Matches in SemOpenAlex for { <https://semopenalex.org/work/W3093022892> ?p ?o ?g. }
Showing items 1 to 100 of
100
with 100 items per page.
- W3093022892 endingPage "430" @default.
- W3093022892 startingPage "418" @default.
- W3093022892 abstract "Glioma is the most common malignancy in the central nervous system with no immediate prospect of a cure. Comprehensive understanding on the pathogenesis of the disorder contributes to a better outcome. Herein, we aimed to investigate whether transcription factors erythroblast transformation-specific (ETS) transcription factor (ELF1), myeloid ecotropic viral integration site 1 (MEIS1), and growth factor independence 1 (GFI1)/F-box/WD repeat-containing protein 7 (FBW7) mediate progression of glioma. ELF1, MEIS1, and GFI1 were upregulated in glioma cells and tissues, as ELF1 was correlated with poor prognosis. Bioinformatics analysis identified the binding between ELF1 and MEIS1 as well as between GFI1 and FBW7, confirmed by chromatin immunoprecipitation (ChIP) experiments. Functional experiment indicated that silencing of ELT1 decreased MEIS1 expression and that overexpression of MEIS1 increased GFI1 expression by activating GFI1 enhancer but decreased FBW7 expression. Importantly, silencing of ELF1 decreased the capacities of proliferation, migration, and invasion of glioma cells whereas it increased apoptosis, supported by increased capase-3 and decreased matrix metalloproteinase-9 (MMP-9) and proliferating cell nuclear antigen (PCNA) expression. Moreover, an in vivo experiment confirmed the inhibitory role of silenced ELF1 in tumor growth, with a decreased level of MEIS1 and GFI1. Taken together, our study elucidated a potential mechanism that ELF1 promoted cell progression by increasing GFI1 and METS1 as well as decreasing FBW7 expression in glioma. Glioma is the most common malignancy in the central nervous system with no immediate prospect of a cure. Comprehensive understanding on the pathogenesis of the disorder contributes to a better outcome. Herein, we aimed to investigate whether transcription factors erythroblast transformation-specific (ETS) transcription factor (ELF1), myeloid ecotropic viral integration site 1 (MEIS1), and growth factor independence 1 (GFI1)/F-box/WD repeat-containing protein 7 (FBW7) mediate progression of glioma. ELF1, MEIS1, and GFI1 were upregulated in glioma cells and tissues, as ELF1 was correlated with poor prognosis. Bioinformatics analysis identified the binding between ELF1 and MEIS1 as well as between GFI1 and FBW7, confirmed by chromatin immunoprecipitation (ChIP) experiments. Functional experiment indicated that silencing of ELT1 decreased MEIS1 expression and that overexpression of MEIS1 increased GFI1 expression by activating GFI1 enhancer but decreased FBW7 expression. Importantly, silencing of ELF1 decreased the capacities of proliferation, migration, and invasion of glioma cells whereas it increased apoptosis, supported by increased capase-3 and decreased matrix metalloproteinase-9 (MMP-9) and proliferating cell nuclear antigen (PCNA) expression. Moreover, an in vivo experiment confirmed the inhibitory role of silenced ELF1 in tumor growth, with a decreased level of MEIS1 and GFI1. Taken together, our study elucidated a potential mechanism that ELF1 promoted cell progression by increasing GFI1 and METS1 as well as decreasing FBW7 expression in glioma. IntroductionMalignant gliomas are among the most common primary brain tumors in adults.1Ostrom Q.T. Gittleman H. Truitt G. Boscia A. Kruchko C. Barnholtz-Sloan J.S. CBTRUS statistical report: primary brain and other central nervous system tumors diagnosed in the united states in 2011–2015.Neuro-oncol. 2018; 20: iv1-iv86Crossref PubMed Scopus (1156) Google Scholar Due to the infiltrative nature of this disease and the localization close to eloquent brain areas, surgical resection fails to cure the disease. Patients diagnosed with a malignant glioma such as glioblastoma have to undergo a fierce clinical course with a survival time of less than 2 years for most patients.2Oike T. Suzuki Y. Sugawara K. Shirai K. Noda S.E. Tamaki T. Nagaishi M. Yokoo H. Nakazato Y. Nakano T. Radiotherapy plus concomitant adjuvant temozolomide for glioblastoma: Japanese mono-institutional results.PLoS ONE. 2013; 8: e78943Crossref PubMed Scopus (65) Google Scholar,3Lesueur P. Lequesne J. Grellard J.M. Dugué A. Coquan E. Brachet P.E. Geffrelot J. Kao W. Emery E. Berro D.H. et al.Phase I/IIa study of concomitant radiotherapy with olaparib and temozolomide in unresectable or partially resectable glioblastoma: OLA-TMZ-RTE-01 trial protocol.BMC Cancer. 2019; 19: 198Crossref PubMed Scopus (58) Google Scholar Based on advances in the molecular characterization of these tumors, disease-associated targets, including epidermal growth factor receptor (EGFR) or vascular EGFR (VEGFR), were identified, which led to the development of new approaches using traditional routes of drug development.4Chinot O.L. Wick W. Mason W. Henriksson R. Saran F. Nishikawa R. Carpentier A.F. Hoang-Xuan K. Kavan P. Cernea D. et al.Bevacizumab plus radiotherapy-temozolomide for newly diagnosed glioblastoma.N. Engl. J. Med. 2014; 370: 709-722Crossref PubMed Scopus (1645) Google Scholar, 5Gilbert M.R. Dignam J.J. Armstrong T.S. Wefel J.S. Blumenthal D.T. Vogelbaum M.A. Colman H. Chakravarti A. Pugh S. Won M. et al.A randomized trial of bevacizumab for newly diagnosed glioblastoma.N. Engl. J. Med. 2014; 370: 699-708Crossref PubMed Scopus (1779) Google Scholar, 6Boxerman J.L. Zhang Z. Safriel Y. Rogg J.M. Wolf R.L. Mohan S. Marques H. Sorensen A.G. Gilbert M.R. Barboriak D.P. Prognostic value of contrast enhancement and FLAIR for survival in newly diagnosed glioblastoma treated with and without bevacizumab: results from ACRIN 6686.Neuro-oncol. 2018; 20: 1400-1410Crossref PubMed Scopus (18) Google Scholar According to the World Health Organization (WHO) classification of tumors of the central nervous system, gliomas can be categorized into four grades (grades I–IV), among which grade IV is also called glioblastoma or glioblastoma multiforme (GBM).7Rogers T.W. Toor G. Drummond K. Love C. Field K. Asher R. Tsui A. Buckland M. Gonzales M. The 2016 revision of the WHO Classification of Central Nervous System Tumours: retrospective application to a cohort of diffuse gliomas.J. Neurooncol. 2018; 137: 181-189Crossref PubMed Scopus (30) Google Scholar Moreover, a gene expression-based molecular classification of glioblastoma has been presented, including proneural, neural, classical, and mesenchymal subtypes.8Brat D.J. Scheithauer B.W. Fuller G.N. Tihan T. Newly codified glial neoplasms of the 2007 WHO Classification of Tumours of the Central Nervous System: angiocentric glioma, pilomyxoid astrocytoma and pituicytoma.Brain Pathol. 2007; 17: 319-324Crossref PubMed Scopus (139) Google Scholar Despite the identification of these different subtypes, no effective targeted therapy for gliomas has been developed in recent decades to improve outcomes. Unfortunately, these strategies failed so far, because the complexity of the disease was underestimated and important factors such as the capability of therapeutics to pass the blood-brain barrier or to penetrate the tumor tissue were not sufficiently considered. This perspective is substantiated by the fact that areas of variant morphology exhibit significant differences in gene expression subtype within a single tumor yet harbor a large number of identical genetic alterations.9Hovelson D.H. Udager A.M. McDaniel A.S. Grivas P. Palmbos P. Tamura S. Lazo de la Vega L. Palapattu G. Veeneman B. El-Sawy L. et al.Targeted DNA and RNA sequencing of paired urothelial and squamous bladder cancers reveals discordant genomic and transcriptomic events and unique therapeutic implications.Eur. Urol. 2018; 74: 741-753Abstract Full Text Full Text PDF PubMed Scopus (38) Google ScholarErythroblast transformation-specific (ETS) family transcription factors play important roles in prostate tumorigenesis, with some acting as oncogenes and others as tumor suppressors. ETS factors compete for binding at some cis-regulatory sequences. Therefore, changes in expression of ETS family members during tumorigenesis can have complex, multimodal effects. Recent research showed that ETS transcription factor 1 (ELF1) could serve as a possible factor for tumor progression.10Feik E. Schweifer N. Baierl A. Sommergruber W. Haslinger C. Hofer P. Maj-Hes A. Madersbacher S. Gsur A. Integrative analysis of prostate cancer aggressiveness.Prostate. 2013; 73: 1413-1426Crossref PubMed Scopus (16) Google Scholar Genome-wide mapping in cell lines indicated that ELF1 has two distinct tumor suppressive roles mediated by distinct cis-regulatory sequences. ELF1 and ELF2, closely related transcription factors to ELF4, also exerted a proliferative effect in various cancer cell lines.11Ando M. Kawazu M. Ueno T. Koinuma D. Ando K. Koya J. Kataoka K. Yasuda T. Yamaguchi H. Fukumura K. et al.Mutational landscape and antiproliferative functions of ELF transcription factors in human cancer.Cancer Res. 2016; 76: 1814-1824Crossref PubMed Scopus (17) Google Scholar Furthermore, knockdown of ELF1 increased docetaxel resistance, indicating that the genomic deletions found in metastatic prostate tumors may promote therapeutic resistance through loss of c1 in glioma and can reduce its ability to recruit the transcription factor myeloid ecotropic viral integration site 1 (MEIS1), and further impair the activation ability of MEIS1 to growth factor independence 1 (GFI1) enhancer, resulting in suppression of proliferation, migration, and invasion and induction of cell apoptosis in glioma cells.12Budka J.A. Ferris M.W. Capone M.J. Hollenhorst P.C. Common ELF1 deletion in prostate cancer bolsters oncogenic ETS function, inhibits senescence and promotes docetaxel resistance.Genes Cancer. 2018; 9: 198-214Crossref PubMed Scopus (10) Google Scholar MEIS1, a transcription factor, exerts important functions in cell fate determination during development and cell proliferation.13Bhanvadia R.R. VanOpstall C. Brechka H. Barashi N.S. Gillard M. McAuley E.M. Vasquez J.M. Paner G. Chan W.C. Andrade J. et al.MEIS1 and MEIS2 expression and prostate cancer progression: a role for HOXB13 binding partners in metastatic disease.Clin. Cancer Res. 2018; 24: 3668-3680Crossref PubMed Scopus (37) Google Scholar GFI1 is located within chromosome 1p22 in the human genome, and as a zinc finger protein, GFI1 mainly functions as a transcriptional repressor by direct or functional interaction with other co-factors.14Lima K. Carlos J.A.E.G. Alves-Paiva R.M. Vicari H.P. Souza Santos F.P. Hamerschlak N. Costa-Lotufo L.V. Traina F. Machado-Neto J.A. Reversine exhibits antineoplastic activity in JAK2V617F-positive myeloproliferative neoplasms.Sci. Rep. 2019; 9: 9895Crossref PubMed Scopus (10) Google Scholar The tumor suppressive mechanisms of these normally expressed ETS factors and their interplay with oncogenic ETS factors are not well understood.In our study, we aimed to investigate the mechanism underlying ELF1 mediating the progression of glioma. Our results revealed that interference of ELF1 in glioma reduced its ability to recruit the transcription factor MEIS1 and further impaired the activation ability of MEIS1 to GFI1 enhancer in glioma cells. Additionally, an animal model was also established to detect the impact of ELF1/MEIS1/GFI1 on tumor growth.ResultsELF1 Is Highly Expressed in Glioma Tissues and Correlates with WHO Grading and KPS Score of PatientsDatasets GEO: GSE12657, GSE35493, GSE104291, and GSE50161 were analyzed by R language, and we found 1,507, 4,173, 2,784, and 4,554 differentially expressed genes, respectively. We found that there were 578 genes expressed in these four datasets through coexpression analyzing using the RobustRankAggreg pack (Figure 1A). Eight key transcription factors were obtained from hTFtarget and Cistrome, including BCL11A, EZH2, FOXM1, HDAC1, ELF1, STAT4, CBX3, and VEZF1 (Figure 1B), among which ELF1 has been implicated as being associated with glioma.15Wang M. Yang C. Liu X. Zheng J. Xue Y. Ruan X. Shen S. Wang D. Li Z. Cai H. Liu Y. An upstream open reading frame regulates vasculogenic mimicry of glioma via ZNRD1-AS1/miR-499a-5p/ELF1/EMI1 pathway.J. Cell. Mol. Med. 2020; 24: 6120-6136Crossref PubMed Scopus (10) Google Scholar The expression data from datasets GEO: GSE35493, GSE35493, GSE104291, and GSE50161 were presented in a boxplot where ELF1 was indicated as highly expressed in glioma, while Gene Expression Profiling Interactive Analysis (GEPIA) analysis of GBM data from GTEx dataset also identified the high expression of ELF1 (Figure 1C).In order to determine whether ELF1 was involved in the occurrence and development of glioma, the expression of ELF1 in brain tissues of glioma patients (n = 60) and normal brain tissues (n = 24) was detected by qRT-PCR. Compared with the normal group, the expression of ELF1 in glioma tissues was significantly increased (Figure 1D). The expression of ELF1 increased with the increase of the WHO grade of glioma (p < 0.05) (Figure 1E). In addition, we analyzed the ELF1 expression and the link between the patient clinical pathological features. According to the ELF1 average expression in gliomas (1.695), it was divided into a high- and low-expression group. The results showed that the expression of ELF1 had an obvious correlation with the WHO classification and Karnofsky performance status (KPS) scores in patients; however, there was no significant correlation between expression of ELF1 and patient’s age, sex, tumor size, and tumor recurrence (Table 1). After Kaplan-Meier analysis, the log-rank test of survival data showed that expression of ELF1 was negatively correlated with survival time and prognosis of patients (Figure 1F).Table 1The Relationship between ELF1 Expression and the Clinicopathological Characteristics of Glioma PatientsIndexNo.ELF1 Expressionp ValuesLow Expression (n = 29)High Expression (n = 31)SexMale3820180.431Female22913Age (Years)≥633516190.794<63251312Tumor Diameter (mm)≥53315180.796<5271413TNMI∼II29281< 0.001III∼IV31130KPS≥7035278< 0.001<7025223RelapseYes3714230.062No23158TNM, tumor-node-metastasis. Open table in a new tab Silencing ELF1 Inhibits the Proliferation, Migration, and Invasion of Glioma Cells and Promotes Cell ApoptosisIn view of the significant upregulation of ELF1 in glioma tissues, in order to determine how it affected the proliferation, migration, and invasion abilities of glioma cells, we performed functional experiments on glioma cells. We constructed small interfering RNA (siRNA) specific to ELF1 (si-ELF1-1, si-ELF1-2, si-ELF1-3), and si-ELF1 exhibited the most significant interference on ELF1 expression, according to the results from qRT-PCR (Figure S4A). After transfection with si-ELF1, the expression of ELF1 was significantly decreased in glioma cells A172, U251, and T98G according to results of qRT-PCR and western blot analysis (Figure 2A; Figure S1A). From the results of a Cell Counting Kit-8 (CCK-8) assay, transwell assay, and annexin V/propidium iodide (PI) dual staining, glioma cell abilities of proliferation, migration, and invasion were significantly inhibited and cell apoptosis was induced after interference of ELF1 (Figures 2B–2E; Figures S1B–S1E). Proliferation-related factor proliferating cell nuclear antigen (PCNA), invasion-related factor matrix metalloproteinase-9 (MMP-9), and apoptosis-related factor cleaved caspase-3 expression was detected by western blot analysis, and results showed that compared with the si-negative control (NC) group, expressions of PCNA and MMP-9 in the si-ELF1 group were significantly decreased, while expressions of cleaved caspase-3 were significantly increased (Figure 2F; Figure S1F).Figure 2Silencing ELF1 Inhibits Proliferation, Migration, and Invasion and Promotes Apoptosis of A172 and U251 Glioma CellsShow full caption(A) Expression of ELF1 in A172 and U251 glioma cells upon si-NC or si-ELF1 was tested by qRT-PCR and western blot analysis. (B) CCK-8 assay of proliferation of A172 and U251 cells upon treatment with si-NC or si-ELF1. (C) Transwell assay of migration of A172 and U251 cells upon treatment with si-NC or si-ELF1 (original magnification, ×200). (D) Transwell assay of migration of A172 and U251 cells upon treatment with si-NC or si-ELF1 (original magnification, × 200). (E) Apoptosis rate of A172 and U251 cells was determined by annexin V/PI flow cytometry. (F) Western blot analysis was used to detect the expression of proliferation-related factor PCNA, invasion-related factor MMP-9, and apoptosis-related factor cleaved caspase-3 of A172 and U251 cells upon treatment with si-NC or si-ELF1. The above values are all measurement data, expressed as mean ± standard deviation. ∗p < 0.05 compared with the si-NC group. An unpaired t test was used between the two groups, and the data of each group at different time points were compared. The cell experiment was repeated three times.View Large Image Figure ViewerDownload Hi-res image Download (PPT)Transcription Factor ELF1 Binds to the MEIS1 Promoter to Promote Its Transcription and Promote the Development of GliomaIn the above studies, we have identified that ELF1 was highly expressed in glioma tissues and can significantly inhibit the proliferation, migration, and invasion of glioma cells after specific interference treatment. Then, we continued our review of relevant literature and found that ELF1, a transcription factor, can be combined into MEIS1 promoter regions, thus affecting the transcription.16Xiang P. Lo C. Argiropoulos B. Lai C.B. Rouhi A. Imren S. Jiang X. Mager D. Humphries R.K. Identification of E74-like factor 1 (ELF1) as a transcriptional regulator of the Hox cofactor MEIS1.Exp. Hematol. 2010; 38: 798-808.e2Abstract Full Text Full Text PDF PubMed Scopus (19) Google Scholar The normalized data of ELF1 and MEIS1 of 84 samples from datasets GEO: GSE12657, GSE35493, GSE104291, and GSE50161 identified a positive correlation between ELF1 and MEIS1 (Figure 3A). GEPIA analysis confirmed the positive correlation upon analysis of GBM data from The Cancer Genome Atlas (TCGA) dataset (Figure 3B). The analysis of the four datasets also revealed that MEIS1 was significant highly expressed in glioma tissues in the GEO: GSE35493 and GSE35492 datasets, while it was highly expressed in GEO: GSE104291 and GSE50161, but not significantly (p > 0.05) (Figure 3C). Based on this evidence, we hypothesized that ELF1 may affect glioma development by promoting the transcription of MEIS1. We initially detected MEIS1 expression in glioma tissues through the qRT-PCR and western blot analysis and identified the elevated expression of MEIS1 in glioma tissues relative to normal brain tissues (Figure 3D). The binding site between ELF1 and MEIS1 promoter obtained by the JASPAR dataset (Figure 3E; Table S1) was detected by a chromatin immunoprecipitation (ChIP) experiment. We applied magnetic protein A beads to precipitation and found that compared with the immunoglobulin G (IgG) group, the promoter DNA of binding MEIS1 in the ELF1 group was significantly increased (Figure 3F; Figure S2A). When si-ELF1 plasmids were transfected into glioma cells, mRNA and protein expressions of MEIS1 were significantly decreased (Figure 3G; Figure S2B).Figure 3ELF1 Binds to the MEIS1 Promoter to Promote the Expression of MEIS1 to Participate in the Development of GliomaShow full caption(A) Normalized expression correlation diagram of ELF1 and MEIS1 drawn from GEO: GSE50161, GSE35493, GSE12657, and GSE104291 datasets. (B) Expression correlation diagram of ELF1 and MEIS1 through GEPIA analysis of GBM data from TCGA dataset. (C) Boxplot of MEIS1 expression from dataset GEO: GSE12657, GSE35493, GSE50161, and GSE104291, as well as GBM data from TCGA dataset and GTEx through GEPIA analysis. ∗p < 0.01. (D) qRT-PCR analysis of MEIS1 expression in clinical glioma tissues (n = 60) and normal brain tissues (n = 24). (E) Binding site of ELF1 and METS1 promoter through the JASPAR dataset. (F) ChIP experiment of METS1 enrichment level in the A172 and U251 cells. (G) Western blot analysis of MEIS1 protein expression in A172 and U251 cells. (H) CCK-8 assay of proliferation of A172 and U251 cells upon treatment with si-NC + oe-NC, si-ELF1 + oe-NC, and si-ELF1 + oe-MEIS1. (I) Transwell assay of migration of A172 and U251 cells upon treatment with si-NC + oe-NC, si-ELF1 + oe-NC, and si-ELF1 + oe-MEIS1 (original magnification, ×200). (J) Transwell assay of invasion of A172 and U251 cells upon treatment with si-NC + oe-NC, si-ELF1 + oe-NC, and si-ELF1 + oe-MEIS1 (original magnification, ×200). (K) Annexin V/PI flow cytometry of A172 and U251 cells upon treatment with si-NC + oe-NC, si-ELF1 + oe-NC, and si-ELF1 + oe-MEIS1. (L) Western blot analysis of PCNA, MMP-9, and cleaved capase-3 protein expression in A172 and U251 cells upon treatment with si-NC + oe-NC, si-ELF1 + oe-NC, and si-ELF1 + oe-MEIS1. ∗p < 0.05 compared with input group or si-NC + oe-NC group; #p < 0.05 compared with IgG group or si-ELF1+ oe-NC group. The data between two groups were analyzed by an unpaired t test. The data among multiple groups were analyzed by ANOVA with Tukey’s post hoc test. Data at different time points among groups were compared by two-way ANOVA, followed by a Bonferroni post-test. The cell experiment was repeated three times.View Large Image Figure ViewerDownload Hi-res image Download (PPT)Furthermore, to determine whether ELF1 was involved in the development of glioma through affecting the transcription of MEIS1, we transfected si-NC + overexpression (oe)-NC, si-ELF1+ oe-NC, and si-ELF1 + oe-MEIS1 into glioma cells A172, U251, and T98G, followed by transwell and CCK-8 assays, as well as annexin V/PI staining. It was clear that treatment with si-ELF1 + oe-NC decreased cell proliferation, migration, and invasion ability, whereas it increased apoptosis, in comparison with that of the control group. Based on si-ELF1, addition of oe-MEIS1 reversed the effect of ELF1 interference on glioma cells, promoting the growth, migration, and invasion of glioma cells, and reducing cell apoptosis (Figures 3H–3K; Figures S2C–S2F).Western blot analysis was then conducted to analyze proliferation factor PCNA, invasion-related protein MMP-9, and apoptosis key factor cleaved caspase-3 upon treatments. The expression of PCNA and MMP-9 in the si-ELF1 + oe-NC group was decreased and the expression of cleaved caspase-3 was increased. Similarly, overexpression of MEIS1 increased the expression of PCNA and MMP-9, and decreased cleaved caspase-3, restoring the expression of these key factors (Figure 3L; Figure S2G). These results suggested that the transcription factor ELF1 may be involved in glioma development by enhancing MEIS1 transcription in glioma.MEIS1 Promotes Glioma Development by Regulating the Activity of the GFI1 PromoterMulti Experiment Matrix (MEM) analysis, a visualization tool gathering publicly available gene expression data and ranking genes by their similarity, in the current study pointed to a co-expression relationship between MEIS1 and GFI1 (Figure 4A). To confirm this relationship in tissues, we then detected GFI1 expression in the glioma and normal control tissues by qRT-PCR and western blot analysis, and found that expression of GFI1 in glioma tissues was higher (Figure 4B). When glioma cells were transfected with overexpression of MEIS1, as demonstrated by western blot analysis, GFI1 expression in cells was increased (Figure 4C; Figure S3A). Then, the ChIP experiment was used to verify the relationship between MEIS1 and GFI1. Overexpression of MEIS1 in A172, U251, and T98G cells led to the enrichment of H3K4me1, H3K27ac, and MEIS1 in the GFI1 promoter and enhancer region with similar results observed in the three cell lines. These results indicated that MEIS1 promoted the expression of GFI1 by activating the enhancer of GFI1 (Figures 4D and 4E; Figures S3B and S3C).Figure 4MEIS1 Regulates GFI1 Enhancer Activity in A172 and U251 Glioma CellsShow full caption(A) MEM analysis of co-expression relationship between MEIS1 and GFI1. (B) qRT-PCR analysis of GFI1 expression in glioma tissues and normal brain tissues. (C) Western blot analysis of GFI1 expression in A172 and U251 cells upon treatment with oe-NC or oe-MEIS1. (D) ChIP of enrichment of H3K4me1, H3K27ac, and MEIS1 in the GFI1 enhancer region upon treatment with oe-NC or oe-MEIS1. (E) ChIP detected the enrichment of H3K4me1, H3K27ac, and MEIS1 in the promoter region of GFI1 upon treatment with oe-NC or oe-MEIS1. (F) Flow cytometry of A172 and U251 cell proliferation upon treatment with oe-NC + si-NC, oe-MEIS1 + si-NC, or oe-MEIS1 + si-GFI1. (G) Transwell assay of migration of A172 and U251 cells upon treatment with oe-NC + si-NC, oe-MEIS1 + si-NC, or oe-MEIS1 + si-GFI1 (original magnification, ×200). (H) Transwell assay of invasion of A172 and U251 cells upon treatment with oe-NC + si-NC, oe-MEIS1 + si-NC, or oe-MEIS1 + si-GFI1 (original magnification, ×200). (I) Flow cytometry of apoptotic rate of A172 and U251 cells upon treatment with oe-NC + si-NC, oe-MEIS1 + si-NC, or oe-MEIS1 + si-GFI1. (J) Western blot analysis of PCNA, MMP-9 and cleaved capase-3 protein expression in A172 and U251 cells upon treatment with oe-NC + si-NC, oe-MEIS1 + si-NC, or oe-MEIS1 + si-GFI1. (K) Correlation diagram of GFI1 and FBX7 expression through normalization of GEO: GSE50161, GSE104291, and GSE3549 datasets. (L) Boxplot of FBX7 expression from GEO: GSE12657, GSE35493, GSE50161, and GSE104291 datasets, as well as GBM data from TCGA dataset and GTEx through GEPIA analysis. (M) MEM analysis of co-expression relationship between GFI1 and FBW7. (N) Western blot analysis of FBW7 protein expression in glioma cells upon all treatments. ∗p < 0.05. Measurement data are expressed as mean ± standard deviation. The data between two groups were analyzed by an unpaired t test. The data among multiple groups were analyzed by ANOVA with Tukey’s post hoc test. Data at different time points among groups were compared by two-way ANOVA, followed by a Bonferroni post-test. The cell experiment was repeated three times.View Large Image Figure ViewerDownload Hi-res image Download (PPT)To further explore the impact between MEIS1 and GFI1 on glial development, we treated U251 cells with overexpressed MEIS1 or interfered GFI1 simultaneously, followed by a CCK-8 assay, transwell assay, and flow cytometry. Compared with the oe-NC+ si-NC group, cell proliferation, migration, and invasion were increased but the apoptosis rate was decreased in the oe-MEIS1 + si-NC group. The interference plasmids of GFI1 (si-GFI1-1, si-GFI1-2, si-GFI1-3) were established and si-GFI1-1 with greatest efficiency of interference was selected to transfect to oe-MEIS1-treated cells (Figure S4B). However, oe-MEIS1 + si-GFI1 reversed the effect of overexpressing MEIS1 on cell progression (Figures 3D–3G and 4F–4I). These results were supported by following the detection of MMP-9, cleaved caspase-3, and PCNA in the cells upon treatment. As displayed in Figure 4J and Figure S3H, interference with GFI1 could reverse the effect of overexpression of MEIS1 on all related proteins, inhibiting the expression of PCNA and MMP-9, and promoting the expression of cleaved caspase-3.In the occurrence of cervical cancer, as reported, GFI1 can inhibit the expression of F-box/WD repeat-containing protein 7 (FBW7) through its correlation with FBW7.17Cai H. Zhang F. Li Z. Gfi-1 promotes proliferation of human cervical carcinoma via targeting of FBW7 ubiquitin ligase expression.Cancer Manag. Res. 2018; 10: 2849-2857Crossref PubMed Scopus (4) Google Scholar Since the GEO: GSE12657 dataset does not include the expression data for FBW7 (named FBXW7 in NCBI), we only normalized the data concerning FBW7 from the other three datasets to assess the correlation between GFI1 and FBW7. We found that there was a negative correlation between GFI1 and FBW7 (Figure 4K). Analysis from the datasets GEO: GSE35493, GSE104291, and GSE35493 indicated poorly expressed expression of FBW7 in glioma, consistent with the results from TCGA and GTEx datasets analyzed by GEPIA (Figure 4L). GFI1 and FBW7 had a significantly co-expressed relationship, as evidenced by MEM analysis (Figure 4M). In addition, we continued to detect the expression of FBW7 in glioma cells by western blot analysis after transfection with MEIS1 or GFI1. As shown in Figure 4N and Figure S3I, compared with the oe-NC + si-NC group, FBW7 expression was significantly lower in the oe-MEIS1 + si-NC group, but additional treatment with si-GFI1 hardly altered expression of FBW7. This evidence elucidated a mechanism that MEIS1 inhibited FBW7 expression through mediating the activity of GFI1 and thereby promotes proliferation, migration, and invasion and inhibits apoptosis of glioma cells.Interference with ELF1 Can Inhibit Glioma Progression In Vivo by the MEIS1/GFI1/FBW7 AxisTo confirm the in vivo anti-tumor effect of ELF1, we developed a mouse model. First, we silenced ELF1 expression in U251 cells and transplanted the cells into the nude mice. Every week, we checked the weight and volume of the tumors. The results showed that the weight and volume of tumor in mice treated with sh-ELF1 were lower than those in the control group (Figures 5A–5C). In addition, western blot analysis showed that sh-ELF1 treatment significantly reduced the expression of ELF1, MEIS1, and GFI1, accompanied with elevated expression of FBW7 (Figure 5D). These results indicated that interference with" @default.
- W3093022892 created "2020-10-22" @default.
- W3093022892 creator A5008140932 @default.
- W3093022892 creator A5025846619 @default.
- W3093022892 creator A5026115126 @default.
- W3093022892 creator A5033510051 @default.
- W3093022892 creator A5062372256 @default.
- W3093022892 creator A5091642462 @default.
- W3093022892 date "2021-03-01" @default.
- W3093022892 modified "2023-09-30" @default.
- W3093022892 title "Transcription Factor ELF1 Activates MEIS1 Transcription and Then Regulates the GFI1/FBW7 Axis to Promote the Development of Glioma" @default.
- W3093022892 cites W1847790984 @default.
- W3093022892 cites W1984806209 @default.
- W3093022892 cites W2022859149 @default.
- W3093022892 cites W2046461201 @default.
- W3093022892 cites W2074071454 @default.
- W3093022892 cites W2084963464 @default.
- W3093022892 cites W2110817748 @default.
- W3093022892 cites W2124736921 @default.
- W3093022892 cites W2129458270 @default.
- W3093022892 cites W2146511318 @default.
- W3093022892 cites W2146512944 @default.
- W3093022892 cites W2156330848 @default.
- W3093022892 cites W2287195562 @default.
- W3093022892 cites W2366536035 @default.
- W3093022892 cites W2775661709 @default.
- W3093022892 cites W2792671843 @default.
- W3093022892 cites W2799978578 @default.
- W3093022892 cites W2884639138 @default.
- W3093022892 cites W2888082724 @default.
- W3093022892 cites W2919931933 @default.
- W3093022892 cites W2928359858 @default.
- W3093022892 cites W2945010680 @default.
- W3093022892 cites W2949599758 @default.
- W3093022892 cites W2950503376 @default.
- W3093022892 cites W2959888234 @default.
- W3093022892 cites W3023549264 @default.
- W3093022892 cites W4234865248 @default.
- W3093022892 doi "https://doi.org/10.1016/j.omtn.2020.10.015" @default.
- W3093022892 hasPubMedCentralId "https://www.ncbi.nlm.nih.gov/pmc/articles/7787950" @default.
- W3093022892 hasPubMedId "https://pubmed.ncbi.nlm.nih.gov/33473327" @default.
- W3093022892 hasPublicationYear "2021" @default.
- W3093022892 type Work @default.
- W3093022892 sameAs 3093022892 @default.
- W3093022892 citedByCount "10" @default.
- W3093022892 countsByYear W30930228922021 @default.
- W3093022892 countsByYear W30930228922022 @default.
- W3093022892 countsByYear W30930228922023 @default.
- W3093022892 crossrefType "journal-article" @default.
- W3093022892 hasAuthorship W3093022892A5008140932 @default.
- W3093022892 hasAuthorship W3093022892A5025846619 @default.
- W3093022892 hasAuthorship W3093022892A5026115126 @default.
- W3093022892 hasAuthorship W3093022892A5033510051 @default.
- W3093022892 hasAuthorship W3093022892A5062372256 @default.
- W3093022892 hasAuthorship W3093022892A5091642462 @default.
- W3093022892 hasBestOaLocation W30930228921 @default.
- W3093022892 hasConcept C104317684 @default.
- W3093022892 hasConcept C138885662 @default.
- W3093022892 hasConcept C179926584 @default.
- W3093022892 hasConcept C2778227246 @default.
- W3093022892 hasConcept C41895202 @default.
- W3093022892 hasConcept C502942594 @default.
- W3093022892 hasConcept C54355233 @default.
- W3093022892 hasConcept C81350143 @default.
- W3093022892 hasConcept C86339819 @default.
- W3093022892 hasConcept C86803240 @default.
- W3093022892 hasConcept C95444343 @default.
- W3093022892 hasConceptScore W3093022892C104317684 @default.
- W3093022892 hasConceptScore W3093022892C138885662 @default.
- W3093022892 hasConceptScore W3093022892C179926584 @default.
- W3093022892 hasConceptScore W3093022892C2778227246 @default.
- W3093022892 hasConceptScore W3093022892C41895202 @default.
- W3093022892 hasConceptScore W3093022892C502942594 @default.
- W3093022892 hasConceptScore W3093022892C54355233 @default.
- W3093022892 hasConceptScore W3093022892C81350143 @default.
- W3093022892 hasConceptScore W3093022892C86339819 @default.
- W3093022892 hasConceptScore W3093022892C86803240 @default.
- W3093022892 hasConceptScore W3093022892C95444343 @default.
- W3093022892 hasLocation W30930228921 @default.
- W3093022892 hasLocation W30930228922 @default.
- W3093022892 hasLocation W30930228923 @default.
- W3093022892 hasOpenAccess W3093022892 @default.
- W3093022892 hasPrimaryLocation W30930228921 @default.
- W3093022892 hasRelatedWork W1491894775 @default.
- W3093022892 hasRelatedWork W1967633405 @default.
- W3093022892 hasRelatedWork W2002456318 @default.
- W3093022892 hasRelatedWork W2018096124 @default.
- W3093022892 hasRelatedWork W2056372829 @default.
- W3093022892 hasRelatedWork W2058339758 @default.
- W3093022892 hasRelatedWork W2091361307 @default.
- W3093022892 hasRelatedWork W2353625357 @default.
- W3093022892 hasRelatedWork W2914991022 @default.
- W3093022892 hasRelatedWork W3166104415 @default.
- W3093022892 hasVolume "23" @default.
- W3093022892 isParatext "false" @default.
- W3093022892 isRetracted "false" @default.
- W3093022892 magId "3093022892" @default.
- W3093022892 workType "article" @default.