Open Access

HNF4G accelerates glioma progression by facilitating NRP1 transcription

  • Authors:
    • Hongmin Che
    • Qi Zheng
    • Zijun Liao
    • Lu Zhang
  • View Affiliations

  • Published online on: February 1, 2023     https://doi.org/10.3892/ol.2023.13688
  • Article Number: 102
  • Copyright: © Che 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

Hepatocyte nuclear factor 4γ (HNF4G) is considered to be a transcription factor and functions as an oncogene in certain types of human cancer. However, the precise functions and the potential molecular mechanisms of HNF4G in glioma remain unclear. Therefore, the present study aimed to elucidate the role of HNF4G in glioma and the underlying mechanism. Western blotting and reverse transcription‑quantitative PCR (RT‑qPCR) demonstrated that HNF4G was highly expressed in glioma tissues and cell lines. The overexpression of HNF4G in LN229 and U251 glioma cells promoted cell proliferation and cell cycle progression, and inhibited apoptosis, while the knockdown of HNF4G suppressed cell proliferation, cell cycle progression and tumor growth, and induced apoptosis. A significant positive association was detected between HNF4G and neuropilin‑1 (NRP1) mRNA expression in glioma tissues. Bioinformatics analysis, chromatin immunoprecipitation‑RT‑qPCR and promoter reporter assays confirmed that HNF4G promoted NRP1 transcription in glioma by binding to its promoter. NRP1 overexpression facilitated glioma cell proliferation and cell cycle progression, and suppressed apoptosis in vitro, while the knockdown of NRP1 inhibited cell proliferation and cell cycle progression, and facilitated apoptosis. NRP1 overexpression reversed the effects induced by HNF4G knockdown on glioma cell proliferation, cell cycle progression and apoptosis. In summary, the present study demonstrated that HNF4G promotes glioma cell proliferation and suppresses apoptosis by activating NRP1 transcription. These findings indicate that HNF4G acts as an oncogene in glioma and may thus be an effective therapeutic target for glioma.

Introduction

Glioma is the most common and lethal intracranial tumor, posing a severe threat to human health and life (1); it is characterized by high morbidity and relapse rates, and a poor survival rate. The annual morbidity rate of cerebral glioma is ~6.1 per 100,000 individuals (2). Depending on its origin, glioma can be classified into astrocytoma or oligodendroglioma. Glioma is divided into grades I–IV by the World Health Organization (WHO); the median survival rates of patients with grades III and IV glioma are 2 and 1 year, respectively (3). Despite numerous advances being made in surgical excision, chemotherapy, radiotherapy, targeted therapy and gene therapy, the currently available therapeutic methods are not sufficient to completely eliminate the glioma, and the therapeutic effect remains unsatisfactory. Therefore, a more in-depth understanding of the precise mechanisms responsible for the development of gliomas is urgently required, which may be beneficial for the identification of novel therapeutic targets.

Hepatocyte nuclear factor 4 (HNF4), which includes HNF4A and HNF4G isoforms, belongs to the nuclear hormone receptor superfamily (4). HNF4G is considered to be a transcription factor. HNF4G is detected in the human kidneys, stomach, pancreas, lungs, bladder and testicles (5). Previous studies have demonstrated that HNF4G is associated with glucose metabolism and hyperuricemia (6,7). In addition, several studies have observed that HNF4G functions as an oncogene, regulating cell growth, apoptosis and invasion in certain types of human cancer, including pancreatic, prostate, bladder, liver and lung cancer (811). However, the function of HNF4G in several other types of tumors, including glioma, has not yet been fully elucidated. In particular, the possible molecular mechanisms underlying the role of HNF4G in gliomas remain unclear.

Therefore, the present study measured the expression of HNF4G in patients with glioma and investigated the function and molecular mechanisms of HNF4G in glioma progression. The present study compared HNF4G expression levels in glioma specimens and cell lines with those in adjacent non-tumor tissues and normal cells, respectively. The association of the mRNA expression of HNF4G and the clinicopathological features of patients with glioma was investigated. In addition, the effect of HNF4G on glioma cell proliferation was examined in vitro and in vivo. Furthermore, molecular mechanistic analyses were conducted to investigate whether HNF4G affected glioma cell proliferation and tumor growth via neuropilin-1 (NRP1).

Materials and methods

Human glioma samples

Human glioma specimens and adjacent non-tumor tissues were obtained during glioma surgery in 59 patients (38 men and 21 women; 33 cases ≥50 years old, 26 cases <50 years old; mean age, 52 years old) between April 2019 and October 2020 at the Department of Pathology, Xi'an Gaoxin Hospital (Xi'an, China). Written informed consent was obtained from each patient. The patients had not been treated with radiotherapy, chemotherapy or other treatment prior to the surgery. Three small sections from each tissue were promptly frozen and stored at −80°C for use in the following experiments. The patient samples were divided into high and low expression groups based on the median gene expression levels (12). The present study was approved by the Ethics Committee of Xi'an Gaoxin Hospital (Xi'an, China; approval no. GXYY-XA-H-2022-068).

Cells and cell culture

The human glioma U87, LN229 and U251 cell lines were purchased from Procell Life Science &Technology Co., Ltd. and immortalized normal human astrocytes (NHAs) were purchased from Shanghai BinsuiBio Co., Ltd. The U87 cell line is not the original glioblastoma cell line established in 1968 at the University of Uppsala; it is the U87 MG ATCC version (CVCL_UE09), which is most probably a glioblastoma, but whose origin is unknown. The cells were cultured in DMEM (Gibco; Thermo Fisher Scientific, Inc.) with 10% fetal calf serum (Gibco; Thermo Fisher Scientific, Inc.) and 1% penicillin-streptomycin (Gibco; Thermo Fisher Scientific, Inc.) at 37°C with 5% CO2.

Animals

A total of 4 male BALB/c nude mice (5 weeks old; 29.1±1.7 g) were purchased from Shanghai SLAC Laboratory Animal Co., Ltd., and fed under pathogen-free conditions. Mice were maintained at a temperature of 26°C, with 50% relative humidity, ventilation 13 times/h and a 10-h light and 14-h dark cycle/day. The food was autoclaved and the drinking water was sterile water with a mixture of vitamins, and both were available ad libitum. The Institutional Animal Care and Use Committee of Xi'an Gaoxin Hospital approved the animal experiments (approval no. GXYY-XA-A-2022-039).

Plasmid construction and transfection

Full-length DNA sequences of HNF4G or NRP1 were incorporated into the pCMV2-GV146 plasmid (Genechem Co. Ltd.), respectively. A reporter plasmid (pGL3-NRP1-luc; Beijing AuGCT DNA-SYN Biotechnology) was also constructed, which included the 490-bp DNA fragment 33484619-33485108 relative to the NRP1 promoter, which was located upstream of the firefly luciferase reporter gene in pGL3-luc. The pGL3-luc and pGL3-NRP1-luc plasmids were transfected into LN229 and U251 glioma cells at 37°C for 48 h using Jet Prime (Polyplus-transfection SA).

Transfection with small interfering RNA (siRNA)

siRNAs were purchased from Shanghai GenePharma Co., Ltd., for the knockdown of HNF4G and NRP1 gene expression. Human HNF4G siRNA-1 (sense, 5′-CGGCACUACAUAAAUGUGATT-3′ and antisense, 5′-UCACAUUUAUGUAGUGCCGTT-3′), HNF4G siRNA-2 (sense, 5′-CGAGUGAGAGAAACACAUUTT-3′ and antisense, 5′-AAUGUGUUUCUCUCACUCGTT-3′), NRP1 siRNA-1 (sense, 5′-GGUUUCUCAGCAAACUACATT-3′ and antisense, 5′-UGUAGUUUGCUGAGAAACCTT-3′), NRP1 siRNA-2 (sense, 5′-CUGGCAUAUCUAUGAGAUUTT-3′ and antisense, 5′-AUCUCAUAGAUAUGCCAGTT-3′) and negative control siRNA (NC-siRNA sense, 5′-UUCACCGACUUUGUCACGUTT-3′ and antisense, 5′-ACGUGACAAAGUCGGUGAATT-3′) were transiently transfected into LN229 and U251 cells at the concentration of 50 nM at 37°C for 24, 48 or 72 h using Jet Prime (Polyplus-transfection SA).

Cell proliferation assay

The LN229 and U251 cells were independently suspended at 20,000 cells/ml in DMEM with 10% fetal calf serum and seeded in 96-well plates (200 µl/well). These cells were transiently transfected with the control vector, HNF4G overexpression vector, NRP1 overexpression vector, NC-siRNA (80 nM), HNF4G siRNA-1, HNF4G siRNA-2, NRP1 siRNA-1, NRP1 siRNA-2, HNF4G siRNA-2 + control vector, or HNF4G siRNA-2 + NRP1 overexpression vector for 24, 48 or 72 h. Cell growth was measured using an MTT assay (MilliporeSigma). MTT diluent (20 µl) was added to each well followed by culturing for 4 h. Dimethylsulfoxide (150 µl) was then added to dissolve the formazan salt. The optical density at 492 nm was examined using a microplate reader (BMG Labtech GmbH).

Cell cycle assay

The LN229 and U251 glioma cells were harvested at 24 h following transfection. The cells were washed using PBS and immobilized with 70% ethyl alcohol at 4°C. The cells were then washed with PBS and prepared as a single cell suspension. Propidium iodide (PI; 0.05 mg/ml; MilliporeSigma) containing RNase A (0.1 mg/ml) was added to each group of cells followed by incubation at room temperature for 10 min for staining. Cell cycle analysis was performed by a flow cytometer (EPICS XL; Beckman Coulter, Inc.) using SYSTEM II Software (version 3.0; Beckman Coulter, Inc.).

Apoptosis assay

At 48 h post-transfection, the LN229 and U251 cells were collected. The cells were washed twice with PBS. The cells were then prepared as single cell suspensions with binding buffer and stained using an Annexin-V-FITC/PI Apoptosis Detection kit (Abcam). The number of apoptotic cells was detected and quantified by a flow cytometer (EPICS XL; Beckman Coulter, Inc.) using SYSTEM II Software (version 3.0; Beckman Coulter, Inc.).

Lentiviral construction

HNF4G short hairpin RNA (shRNA) was incorporated into a lentiviral vector (GV118; Shanghai GeneChem Co., Ltd.) for the silencing of HNF4G expression. The sequences were as follows: Negative control (sh-Ctrl), 5′-AAAAGAGGCTTGCACAGTGCATTCAAGACGTGCACTGTGCAAGCCTCTTTT-3′; and HNF4G shRNA, 5′-TCGAGTGAGAGAAACACATTTTCTCGAGAATGTGTTTCTCTCACTCGTTTTTTC-3′. The U251 cells were cultured in 12-well plates. The solution of lentiviral vector (0.5 ml 4×108 TU/ml) was then used to infect the U251 cells with Polybrene 5 µg/ml (Shanghai GeneChem Co., Ltd.) for 10 h at 37°C, after which the culture medium was replaced with DMEM containing 10% fetal calf serum. At 2 days post-infection, puromycin (cat. no. P9620; 25 µg/ml; MilliporeSigma) was added in infected cells and the culture was continued for 6 days for the tumor transplantation experiment.

Tumorigenicity assay

Following infection with sh-Ctrl or HNF4G shRNA, the U251 cells were prepared as a single-cell suspension in DMEM. Tumorigenicity was detected using 6-week-old BALB/c nude mice (n=4/group). The infected U251 cells (2×106) were resuspended in 100 µl DMEM and injected subcutaneously into the posterior flanks of the mice. The transplanted tumors were measured using a Vernier caliper every third day. The length (L) and width (W) of the tumors were used to calculate the tumor volume (V) using the following formula: V=(L × W2)/2. The nude mice were euthanized by anesthesia with 3% isoflurane followed by the injection of pentobarbital sodium (150 mg/kg) 31 days after the injection of the cells. The cessation of breathing and heartbeat were considered as confirmation of death. The tumors were then isolated and weighed, after which the tumor tissues were frozen for use in further assays.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from the glioma samples, mouse tumor tissues and glioma cells using an RNA Extraction Kit (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. The RNA was reversed transcribed into cDNA using the PrimeScript II 1st strand cDNA kit (Takara Bio, Inc.) according to the manufacturer's protocol. qPCR was then performed using SYBR Premix Ex Taq (Takara Biotechnology Co., Ltd.). Reaction conditions were as follows: Initial denaturation at 95°C for 5 min, followed by denaturation at 95°C for 10 sec, annealing at 55°C for 25 sec and extension at 72°C for 10 sec, for 45 cycles. The primer sequences used were as follows: HNF4G forward, 5′-ACAGAATAAGCACCAGAAG-3′ and reverse, 5′-TCACAGACATCACCAATAC-3′; NRP1 forward, 5′-CGTGGAAGTCTTCGATGGAG-3′ and reverse, 5′-AAGAAATGGCCCTGAAGACA-3′; and GAPDH forward, 5′-GCCGTATCGCTCAGACAC-3′ and reverse, 5′-GCCTAATACGACCAAATCC-3′. The qPCR was performed using an IQ5 Multicolor qRT-PCR Detection System (Bio-Rad Laboratories, Inc.). The 2−ΔΔCq method (13) was used to analyze the expression of the target genes using GAPDH as the reference gene.

Western blot analysis

Protein was extracted from the glioma samples, mouse tumor tissue and glioma cells using RIPA buffer (MilliporeSigma) with protease inhibitors. Protein quantification was performed with a BCA kit (Beyotime Institute of Biotechnology) according to the manufacturer's protocol. Equal amounts of protein samples (30 µg) were subjected to sodium dodecyl sulfate-polyacrylamide (10%) gel electrophoresis followed by transfer onto nitrocellulose membranes. After blocking the membranes with skimmed milk (5%) at room temperature for 2 h, primary antibodies were added followed by incubation at 4°C for 10 h. The primary antibodies comprised HNF4G antibody (cat. no. HPA005438; 1:1,000; MilliporeSigma), NRP1 antibody (cat. no. sc-5307; 1:1,000; Santa Cruz Biotechnology, Inc.) and GAPDH antibody (cat. no. sc-47724; 1:1,000; Santa Cruz Biotechnology, Inc.). The appropriate anti-rabbit (cat. no. sc-2357) or anti-mouse (cat. no. sc-2005) horseradish peroxidase-linked secondary antibody (both 1:2,000; Santa Cruz Biotechnology, Inc.) was then added followed by incubation at room temperature for 4 h. The membranes were finally incubated with enhanced chemiluminescence reagent (GE Healthcare; Cytiva) for chemiluminescence detection. The protein bands were scanned using Syngene G:BOX Chemi XX6 system (Syngene) and quantified using GeneTools software (version 3.06.02; Syngene). GAPDH was used to normalize the protein expression data.

Chromatin immunoprecipitation (ChIP)-RT-qPCR

After crosslinking the LN229 and U251 cells with formaldehyde (1%) at room temperature for 18 min, glycine was added for quenching. The cells were resuspended in SDS lysis buffer [50 mM Tris-HCl (pH 8.1), 10 mM EDTA and 1% SDS with freshly added PIC at 1 µl PIC/800 µl total volume]. The LN229 and U251 cells were then sonicated using an ultrasonic processor and nuclear lysates were extracted, which contained chromatin that had been broken into ~200-bp DNA fragments. HNF4G or IgG (cat. nos. HPA005438 and I8765; both 1:100; MilliporeSigma) antibodies were incubated with the extracted DNA fragments for 13 h at 4°C. Dynabeads Protein A (200 µl; Thermo Fisher Scientific, Inc.) was added in 500 µl lysis buffer and agitated in a shaker for 2 h. The beads were centrifuged at 12,000 × g for 1 min at room temperature. The supernatant was removed and transfered to a new 1.5-ml microfuge tube. The DNA-protein complexes were washed in TE buffer at 65°C. Subsequently, the crosslinking was reversed for 8 h at 65°C. The QIAquick® PCR purification Kit (Qiagen GmbH) was used to extract the binding DNA fragments. The predicted binding NRP1 DNA fragments (UCSC Genome Browser; Human Feb. 2009, GRCh37/hg19) (14) were verified using RT-qPCR according to the aforementioned protocol. The primer sequences used were as follows: Primer 1 forward, 5′-GCATTGACTTAGCCAGAAGGTGACA-3′ and reverse, 5′-GCTTTCCTGTTTCTCCATTGTCTGA-3′; primer 2 forward, 5′-ATGACTCAGACAATGGAGAAACAGG-3′ and reverse, 5′-ACAAGTTCAATCCAAACCACGCGGG-3′; primer 3 forward, 5′-GTAGACCCGCGTGGTTTGGATTGAA-3′ and reverse, 5′-GCCAGTCGGTCCTGTCACAGAGTCT-3′; primer 4 forward, 5′-CAGTAGACTCTGTGACAGGACCGAC-3′ and reverse, 5′-ATTGTCAGAGCAGGAGCGGTTTTGT-3′; and primer 5 forward, 5′-TAACAAAACCGCTCCTGCTCTGACA-3′ and reverse, 5′-GTTAAAAAAAATAAAAGTGAACAAC-3′. The target areas of the primers are shown in Fig. S1.

Luciferase reporter gene assay

The LN229 and U251 cells were plated into 96-well plates. Each group was established in six parallel wells. The LN229 and U251 cells were transfected with pGL3-luc or pGL3-NRP1-luc, and co-transfected with pGL3-NRP1-luc and HNF4G siRNAs or HNF4G overexpression vector at 37°C for 2 days. The luciferase activity was detected using a Dual-Luciferase Reporter Assay System (Promega Corporation). Luciferase activity was normalized to Renilla luciferase activity.

The cancer genome atlas (TCGA) data analysis

To verify that HNF4G regulates NRP1, microarray data were collected from the public database, TCGA (http://gepia.cancer-pku.cn/detail.php?gene=HNF4G), and the correlation between HNF4G and NRP1 expression was analyzed by using simple linear regression.

Statistical analysis

All statistical data were analyzed using SPSS 25.0 software (IBM Corp.). All experiments were performed with at least three independent assays. Data are presented as the mean ± standard deviation. Unpaired Student's t-test or one-way ANOVA was performed to analyze the differences between two or multiple independent groups, respectively. The ANOVA test followed by the Bonferroni post hoc test was used for pairwise comparisons. Paired Student's t-test was used to analyze differences in HNF4G and NRP1 mRNA levels between glioma tissue and normal tissue. Fisher's exact test was used to analyze the association between HNF4G mRNA expression and clinicopathological features. Pearson's correlation analysis was conducted to analyze the correlation between HNF4G and NRP1 expression. P<0.05 was considered to indicate a statistically significant difference.

Results

HNF4G expression is upregulated in human glioma samples and cell lines

To explore the role of HNF4G in glioma, the expression of HNF4G in glioma specimens and cell lines was evaluated using RT-qPCR and western blot analysis. The results revealed that the mRNA and protein expression levels of HNF4G were significantly upregulated in human glioma samples compared with those in adjacent normal tissues (P<0.001; Fig. 1A and C). Moreover, the high mRNA expression of HNF4G was associated with the WHO pathological grade, isocitrate dehydrogenase [NADP(+)] 1 (IDH1) mutation, tumor size and the Karnofsky Performance Scale (KPS) score (all P<0.001; Table I). The HNF4G mRNA and protein expression levels were also significantly higher in the U87, LN229 and U251 human glioma cell lines than in the NHAs (P<0.001; Fig. 1B and D).

Table I.

Association between HNF4G mRNA expression and clinicopathological features in patients with glioma.

Table I.

Association between HNF4G mRNA expression and clinicopathological features in patients with glioma.

HNF4G mRNA expression

CharacteristicsPatients (n)High (n=49)Low (n=10)aP-value
Sex 0.907
  Male38317
  Female21183
Age, years 0.839
  ≥5033276
  <5026224
WHO grade <0.001
  I + II20128
  III + IV39372
IDH1 <0.001
  Mutation27 (Nod, 13; Cod, 16)252
  Wild-type32 (Nod, 28; Cod, 6)248
1p/19q 0.065
  No deletion38317
  Codeletion21183
Tumor size, cm <0.001
  ≥535323
  <524177
KPS score <0.001
  <8023176
  ≥8036324

a P-values calculated by Fisher's exact test. HNF4G, hepatocyte nuclear factor 4γ; WHO, World Health Organization; IDH1, isocitrate dehydrogenase [NADP(+)] 1; Nod, no deletion; Cod, codeletion; KPS, Karnofsky Performance Scale.

HNF4G promotes glioma cell proliferation in vitro and in vivo

To investigate the biological function of HNF4G in human glioma, LN229 and U251 cells were stably transfected with HNF4G overexpression plasmid, control empty plasmid, HNF4G siRNAs or NC-siRNA, independently. The HNF4G overexpression plasmid significantly upregulated the mRNA and protein expression levels of HNF4G in the LN229 and U251 cells; by contrast, the HNF4G siRNAs significantly downregulated the mRNA and protein expression levels of HNF4G (P<0.001; Fig. 2A and B). The results of MTT assays revealed that HNF4G overexpression significantly promoted glioma cell proliferation (P<0.001; Fig. 2C) after 48 and 72 h, whereas the silencing of HNF4G significantly suppressed glioma cell proliferation at these time points (P<0.001; Fig. 2D). Cell cycle analysis revealed that HNF4G overexpression led to a significant reduction in the number of cells in the G0/G1 phase and significant increases in the number of cells in the S and G2/M phases (P<0.001; Figs. 2E and S2A); by contrast, the silencing of HNF4G resulted in the significant accumulation of cells in the G0/G1 phase and a corresponding reduction in the number of cells in the S and G2/M phases (P<0.001; Figs. 2F and S2B). Moreover, analysis of cell apoptosis using flow cytometry revealed that HNF4G overexpression reduced the rates of early and late apoptosis (Figs. 2G and S2C), whereas the silencing of HNF4G enhanced the rate of early- and late-apoptotic cells to a significant extent (P<0.001; Figs. 2H and S2D). Subsequently, an HNF4G shRNA lentiviral vector was constructed and stably transfected U251 gliomas were generated. Nude mice were subcutaneously injected in the posterior flanks with HNF4G shRNA-infected or sh-Ctrl-infected U251 cells and tumor growth was monitored for 31 days. Observation of the excised tumors revealed that HNF4G shRNA markedly inhibited tumor growth compared with that in the sh-Ctrl group, in which the largest tumor diameter was 9 mm (Fig. 2I). The volumes and weights of the tumors derived from HNF4G shRNA-transfected cells were significantly lower than those derived from sh-Ctrl-transfected cells (P<0.001; Fig. 2J and K). The results also revealed that HNF4G shRNA significantly suppressed the mRNA and protein expression levels of HNF4G in the xenograft tumors (P<0.001; Fig. 2L and M). These results suggest that HNF4G facilitated glioma cell proliferation and tumor growth, and suppressed cell apoptosis.

HNF4G activates NRP1 transcription by binding to its promoter

To determine the mechanisms underlying the effect of HNF4G in the modulation of glioma development, the UCSC Genome Browser was used to predict and select the downstream target gene of HNF4G. The results indicated that HNF4G binds to the promoter region of the NRP1 gene (Fig. 3A). ChIP-RT-qPCR further confirmed that HNF4G bound to the promoter regions of NRP1 in LN229 and U251 cells, and primarily bound to the segments of primers 3–5 (P<0.001; Fig. 3B). Promoter reporter assays were used to verify whether HNF4G regulates NRP1 transcription by binding to its promoter. The binding sequence of NRP1 was inserted downstream of the luciferase gene. The LN22 and U251 cells were transfected with the constructed plasmids for 2 days and the luciferase activity was detected. The results revealed that the luciferase activity in the cells transfected with pGL3-NRP1-luc was significantly higher than that in the cells transfected with pGL3-luc (P<0.001; Fig. 3C). When the glioma cells were co-transfected with pGL3-NRP1-luc and the HNF4G overexpression vector, the luciferase activity was significantly enhanced compared with that in the control vector group (P<0.001; Fig. 3D). A significant reduction in luciferase activity was observed in the cells co-transfected with pGL3-NRP1-luc and HNF4G siRNA compared with that in the cells co-transfected with pGL3-NRP1-luc and NC-siRNA (P<0.001; Fig. 3E).

The analysis of patient tissues showed that the mRNA expression of NRP1 was also significantly upregulated in glioma samples compared with that in the adjacent normal tissues (P<0.001; Fig. 3F). In addition, a significant positive association between HNF4G and NRP1 mRNA expression was detected in the glioma tissues (P<0.001; Fig. 3G). TCGA data also revealed that HNF4G expression was positively associated with NRP1 expression in glioma (P<0.001; Fig. 3H). HNF4G overexpression significantly upregulated the mRNA and protein expression levels of NRP1 in LN229 and U251 cells, while HNF4G siRNA significantly downregulated them (P<0.001; Fig. 3I, K and L). Furthermore, HNF4G shRNA significantly suppressed the mRNA and protein expression levels of NRP1 in xenograft tumors (P<0.001; Fig. 3J and M). Together, these data demonstrate that HNF4G promoted NRP1 transcription by binding to its promoter in glioma cells.

NRP1 promotes glioma cell proliferation

To explore the role of NRP1 in human glioma, the LN229 and U251 cells were stably transfected with NRP1 overexpression plasmid, control empty plasmid, NRP1 siRNAs or NC-siRNA, independently. NRP1 overexpression significantly increased the mRNA and protein expression levels of NRP1 in the cells, whereas the knockdown of NRP1 significantly suppressed the mRNA and protein expression levels of NRP1 (P<0.001; Fig. 4A and B). The results of MTT assays revealed that NRP1 overexpression markedly promoted glioma cell proliferation after 48 and 72 h (P<0.001; Fig. 4C), while the knockdown of NRP1 substantially inhibited glioma cell growth at these time points (P<0.001; Fig. 4D). Flow cytometry revealed that NRP1 overexpression induced significant changes in the cell cycle, resulting in a marked significant reduction in the proportion of cells in the G0/G1 phase and an increase in the proportion of cells in the S and G2/M phases (P<0.001; Figs. 4E and S3A). However, the knockdown of NRP1 led to the significant accumulation of cells in the G0/G1 phase and a reduction in the proportion of cells in the S and G2/M phases (P<0.001; Figs. 4F and S3B). In addition, flow cytometry revealed that NRP1 overexpression significantly decreased the rates of early and late apoptosis (P<0.001; Figs. 4G and S3C), whereas the knockdown of NRP1 significantly increased the rates of early and late apoptosis (P<0.001; Figs. 4H and S3D). These results demonstrate that NRP1 had oncogenic effects.

HNF4G facilitates glioma cell growth by upregulating NRP1 expression

To validate the previous findings which indicated that HNF4G enhances glioma cell proliferation by modulating NRP1 transcription, HNF4G siRNA-2 and NRP1 overexpression vector were co-transfected into LN22 and U251 cells. The knockdown of HNF4G decreased the NRP1 mRNA and protein expression levels in LN229 and U251 cells, whereas co-transfection with the NRP1 overexpression vector reversed this effect (P<0.001; Fig. 5A and B). Furthermore, MTT assay results revealed that NRP1 overexpression reversed the effects of HNF4G knockdown on cell proliferation (P<0.001; Fig. 5C). Cell cycle analysis confirmed that HNF4G knockdown led to a significant accumulation of cells in the G0/G1 phase and a reduction in the proportion of cells in the S and G2/M phase, while co-transfection with NRP1 overexpression vector counteracted the effects of HNF4G knockdown on the cell cycle (P<0.001; Figs. 5D and S4A). Furthermore, apoptosis analysis showed that while the knockdown of HNF4G significantly increased the proportions of early- and late-apoptotic cells, co-transfection with NRP1 overexpression vector eliminated the effects of HNF4G siRNA-2 on apoptosis (P<0.001; Figs. 5E and S4B). These findings confirm that HNF4G promoted glioma cell proliferation and suppressed cell apoptosis by promoting NRP1 expression.

Discussion

As a major transcription factor, HNF4G is frequently upregulated in several types of cancer and functions as a pivotal oncogene (8,11). Previous studies have demonstrated that HNF4G facilitates the proliferation and invasion of bladder cancer cells (10,15). Wang et al (8) found that the overexpression of HNF4G promoted the progression and metastasis of pancreatic ductal adenocarcinoma. In addition, Shukla et al (9) observed that the HNF4G/HNF1A transcription loop accelerated prostate cancer cell proliferation and oncogenesis. Furthermore, Wang et al (11) demonstrated that the expression of HNF4G was significantly upregulated in lung cancer tissues, and that the overexpression of HNF4G facilitated lung cancer cell proliferation and tumorigenesis. In the present study, it was observed that the expression of HNF4G was upregulated in human glioma tissues and cell lines. The high mRNA expression of HNF4G was associated with the WHO pathological grade, IDH1 mutation, tumor size and KPS score. HNF4G promoted glioma cell proliferation in vitro, as well as cell cycle G1-S phase transition and tumor growth. Moreover, experiments in which HNF4G was overexpressed or knocked down revealed that HNF4G markedly suppressed apoptosis. The previous studies found that HNF4G facilitated the proliferation, invasion and metastasis in certain cancer types, and the present study revealed similar findings for glioma; specifically, HNF4G promoted glioma cell proliferation and cell cycle transition, and suppressed apoptosis. However, metastasis was not evaluated. These findings indicate that HNF4G functions as an oncogene in human glioma, and thus has potential for use as a novel therapeutic target for glioma.

HNF4G promotes bladder cancer progression by facilitating the transcription of hyaluronan synthase 2 (10). It has been reported that HNF4G and HNF1A activate enhancer chromatin and upregulate the expression of gastrointestinal transcriptome genes (9,16). In the present study, to investigate the potential molecular mechanism of HNF4G in glioma, NRP1 was predicted as a downstream target gene of HNF4G by bioinformatics analysis. ChIP-RT-qPCR and promoter reporter assays verified that HNF4G upregulated NRP1 transcription by binding to its promoter. Furthermore, HNF4G expression positively associated with NRP1 expression in glioma. The overexpression and silencing of HNF4G revealed that HNF4G promoted NRP1 expression in glioma. These findings confirm the prediction that HNF4G upregulates NRP1 expression by binding to its promoter region in glioma cells.

NRP1 is a transmembrane glycoprotein and a multi-functional co-receptor for several signaling pathways, such as hepatocyte growth factor (HGF), semaphorins, platelet-derived growth factor and vascular endothelial growth factor (VEGF) (1719). NRP1 has a pivotal role in embryonic angiogenesis and neurogenesis (20,21). Moreover, NRP1 can regulate cell mitogenesis, migration, motility, proliferation, survival and apoptosis by binding to VEGF and HGF (2226). There is evidence to indicate that NRP1 is involved in tumorigenesis and progression. For example, studies have shown that the upregulation of NRP1 promotes pancreatic cancer progression by modulating the HGF/c-Met pathway (27), VEGF-A/NRP1 signaling promotes breast cancer metastasis (28), and NRP1 facilitates oral squamous cell carcinoma cell growth and invasion by interacting with CMTM6 (29). In addition, the VEGFR-NRP1 axis has been demonstrated to promote the angiogenesis, growth and metastasis of gastric cancer (30). Furthermore, NRP1 has been shown to facilitate nasopharyngeal carcinoma cell migration and invasion (31). In glioma, previous studies have established that NRP1 promotes glioma cell proliferation, invasion, migration and tumor growth, and suppresses cell apoptosis (3234). In the present study, it was confirmed that NRP1 promoted glioma cell proliferation and inhibited cell apoptosis. Additionally, NRP1 overexpression reversed the effects induced by HNF4G knockdown on cell proliferation, cell cycle progression and apoptosis. These findings demonstrate that HNF4G promoted glioma cell proliferation and suppressed cell apoptosis by promoting NRP1 transcription.

In conclusion, HNF4G is considered as an oncogene in glioma. In the present study, the results demonstrated that HNF4G expression was upregulated in glioma. HNF4G facilitated cell proliferation and cell cycle progression, and inhibited apoptosis by promoting NRP1 transcription. These results indicate that HNF4G may be an effective therapeutic target for glioma.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

This study was supported by the Shaanxi Province Natural Science Foundation (grant nos. 2015JM8419 and 2021JM-582) and the Key Research and Development Program of Shaanxi (grant no. 2019SF-217).

Availability of data and materials

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

Authors' contributions

HC and LZ designed the experiments. HC, QZ and LZ conducted the experiments. ZL provided research materials and analyzed data. LZ, HC and LZ wrote the manuscript. All authors read and approved the final version of the manuscript. HC and LZ confirm the authenticity of all the raw data.

Ethics approval and consent to participate

The present study was approved by the Ethics Committee of Xi'an Gaoxin Hospital (approval no. GXYY-XA-2022-052). All patients provided written informed consent. The Institutional Animal Care and Use Committee of Xi'an Gaoxin Hospital approved the animal experiments (approval no. GXYY-XA-A-2022-039).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Chen J, Wu X, Xing Z, Ma C, Xiong W, Zhu X and He X: FOXG1 expression is elevated in glioma and inhibits glioma cell apoptosis. J Cancer. 9:778–783. 2018. View Article : Google Scholar : PubMed/NCBI

2 

Ostrom QT, Cote DJ, Ascha M, Kruchko C and Barnholtz-Sloan JS: Adult glioma incidence and survival by race or ethnicity in the United States from 2000 to 2014. JAMA Oncol. 4:1254–1262. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Komori T, Sasaki H and Yoshida K: Revised WHO classification of tumours of the central nervous system: Summary of the revision and perspective. No Shinkei Geka. 44:625–635. 2016.PubMed/NCBI

4 

Bertrand S, Brunet FG, Escriva H, Parmentier G, Laudet V and Robinson-Rechavi M: Evolutionary genomics of nuclear receptors: From twenty-five ancestral genes to derived endocrine systems. Mol Biol Evol. 21:1923–1937. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Drewes T, Senkel S, Holewa B and Ryffel GU: Human hepatocyte nuclear factor 4 isoforms are encoded by distinct and differentially expressed genes. Mol Cell Biol. 16:925–931. 1996. View Article : Google Scholar : PubMed/NCBI

6 

Baraille F, Ayari S, Carrière V, Osinski C, Garbin K, Blondeau B, Guillemain G, Serradas P, Rousset M, Lacasa M, et al: Glucose tolerance is improved in mice invalidated for the nuclear receptor HNF-4γ: A critical role for enteroendocrine cell lineage. Diabetes. 64:2744–2756. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Chen BD, Chen XC, Pan S, Yang YN, He CH, Liu F, Ma X, Gai MT and Ma YT: TT genotype of rs2941484 in the human HNF4G gene is associated with hyperuricemia in Chinese Han men. Oncotarget. 8:26918–26926. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Wang C, Zhang T, Liao Q, Dai M, Guo J, Yang X, Tan W, Lin D, Wu C and Zhao Y: Metformin inhibits pancreatic cancer metastasis caused by SMAD4 deficiency and consequent HNF4G upregulation. Protein Cell. 12:128–144. 2021. View Article : Google Scholar : PubMed/NCBI

9 

Shukla S, Cyrta J, Murphy DA, Walczak EG, Ran L, Agrawal P, Xie Y, Chen Y, Wang S, Zhan Y, et al: Aberrant activation of a gastrointestinal transcriptional circuit in prostate cancer mediates castration resistance. Cancer Cell. 32:792–806.e7. 2017. View Article : Google Scholar : PubMed/NCBI

10 

Okegawa T, Ushio K, Imai M, Morimoto M and Hara T: Orphan nuclear receptor HNF4G promotes bladder cancer growth and invasion through the regulation of the hyaluronan synthase 2 gene. Oncogenesis. 2:e582013. View Article : Google Scholar : PubMed/NCBI

11 

Wang J, Zhang J, Xu L, Zheng Y, Ling D and Yang Z: Expression of HNF4G and its potential functions in lung cancer. Oncotarget. 9:18018–18028. 2017. View Article : Google Scholar : PubMed/NCBI

12 

Wang Z, Ni F, Yu F, Cui Z, Zhu X and Chen J: Prognostic significance of mRNA expression of CASPs in gastric cancer. Oncol Lett. 18:4535–4554. 2019.PubMed/NCBI

13 

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 : PubMed/NCBI

14 

Kent WJ, Sugnet CW, Furey TS, Roskin KM, Pringle TH, Zahler AM and Haussler D: The human genome browser at UCSC. Genome Res. 12:996–1006. 2002. View Article : Google Scholar : PubMed/NCBI

15 

Sun H, Tian J, Xian W, Xie T and Yang X: miR-34a inhibits proliferation and invasion of bladder cancer cells by targeting orphan nuclear receptor HNF4G. Dis Markers. 2015:8792542015. View Article : Google Scholar : PubMed/NCBI

16 

Chen L, Toke NH, Luo S, Vasoya RP, Fullem RL, Parthasarathy A, Perekatt AO and Verzi MP: A reinforcing HNF4-SMAD4 feed-forward module stabilizes enterocyte identity. Nat Genet. 51:777–785. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Hong TM, Chen YL, Wu YY, Yuan A, Chao YC, Chung YC, Wu MH, Yang SC, Pan SH, Shih JY, et al: Targeting neuropilin 1 as an antitumor strategy in lung cancer. Clin Cancer Res. 13:4759–4768. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Karjalainen K, Jaalouk DE, Bueso-Ramos CE, Zurita AJ, Kuniyasu A, Eckhardt BL, Marini FC, Lichtiger B, O'Brien S, Kantarjian HM, et al: Targeting neuropilin-1 in human leukemia and lymphoma. Blood. 117:920–927. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Gelfand MV, Hagan N, Tata A, Oh WJ, Lacoste B, Kang KT, Kopycinska J, Bischoff J, Wang JH and Gu C: Neuropilin-1 functions as a VEGFR2 co-receptor to guide developmental angiogenesis independent of ligand binding. Elife. 3:e037202014. View Article : Google Scholar : PubMed/NCBI

20 

Gu C, Rodriguez ER, Reimert DV, Shu T, Fritzsch B, Richards LJ, Kolodkin AL and Ginty DD: Neuropilin-1 conveys semaphorin and VEGF signaling during neural and cardiovascular development. Dev Cell. 5:45–57. 2003. View Article : Google Scholar : PubMed/NCBI

21 

Bagri A and Tessier-Lavigne M: Neuropilins as semaphorin receptors: In vivo functions in neuronal cell migration and axon guidance. Adv Exp Med Biol. 515:13–31. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Pan Q, Chanthery Y, Liang WC, Stawicki S, Mak J, Rathore N, Tong RK, Kowalski J, Yee SF, Pacheco G, et al: Blocking neuropilin-1 function has an additive effect with anti-VEGF to inhibit tumor growth. Cancer Cell. 11:53–67. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Sulpice E, Plouët J, Bergé M, Allanic D, Tobelem G and Merkulova-Rainon T: Neuropilin-1 and neuropilin-2 act as coreceptors, potentiating proangiogenic activity. Blood. 111:2036–2045. 2008. View Article : Google Scholar : PubMed/NCBI

24 

Jia H, Cheng L, Tickner M, Bagherzadeh A, Selwood D and Zachary I: Neuropilin-1 antagonism in human carcinoma cells inhibits migration and enhances chemosensitivity. Brit J Cancer. 102:541–552. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Lee P, Goishi K, Davidson AJ, Mannix R, Zon L and Klagsbrun M: Neuropilin-1 is required for vascular development and is a mediator of VEGF-dependent angiogenesis in zebrafish. Proc Natl Acad Sci USA. 99:10470–10475. 2002. View Article : Google Scholar : PubMed/NCBI

26 

Raskopf E, Vogt A, Standop J, Sauerbruch T and Schmitz V: Inhibition of neuropilin-1 by RNA-interference and its angiostatic potential in the treatment of hepatocellular carcinoma. Z Gastroenterol. 48:21–27. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Matsushita A, Götze T and Korc M: Hepatocyte growth factor-mediated cell invasion in pancreatic cancer cells is dependent on neuropilin-1. Cancer Res. 67:10309–10316. 2007. View Article : Google Scholar : PubMed/NCBI

28 

Song Y, Zeng S, Zheng G, Chen D, Li P, Yang M, Luo K, Yin J, Gu Y, Zhang Z, et al: FOXO3a-driven miRNA signatures suppresses VEGF-A/NRP1 signaling and breast cancer metastasis. Oncogene. 40:777–790. 2021. View Article : Google Scholar : PubMed/NCBI

29 

Zheng Y, Wang C, Song A, Jiang F, Zhou J, Li G, Zhang W, Ye J, Ding X, Zhang W, et al: CMTM6 promotes cell proliferation and invasion in oral squamous cell carcinoma by interacting with NRP1. Am J Cancer Res. 10:1691–1709. 2020.PubMed/NCBI

30 

Mei B, Chen J, Yang N and Peng Y: The regulatory mechanism and biological significance of the Snail-miR590-VEGFR-NRP1 axis in the angiogenesis, growth and metastasis of gastric cancer. Cell Death Dis. 11:2412020. View Article : Google Scholar : PubMed/NCBI

31 

Huang Z, Cheng C, Xiong H, Wang Y, Chen KK, Yang J, Xiao B, Zhang R, Li S and Sang Y: NRP1 promotes cell migration and invasion and serves as a therapeutic target in nasopharyngeal carcinoma. Int J Clin Exp Pathol. 11:2460–2469. 2018.PubMed/NCBI

32 

Evans IM, Yamaji M, Britton G, Pellet-Many C, Lockie C, Zachary IC and Frankel P: Neuropilin-1 signaling through p130Cas tyrosine phosphorylation is essential for growth factor-dependent migration of glioma and endothelial cells. Mol Cell Biol. 31:1174–1185. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Higgins DMO, Caliva M, Schroeder M, Carlson B, Upadhyayula PS, Milligan BD, Cheshier SH, Weissman IL, Sarkaria JN, Meyer FB and Henley JR: Semaphorin 3A mediated brain tumor stem cell proliferation and invasion in EGFRviii mutant gliomas. BMC Cancer. 20:12132020. View Article : Google Scholar : PubMed/NCBI

34 

Hamerlik P, Lathia JD, Rasmussen R, Wu Q, Bartkova J, Lee M, Moudry P, Bartek J Jr, Fischer W, Lukas J, et al: Autocrine VEGF-VEGFR2-neuropilin-1 signaling promotes glioma stem-like cell viability and tumor growth. J Exp Med. 209:507–520. 2012. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2023
Volume 25 Issue 3

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
Che H, Zheng Q, Liao Z and Zhang L: HNF4G accelerates glioma progression by facilitating NRP1 transcription. Oncol Lett 25: 102, 2023
APA
Che, H., Zheng, Q., Liao, Z., & Zhang, L. (2023). HNF4G accelerates glioma progression by facilitating NRP1 transcription. Oncology Letters, 25, 102. https://doi.org/10.3892/ol.2023.13688
MLA
Che, H., Zheng, Q., Liao, Z., Zhang, L."HNF4G accelerates glioma progression by facilitating NRP1 transcription". Oncology Letters 25.3 (2023): 102.
Chicago
Che, H., Zheng, Q., Liao, Z., Zhang, L."HNF4G accelerates glioma progression by facilitating NRP1 transcription". Oncology Letters 25, no. 3 (2023): 102. https://doi.org/10.3892/ol.2023.13688