ, Portland HB, Ewings K, Dmitrovsky F, DiPetrillo K, Spinella MJ and Dmitrovsky E: Characterization and tissue-specific expression of human GSK-3-binding proteins FRAT1 and FRAT2. Gene. 291:17–27. 2002. View Article : Google Scholar : PubMed/NCBI

18 

Yost C, Farr GH III, Pierce SB, Ferkey DM, Chen MM and Kimelman D: GBP, an inhibitor of GSK-3, is implicated in Xenopus development and oncogenesis. Cell. 93:1031–1041. 1998. View Article : Google Scholar : PubMed/NCBI

19 

Yuan Y, Yang Z, Miao X, Li D, Liu Z and Zou Q: The clinical significance of FRAT1 and ABCG2 expression in pancreatic ductal adenocarcinoma. Tumour Biol. 36:9961–9968. 2015. View Article : Google Scholar : PubMed/NCBI

20 

Jonkers J, van Amerongen R, van der Valk M, Robanus-Maandag E, Molenaar M, Destrée O and Berns A: In vivo analysis of Frat1 deficiency suggests compensatory activity of Frat3. Mech Dev. 88:183–194. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Saitoh T, Mine T and Katoh M: Molecular cloning and expression of proto-oncogene FRAT1 in human cancer. Int J Oncol. 20:785–789. 2002.PubMed/NCBI

22 

Jonkers J, Korswagen HC, Acton D, Breuer M and Berns A: Activation of a novel proto-oncogene, Frat1, contributes to progression of mouse T-cell lymphomas. EMBO J. 16:441–450. 1997. View Article : Google Scholar : PubMed/NCBI

23 

Jonkers J, Weening JJ, van der Valk M, van der Valk M, Bobeldijk R and Berns A: Overexpression of Frat1 in transgenic mice leads to glomerulosclerosis and nephrotic syndrome, and provides direct evidence for the involvement of Frat1 in lymphoma progression. Oncogene. 18:5982–5990. 1999. View Article : Google Scholar : PubMed/NCBI

24 

Wang Y, Hewitt SM, Liu S, Zhou X, Zhu H, Zhou C, Zhang G, Quan L, Bai J and Xu N: Tissue microarray analysis of human FRAT1 expression and its correlation with the subcellular localisation of beta-catenin in ovarian tumours. Br J Cancer. 94:686–691. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Zhang Y, Han Y, Zheng R, Yu JH, Miao Y, Wang L and Wang EH: Expression of Frat1 correlates with expression of β-catenin and is associated with a poor clinical outcome in human SCC and AC. Tumour Biol. 33:1437–1444. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Zhang Y, Yu JH, Lin XY, Miao Y, Han Y, Fan CF, Dong XJ, Dai SD and Wang EH: Overexpression of Frat1 correlates with malignant phenotype and advanced stage in human non-small cell lung cancer. Virchows Arch. 459:255–263. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Fan WH, Du FJ, Liu XJ and Chen N: Knockdown of FRAT1 inhibits hypoxia-induced epithelial-to-mesenchymal transition via suppression of the Wnt/β-catenin pathway in hepatocellular carcinoma cells. Oncol Rep. 36:2999–3004. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Guo G, Kuai D, Cai S, Xue N, Liu Y, Hao J, Fan Y, Jin J, Mao X, Liu B, et al: Knockdown of FRAT1 expression by RNA interference inhibits human glioblastoma cell growth, migration and invasion. PLoS One. 8:e612062013. View Article : Google Scholar : PubMed/NCBI

29 

Gao YF, Mao XY, Zhu T, Mao CX, Liu ZX, Wang ZB, Li L, Li X, Yin JY, Zhang W, et al: COL3A1 and SNAP91: Novel glioblastoma markers with diagnostic and prognostic value. Oncotarget. 7:70494–70503. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Duarte CW, Willey CD, Zhi D, Cui X, Harris JJ, Vaughan LK, Mehta T, McCubrey RO, Khodarev NN, Weichselbaum RR and Gillespie GY: Expression signature of IFN/STAT1 signaling genes predicts poor survival outcome in glioblastoma multiforme in a subtype-specific manner. PLoS One. 7:e296532012. View Article : Google Scholar : PubMed/NCBI

31 

Wang L, Cao C, Ma Q, Zeng Q, Wang H, Cheng Z, Zhu G, Qi J, Ma H, Nian H and Wang Y: RNA-seq analyses of multiple meristems of soybean: Novel and alternative transcripts, evolutionary and functional implications. BMC Plant Biol. 14:1692014. View Article : Google Scholar : PubMed/NCBI

32 

Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B and Ideker T: Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 13:2498–2504. 2003. View Article : Google Scholar : PubMed/NCBI

33 

Wu J, Mao X, Cai T, Luo J and Wei L: KOBAS server: A web-based platform for automated annotation and pathway identification. Nucleic Acids Res. 34:W720–W724. 2006. View Article : Google Scholar : PubMed/NCBI

34 

Sherman BT, Huang da W, Tan Q, Guo Y, Bour S, Liu D, Stephens R, Baseler MW, Lane HC and Lempicki RA: DAVID Knowledgebase: A gene-centered database integrating heterogeneous gene annotation resources to facilitate high-throughput gene functional analysis. BMC Bioinformatics. 8:4262007. View Article : Google Scholar : PubMed/NCBI

35 

Kramer A, Green J, Pollard J Jr and Tugendreich S: Causal analysis approaches in ingenuity pathway analysis. Bioinformatics. 30:523–530. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Culbert AA, Brown MJ, Frame S, Hagen T, Cross DA, Bax B and Reith AD: GSK-3 inhibition by adenoviral FRAT1 overexpression is neuroprotective and induces Tau dephosphorylation and beta-catenin stabilisation without elevation of glycogen synthase activity. FEBS Lett. 507:288–294. 2001. View Article : Google Scholar : PubMed/NCBI

37 

Hagen T, Cross DA, Culbert AA, West A, Frame S, Morrice N and Reith AD: FRAT1, a substrate-specific regulator of glycogen synthase kinase-3 activity, is a cellular substrate of protein kinase A. J Biol Chem. 281:35021–35029. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Li L, Yuan H, Weaver CD, Mao J, Farr GH III, Sussman DJ, Jonkers J, Kimelman D and Wu D: Axin and Frat1 interact with Dvl and GSK, bridging Dvl to GSK in Wnt-mediated regulation of LEF-1. EMBO J. 18:4233–4240. 1999. View Article : Google Scholar : PubMed/NCBI

39 

Thomas GM, Frame S, Goedert M, Nathke I, Polakis P and Cohen P: A GSK3-binding peptide from FRAT1 selectively inhibits the GSK3-catalysed phosphorylation of axin and beta-catenin. FEBS Lett. 458:247–251. 1999. View Article : Google Scholar : PubMed/NCBI

40 

Jamieson C, Sharma M and Henderson BR: Wnt signaling from membrane to nucleus: β-catenin caught in a loop. Int J Biochem Cell Biol. 44:847–850. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Yong Z, Yu JH, Lin XY, Miao Y, Han Y, Fan CF, Dong XJ, Dai SD and Wang EH: Overexpression of Frat1 correlates with malignant phenotype and advanced stage in human non-small cell lung cancer. Virchows Archiv. 459:255–263. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Guo G, Liu B, Zhong C, Zhang X, Mao X, Wang P, Jiang X, Huo J, Jin J, Liu X and Chen X: FRAT1 expression and its correlation with pathologic grade, proliferation, and apoptosis in human astrocytomas. Med Oncol. 28:1–6. 2011. View Article : Google Scholar : PubMed/NCBI

43 

Ihle JN: STATs: Signal transducers and activators of transcription. Cell. 84:331–334. 1996. View Article : Google Scholar : PubMed/NCBI

44 

Bowman T, Garcia R, Turkson J and Jove R: STATs in oncogenesis. Oncogene. 19:2474–2488. 2000. View Article : Google Scholar : PubMed/NCBI

45 

Matikainen S, Sareneva T, Ronni T, Lehtonen A, Koskinen PJ and Julkunen I: Interferon-alpha activates multiple STAT proteins and upregulates proliferation-associated IL-2Ralpha, c-myc, and pim-1 genes in human T cells. Blood. 93:1980–1991. 1999. View Article : Google Scholar : PubMed/NCBI

46 

Bromberg JF, Wrzeszczynska MH, Devgan G, Zhao Y, Pestell RG, Albanese C and Darnell JE Jr: Stat3 as an oncogene. Cell. 98:295–303. 1999. View Article : Google Scholar : PubMed/NCBI

47 

Kharma B, Baba T, Matsumura N, Kang HS, Hamanishi J, Murakami R, McConechy MM, Leung S, Yamaguchi K, Hosoe Y, et al: STAT1 drives tumor progression in serous papillary endometrial cancer. Cancer Res. 74:6519–6530. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Lilly M and Kraft A: Enforced expression of the Mr 33,000 Pim-1 kinase enhances factor-independent survival and inhibits apoptosis in murine myeloid cells. Cancer Res. 57:5348–5355. 1997.PubMed/NCBI

49 

Moroy T, Grzeschiczek A, Petzold S and Hartmann KU: Expression of a Pim-1 transgene accelerates lymphoproliferation and inhibits apoptosis in lpr/lpr mice. Proc Natl Acad Sci USA. 90:10734–10738. 1993. View Article : Google Scholar : PubMed/NCBI

50 

Meng D, Chen Y, Yun D, Zhao Y, Wang J, Xu T, Li X, Wang Y, Yuan L, Sun R, et al: High expression of N-myc (and STAT) interactor predicts poor prognosis and promotes tumor growth in human glioblastoma. Oncotarget. 6:4901–4919. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Gouilleux-Gruart V, Gouilleux F, Desaint C, Claisse JF, Capiod JC, Delobel J, Weber-Nordt R, Dusanter-Fourt I, Dreyfus F, Groner B and Prin L: STAT-related transcription factors are constitutively activated in peripheral blood cells from acute leukemia patients. Blood. 87:1692–1697. 1996. View Article : Google Scholar : PubMed/NCBI

52 

Grandis JR, Drenning SD, Chakraborty A, Zhou MY, Zeng Q, Pitt AS and Tweardy DJ: Requirement of Stat3 but not Stat1 activation for epidermal growth factor receptor-mediated cell growth In vitro. J Clin Invest. 102:1385–1392. 1998. View Article : Google Scholar : PubMed/NCBI

53 

Gouilleux-Gruart V, Debierre-Grockiego F, Gouilleux F, Capiod JC, Claisse JF, Delobel J and Prin L: Activated Stat related transcription factors in acute leukemia. Leuk Lymphoma. 28:83–88. 1997. View Article : Google Scholar : PubMed/NCBI

54 

Bromberg JF: Activation of STAT proteins and growth control. Bioessays. 23:161–169. 2001. View Article : Google Scholar : PubMed/NCBI

55 

Catlett-Falcone R, Dalton WS and Jove R: STAT proteins as novel targets for cancer therapy. Signal transducer an activator of transcription. Curr Opin Oncol. 11:490–496. 1999. View Article : Google Scholar : PubMed/NCBI

56 

Darnell JE Jr, Kerr IM and Stark GR: Jak-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science. 264:1415–1421. 1994. View Article : Google Scholar : PubMed/NCBI

57 

Hackenmiller R, Kim J, Feldman RA and Simon MC: Abnormal Stat activation, hematopoietic homeostasis, and innate immunity in c-fes-/- mice. Immunity. 13:397–407. 2000. View Article : Google Scholar : PubMed/NCBI

58 

Delwar ZM, Kuo Y, Wen YH, Rennie PS and Jia W: Oncolytic virotherapy blockade by microglia and macrophages requires STAT1/3. Cancer Res. 78:718–730. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2020
Volume 20 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guo G, Wang S, Hao Y, Ren Y, Wu Y, Zhang J and Liu D: STAT1 is a modulator of the expression of frequently rearranged in advanced T‑cell lymphomas 1 expression in U251 cells. Oncol Lett 20: 248-256, 2020
APA
Guo, G., Wang, S., Hao, Y., Ren, Y., Wu, Y., Zhang, J., & Liu, D. (2020). STAT1 is a modulator of the expression of frequently rearranged in advanced T‑cell lymphomas 1 expression in U251 cells. Oncology Letters, 20, 248-256. https://doi.org/10.3892/ol.2020.11555
MLA
Guo, G., Wang, S., Hao, Y., Ren, Y., Wu, Y., Zhang, J., Liu, D."STAT1 is a modulator of the expression of frequently rearranged in advanced T‑cell lymphomas 1 expression in U251 cells". Oncology Letters 20.1 (2020): 248-256.
Chicago
Guo, G., Wang, S., Hao, Y., Ren, Y., Wu, Y., Zhang, J., Liu, D."STAT1 is a modulator of the expression of frequently rearranged in advanced T‑cell lymphomas 1 expression in U251 cells". Oncology Letters 20, no. 1 (2020): 248-256. https://doi.org/10.3892/ol.2020.11555