Open Access

Panoramic RNA expression of fibroblast growth factors in human glioblastoma tissues and the impact on the survival of patients

  • Authors:
    • Kun Zhao
    • Jiakun Xu
    • Beichuan Zhao
  • View Affiliations

  • Published online on: May 14, 2024     https://doi.org/10.3892/ol.2024.14450
  • Article Number: 317
  • Copyright: © Zhao 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

Fibroblast growth factors (FGFs) have a key role in various critical steps of tumor growth and progression through effects on angiogenesis, inflammation and the growth and invasion of malignant cells. Nevertheless, the role of the FGF family in human glioblastoma (GBM) has been rarely studied. The objective of the present study was to assess the RNA expression of all FGF family members in tissues obtained from patients with GBM and to analyze the association between FGF expression and the survival of these patients. For this, the RNA expression of FGF family members in the malignant and proximal tissues of 12 patients with GBM was determined by analyzing high‑throughput RNA transcriptome sequencing data uploaded to the National Center for Biotechnology Information database. The relationship between FGF genes and the survival of patients with GBM and glioma was also respectively studied by analyzing data from The Cancer Genome Atlas database using the Gene Expression Profiling Interactive Analysis tool. The results showed that the expression of FGF1, FGF17, FGF20 and FGF22 in GBM tissues was lower than that in adjacent tissues, with a difference of >2 times. Analysis of the overall survival of patients with GBM indicated there were no significant relationships between the expression of FGF1, FGF17, FGF20, FGF22 and overall survival. Analysis of the overall survival of patients with glioma showed that glioma patients with low FGF1 expression achieved a longer survival time than patients with high FGF1 expression; however, high expression of FGF17 and FGF22 indicated a longer survival time. In summary, the results of the present study demonstrated the panoramic expression of FGF family members in patients with GBM, and indicated that FGF1, FGF17 and FGF22 did not affect the survival of patients with GBM, but had a notable influence on the survival of patients with glioma.

Introduction

Human gliomas are the most common and malignant primary tumors of the central nervous system (CNS). Despite the development of surgery, chemotherapy and radiation therapy, the median survival time of patients with this disease remains poor (1). Glioblastoma (GBM), the most lethal form of glioma, has a median overall survival time of only 15 months, which has remained unchanged over several years (2). In addition to the rapid proliferation, extensive invasion, tumor genetic heterogeneity and treatment resistance of GBM, the poor prognosis of patients also results from an insufficient understanding of the molecular pathogenesis and the lack of markers for timely diagnosis and sensitive treatments (2,3). Therefore, there is an urgent need to explore the reliable biomarkers of GBM progression to improve the treatment of this malignancy.

Fibroblast growth factors (FGFs) are broad-spectrum mitogens that regulate a number of cellular functions, including migration, proliferation, differentiation and survival (4). FGFs have been studied since the 1960s with regards to their function in fibroblast driven growth (5), and were formally purified and characterized nearly a decade after first identification (6). In mammals, FGFs exert their pleiotropic functions through binding and activating high-affinity tyrosine kinase receptors such as the fibroblast growth factor receptor (FGFR), which is encoded by four genes (FGFR1, FGFR2, FGFR3 and FGFR4) and FGFRL (a truncated FGFR without an intracellular domain) (4). The FGF/FGFR signaling system regulates a number of biological processes, such as embryogenesis, wound repair, tissue homeostasis and angiogenesis (7). Dysfunction of FGF/FGFR signaling has been observed in a variety of human diseases, such as chronic kidney disease, congenital craniosynostosis, dwarfism syndromes, obesity, insulin resistance and cancer. A number of studies have demonstrated that activation of the FGF/FGFR signaling system plays a key role in a variety of critical steps in tumor growth and progression through its effects on tumor and stromal cells, affecting angiogenesis, inflammation and tumor growth (8,9).

FGFs are highly associated with the development and progression of various human malignancies, including urothelial cancer, multiple myeloma, prostate cancer and hepatocellular carcinoma (4). Considering the importance of FGF signaling, it has attracted considerable interest in the study of multiple malignancies, where it plays a role in the proliferation, survival, self-renewal and invasion of tumor cells (10). However, the role of FGF family members in GBM has been rarely explored, and most studies examining the role of FGFs were conducted in vitro. It has been reported that FGF2 is a oncogenic factor in glioma (11) and contributes to proliferation (12), the vascularization of glioma cells (13,14) and cancer stem cell self-renewal (15). FGF9 has been shown to potentially regulate the proliferation of human glioma cells either in the presence or absence of endogenous growth factor expression (16). Continued efforts to understand the correlation of FGFs with human GBM tissues will play a significant role in driving the discovery of novel diagnostic approaches and new therapies.

In the present study, the RNA expression of FGF members in human GBM tissues was first investigated by analyzing high-throughput RNA transcriptome sequencing data, then the effect of different FGF members on the overall survival of patients was further clarified. The present study therefore aimed to provide potential targets for diagnosis methods and the treatment of this malignancy.

Materials and methods

Human GBM and adjacent non-cancerous brain tissues (ANCBTs)

Eligible patients with newly diagnosed and histologically confirmed GBM [World Health Organization (WHO) grade IV astrocytoma] (17), and who had undergone maximal safe surgery were included in the present study. Patients with GBM who had undergone radiotherapy, chemotherapy, immunotherapy and other new therapies were excluded from the present study. Human cancerous tissues and ANCBTs were collected from 12 patients with GBM admitted to the Department of Neurosurgery at The First Affiliated Hospital of Sun Yat-sen University (Guangzhou, China) between December, 2016 and November, 2017, with written patient consent. Molecular and immunohistochemical biomarkers used for the diagnosis of these 12 patients with GBM included isocitrate dehydrogenase 1, glial fibrillary acidic protein, vimentin, p53, Ki-67 and O-6-methylguanine-DNA methyltransferase, information of which was obtained from the medical records from The Department Pathology at The First Affiliated Hospital of Sun Yat-sen University. Written patient consent was also obtained for the release of potentially personally-identifying clinical information. The present study was approved by the Institutional Ethics Review Board of The First Affiliated Hospital of Sun Yat-sen University [approval no. (2016)279] and complied with all relevant ethical regulations regarding human participants.

Analyzing high-throughput RNA transcriptome sequencing data

Previously, our team conducted high-throughput RNA transcriptome sequencing on tumors and adjacent tissues of the aforementioned 12 patients with GBM. The high-throughput RNA transcriptome sequencing was conducted and the procedure was clearly described in our previous study (18). The raw high-throughput RNA sequencing data was uploaded to the National Center for Biotechnology Information database with the (accession no. PRJNA525736). In the present study, the expression of FGF family members in GBM and proximal tissues was analyzed using this sequencing data. Data were mapped to the reference genome using the software TopHat2 (v.2.1.1), and then transcript abundance was quantified using RSEM (v.1.2.19).

Overall survival analysis

To further investigate the relationship between the expression of FGF family members and the overall survival of patients with GBM and glioma respectively, the ‘Survival Analysis’ tool (version 1.0) on Gene Expression Profiling Interactive Analysis (GEPIA; http://gepia.cancer-pku.cn/index.html) was used. The GBM and glioma datasets were respectively selected, and FGF1, FGF17, FGF20 and FGF22 (distinguishing high and low expression according to the median expression) were selected as the genes of interest. GEPIA datasets were originally from TCGA database. GEPIA used the log-rank test (sometimes termed the Mantel-Cox test) for the hypothesis evaluation, and P<0.05 was considered to indicate a statistically significant difference. The Cox proportional hazard ratio and the 95% confidence interval information were also included in the survival plot (19).

Statistical analysis

The data were analyzed using paired Student's t-test or χ2 test. Statistical analysis was performed using GraphPad Prism 8.0 (Dotmatics) and SPSS (version 25.0; IBM Corp.) statistical software. P<0.05 was considered to indicate a statistically significant difference.

Results

FGF1, FGF17, FGF20 and FGF22 were differentially expressed in human GBMs and ANCBTs at the RNA level

Human cancerous GBM tissues and ANCBTs were collected from 12 patients. The median age of these patients was 45.5 years (range, 29–59 years), and there were 7 (58.3%) male and 5 (41.7%) female patients. All patients were diagnosed with GBM (WHO grade IV astrocytoma) and underwent maximal surgical resection. The clinicopathological characteristics of the patients with GBM are shown in Table I.

Table I.

Clinicopathological characteristics of the patients with glioblastoma included in the present study.

Table I.

Clinicopathological characteristics of the patients with glioblastoma included in the present study.

Patient no.Age, yearsSexStageTherapyIDH-1GFAPVimentinp53Ki-67, %MGMT
159MaleIVResection-++++, 20+
256FemaleIVResection-++++, 40-
329MaleIVResection-++++, 60+
436FemaleIVResection-++++, 60+
554MaleIVResection-++++, 50+
645MaleIVResection-++++, 30+
744MaleIVResection-++++, 60+
859FemaleIVResection-++++, 20-
952MaleIVResection-++-+, 80+
1046MaleIVResection-++++, 70+
1129FemaleIVResection-++++, 50+
1237FemaleIVResection-++++, 30-

[i] GFAP, glial fibrillary acidic protein; IDH-1, isocitrate dehydrogenase 1; MGMT, O-6-methylguanine-DNA methyltransferase.

A panoramic visualization analysis was performed on the RNA expression of all members of the FGF family in human GBM tissues and ANCBTs (Table II). The result showed that the expression levels of four members, FGF1, FGF17, FGF20 and FGF22, were significantly lower in GBM tissues compared with the adjacent tissues, with a difference of >2 times (P<0.05; Fig. 1 and Table II). However, the expression of other FGF members in human GBM tissues and ANCBTs did not achieve both a difference of >2 times and statistical significance (Fig. 2 and Table II). To the best of our knowledge, these finding are the first to show the whole expression level of FGF family members in human GBM tissues and ANCBTs.

Table II.

Expression of the FGF family members in the high-throughput RNA sequencing data obtained from 12 pairs of human GBM tissues and ANCBTs.

Table II.

Expression of the FGF family members in the high-throughput RNA sequencing data obtained from 12 pairs of human GBM tissues and ANCBTs.

IDSymbolGBM, average FPKMANCBT, average FPKMGBM/ANCBT, fold changeP-value
ENSG00000113578FGF149.8383115.15332.31050.0003
ENSG00000078579FGF200.33500.95672.85570.0019
ENSG00000070388FGF220.13920.40672.92220.0047
ENSG00000158815FGF171.06922.18832.04680.0137
ENSG00000161958FGF1117.460010.52250.60270.0082
ENSG00000156427FGF180.07580.14251.87910.0302
ENSG00000129682FGF135.287510.12831.91550.0462
ENSG00000070193FGF100.15000.29171.94440.0567
ENSG00000118972FGF230.00330.00000.00000.1039
ENSG00000140285FGF70.41500.55001.32530.1260
ENSG00000138675FGF50.24750.43421.75420.1942
ENSG00000111241FGF60.00000.01080.0108/00.1990
ENSG00000075388FGF40.00000.00080.0008/00.3388
ENSG00000186895FGF30.01330.00000.00000.3388
ENSG00000196468FGF160.02080.03581.72000.4303
ENSG00000162344FGF190.00250.00080.33330.4382
ENSG00000102678FGF91.26001.45831.15740.6505
ENSG00000102466FGF1410.30179.44830.91720.7533
ENSG00000114279FGF1225.526727.54921.07920.7617
ENSG00000138685FGF210.884210.69170.98230.8232
ENSG00000105550FGF210.00670.00751.12500.9089
ENSG00000107831FGF80.10170.10171.00001.0000

[i] ANCBT, adjacent non-cancerous brain tissue; FGF, fibroblast growth factor; FPKM, fragments per kilobase of transcript per million mapped reads.

FGF1, FGF17 and FGF22 expression had a notable influence on the survival of patients with glioma

The impact of the expression of the aforementioned four FGF genes (FGF1, FGF17, FGF20 and FGF22) on the survival of patients with GBM and glioma was respectively analyzed using data from The Cancer Genome Atlas (TCGA) database. In TCGA dataset, the high FGF groups represented patients with ≥50% FGF expression compared with all patients, and the low FGF groups represented patients with <50% FGF expression compared with all patients. The expression levels of FGF1 (P=0.24), FGF17 (P=0.82), FGF20 (P=0.49) and FGF20 (P=0.87) were not significantly correlated with the overall survival of the patients with GBM (Fig. 3). The relationship between these aforementioned FGFs and the overall survival of patients with glioma was then investigated. It was found that patients with glioma with low FGF1 expression achieved a longer survival time than patients with high FGF1 expression. Conversely, high expression of FGF17 and FGF22 indicated a longer survival time. In particular, the expression of FGF17 was most closely related to the survival of patients with glioma (P<0.001) (Fig. 4).

Discussion

The FGF family contains 22 members, which are typically divided into seven subfamilies based on evolutionary relationships, sequence similarity and biochemical functions, including the FGF1 (FGF1 and FGF2), FGF4 (FGF4, FGF5 and FGF6), FGF7 (FGF3, FGF7, FGF10 and FGF22), FGF8 (FGF8, FGF17 and FGF18), FGF9 (FGF9, FGF16 and FGF20), FGF19 (FGF19, FGF21 and FGF23) and FGF11 (FGF11, FGF12, FGF13 and FGF14) subfamilies (4,20). The results of the present study showed that the FGF1, FGF20, FGF22 and FGF17 expression levels were lower in GBM tissues compared with adjacent tissues, with a >2 times difference.

Among the FGF family members, FGF2 is one of the best characterized FGFs present in the CNS, and plays an important role in astrocyte proliferation, migration and maturation (2123). FGF2 also promotes glioma cell growth, vascularization and cancer stem cell self-renewal (10). Experimental studies using FGF2-specific antisense oligonucleotides or antibodies to block glioma cell proliferation (24) and angiogenesis (25) have highlighted the relevance of the therapeutic targeting of FGF2 to increase survival time in glioma animal models. However, the expression of FGF2 in patients with GBM has not been focused upon. In the present study, a significant difference in FGF2 expression was not observed between tumor and adjacent tissues in patients with GBM. The expression of FGF2 was not correlated with the overall survival of patients with GBM.

FGF1 (also termed acidic FGF) is an autocrine/paracrine hormone that has historically been considered as a mitogen and has also attracted attention as an antidiabetic agent (26,27). In addition, FGF1 also promotes the repair process of damaged vessels (28). Thus, FGF1 could promote proliferation and resistance to chemotherapy in human pancreatic cancer cell lines (29). The characteristic ability of FGF1 promoting tumor cell proliferation and chemotherapy resistance may explain the finding that glioma patients with low FGF1 expression had longer overall survival times than patients with high FGF1 expression. However, the specific impact of FGF1 on glioma cells still needs further study. High expression of FGF1 has been found in non-small cell lung, ovarian, breast and prostate malignancies (3032). The results of the present study showed that FGF1 expression was 2 times lower in GBM tissues compared with adjacent tissues, which was inconsistent with a previous result indicating that FGF1 was upregulated in human glioma tissues and cell lines (33). Thus, further studies are needed to compare the expression levels of FGF1 in human GBM tissues and ANCBTs. The present study found that GBM patients with high FGF1 expression (n=81) did not achieve a significantly different survival time compared with GBM patients with low FGF1 expression (n=81). However, glioma patients with low FGF1 expression (n=338) had longer overall survival times compared with glioma patients with high FGF1 expression (n=338). The paradox with regard to the different actions of FGF1 expression on GBM and glioma maybe due to the relatively small number of patients with GBM that were included.

FGF17, a member of the FGF8/17/18 subfamily, contains an N-terminal cleaved signal peptide and can activate IIIc splice variants of FGFR 1–3 and FGFR4 (20). FGF17 is commonly expressed in the brain, endometrium, adrenal glands, thyroid and spleen (34). A recent study showed that FGF17 can induce the proliferation of oligodendrocyte progenitor cells in the brain, thereby improving memory in mice (35). It has also been shown that FGF17 is highly expressed in prostate cancer and aberrantly expressed in acute leukemia (34,36). However, to the best of our knowledge, there have been no reports considering the effect of FGF17 expression on overall survival in GBM. In the present study, the results of the RNA sequencing analysis showed that FGF17 expression was >2 times higher in adjacent tissues compared with tumor tissues in patients with GBM. In addition, the results of TCGA data analysis demonstrated that glioma patients with high FGF17 expression (n=337) achieved a longer survival time compared with glioma patients with low FGF17 expression (n=337). However, GBM patients with high FGF17 expression (n=81) did not achieve a significantly different survival time compared with GBM patients with low FGF17 expression (n=80). This is maybe due to small number of patients with GBM that were included, and further studies including greater numbers may change the association between the expression of FGF17 and the overall survival of patients with GBM.

FGF20 is a paracrine growth factor, the orthologs of which are highly conserved among vertebrates (37). FGF20 is predominantly expressed in the brain (38) and has being suggested to be a vital factor involved in brain development and neuronal homeostasis (39). FGF20 could enhance the survival of dopaminergic neurons in Parkinson's disease, and this means that FGF20 may have a neuroprotective function in the brain (40). A previous study found that FGF20 expression was upregulated in glioma cells following treatment with glucocorticoids (GCs), while a reduction in FGF20 expression in glioma cells significantly blocked the effect of GCs on macrophage polarization (41). However, to the best of our knowledge, there has been no research examining FGF20 expression in patients with GBM and it remains to be determined whether FGF20 expression is associated with the prognosis of these patients. In the present study, the results showed that FGF20 expression in human GBM tissues was nearly 3-fold lower than that in ANCBTs. However, the results from TCGA database analysis did not identify a relationship between the FGF20 expression level and the overall survival of patients with GBM and glioma, respectively.

FGF22, a target-derived presynaptic organizer critical in the formation of novel excitatory synapses during development and the organization of synapses in adult brains (42,43), has become a crucial endogenous contributor of detour circuit formation (44). Previous studies have shown that FGF22 plays a critical role as a presynaptic organizer in the formation of new synapses in adult remodeling spinal cords (45) and that delivery of FGF22 may promote organization of the presynapse, promoting the plasticity of healthy supraspinal axons and thereby contributing to the recovery of function in incomplete spinal cord injury (44). However, there have been no studies examining the relationship between FGF22 and GBM and whether FGF22 expression affects the survival of patients with GBM. In the present study it was shown that the FGF22 expression level in adjacent tissues was nearly three times higher than that in GBM tissues. In addition, following analysis of TCGA data, it was found that GBM patients with high FGF22 expression (n=79) did not achieve a significantly different survival time compared with GBM patients with low FGF22 expression (n=78). However, it was found that glioma patients with high FGF22 expression (n=337) had a longer survival time than glioma patients with low FGF22 expression (n=333).

In summary, in the present study, the RNA expression levels of FGF family members in human GBM tissues were first investigated by analyzing high-throughput RNA transcriptome sequencing data. It was found that the expression levels of four members, FGF1, FGF20, FGF22 and FGF17, were >2 times lower in GBM tissues compared with adjacent tissues. The different expression levels of FGF1, FGF17, FGF20 and FGF22 did not have a significant influence on the overall survival of the patients with GBM. However, it was identified that patients with glioma with low FGF1 expression achieved longer overall survival time than patients with high FGF1 expression. By contrast, high expression of FGF17 and FGF22 indicated a longer overall survival time. The expression level of FGF17 was most closely related to the longer overall survival time of patients with glioma. The present study is therefore expected to provide a certain research basis for clinical gene therapy of these malignancies.

However, there are some limitations to the present study. The present study mainly focused on the RNA levels of FGFs in patients with GBM through analyzing RNA transcriptome sequencing data, and the FGF protein levels were therefore not detected in the present study. The protein levels of FGFs in human GBM will be examined in a further study. In addition, further research is still needed to explore whether and how FGF1, FGF17, FGF20 and FGF22 affect the proliferation or invasion abilities of human GBM. Moreover, the overall survival curves were obtained through the GEPIA analysis tool, which generates survival curves based on data from TCGA database. In addition, since whole datasets could not be obtained from all research within TCGA and as FGF expression was only tested in 12 patients in The First Affiliated Hospital of Sun Yat-sen University, survival outcomes in this study could only be obtained via the GEPIA analysis tool, which may have led to a bias in the overall survival data analysis.

Acknowledgements

Not applicable.

Funding

The present study was supported by The National Natural Science Foundation Youth Project of China (grant no. 82103254).

Availability of data and materials

The data generated in the present study are included in the figures and/or tables of this article.

Author's contributions

KZ designed the study, analyzed the sequencing data and wrote the manuscript. JX and BZ made contributions to the analysis and interpretation of data. KZ and JX confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Human GBM tissues and adjacent non-cancerous brain tissues were collected from the Neurosurgery Department of the First Affiliated Hospital of Sun Yat-sen University (Guangzhou, China) with written patient consent. The present study was approved by The Institutional Ethics Review Board of the First Affiliated Hospital of Sun Yat-sen University [approval no. (2016)279] and complied with all relevant ethical regulations regarding human participants.

Patient consent for publication

Written patient consent was obtained for the publication of potentially personally-identifying clinical information.

Competing interests

The authors declare that they have no competing interests.

References

1 

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

2 

Cheng J, Meng J, Zhu L and Peng Y: Exosomal noncoding RNAs in Glioma: Biological functions and potential clinical applications. Mol Cancer. 19:662020. View Article : Google Scholar : PubMed/NCBI

3 

Alifieris C and Trafalis DT: Glioblastoma multiforme: Pathogenesis and treatment. Pharmacol Ther. 152:63–82. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Xie Y, Su N, Yang J, Tan Q, Huang S, Jin M, Ni Z, Zhang B, Zhang D, Luo F, et al: FGF/FGFR signaling in health and disease. Signal Transduct Target Ther. 5:1812020. View Article : Google Scholar : PubMed/NCBI

5 

Foley JF, Kennedy BJ and Ross JD: A factor from HeLa cells promoting colonial growth of human fibroblast-like cells in culture. Cancer Res. 23:368–371. 1963.PubMed/NCBI

6 

Gospodarowicz D: Localisation of a fibroblast growth factor and its effect alone and with hydrocortisone on 3T3 cell growth. Nature. 249:123–127. 1974. View Article : Google Scholar : PubMed/NCBI

7 

Presta M, Chiodelli P, Giacomini A, Rusnati M and Ronca R: Fibroblast growth factors (FGFs) in cancer: FGF traps as a new therapeutic approach. Pharmacol Ther. 179:171–187. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Giacomini A, Chiodelli P, Matarazzo S, Rusnati M, Presta M and Ronca R: Blocking the FGF/FGFR system as a ‘two-compartment’ antiangiogenic/antitumor approach in cancer therapy. Pharmacol Res. 107:172–185. 2016. View Article : Google Scholar : PubMed/NCBI

9 

Presta M, Dell'Era P, Mitola S, Moroni E, Ronca R and Rusnati M: Fibroblast growth factor/fibroblast growth factor receptor system in angiogenesis. Cytokine Growth Factor Rev. 16:159–178. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Jimenez-Pascual A, Mitchell K, Siebzehnrubl FA and Lathia JD: FGF2: A novel druggable target for glioblastoma? Expert Opin Ther Targets. 24:311–318. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Haley EM and Kim Y: The role of basic fibroblast growth factor in glioblastoma multiforme and glioblastoma stem cells and in their in vitro culture. Cancer Lett. 346:1–5. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Bian XW, Du LL, Shi JQ, Cheng YS and Liu FX: Correlation of bFGF, FGFR-1 and VEGF expression with vascularity and malignancy of human astrocytomas. Anal Quant Cytol Histol. 22:267–274. 2000.PubMed/NCBI

13 

Joy A, Moffett J, Neary K, Mordechai E, Stachowiak EK, Coons S, Rankin-Shapiro J, Florkiewicz RZ and Stachowiak MK: Nuclear accumulation of FGF-2 is associated with proliferation of human astrocytes and glioma cells. Oncogene. 14:171–183. 1997. View Article : Google Scholar : PubMed/NCBI

14 

Toyoda K, Tanaka K, Nakagawa S, Thuy DH, Ujifuku K, Kamada K, Hayashi K, Matsuo T, Nagata I and Niwa M: Initial contact of glioblastoma cells with existing normal brain endothelial cells strengthen the barrier function via fibroblast growth factor 2 secretion: A new in vitro blood-brain barrier model. Cell Mol Neurobiol. 33:489–501. 2013. View Article : Google Scholar : PubMed/NCBI

15 

Pollard SM, Yoshikawa K, Clarke ID, Danovi D, Stricker S, Russell R, Bayani J, Head R, Lee M, Bernstein M, et al: Glioma stem cell lines expanded in adherent culture have tumor-specific phenotypes and are suitable for chemical and genetic screens. Cell Stem Cell. 4:568–580. 2009. View Article : Google Scholar : PubMed/NCBI

16 

Miyagi N, Kato S, Terasaki M, Shigemori M and Morimatsu M: Fibroblast growth factor-2 and −9 regulate proliferation and production of matrix metalloproteinases in human gliomas. Int J Oncol. 12:1085–1090. 1998.PubMed/NCBI

17 

Louis DN, Perry A, Reifenberger G, von Deimling A, Figarella-Branger D, Cavenee WK, Ohgaki H, Wiestler OD, Kleihues P and Ellison DW: The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A summary. Acta Neuropathol. 131:803–820. 2016. View Article : Google Scholar : PubMed/NCBI

18 

Gao X, Xia X, Li F, Zhang M, Zhou H, Wu X, Zhong J, Zhao Z, Zhao K, Liu D, et al: Circular RNA-encoded oncogenic E-cadherin variant promotes glioblastoma tumorigenicity through activation of EGFR-STAT3 signalling. Nat Cell Biol. 23:278–291. 2021. View Article : Google Scholar : PubMed/NCBI

19 

Tang Z, Li C, Kang B, Gao G, Li C and Zhang Z: GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 45((W1)): W98–W102. 2017. View Article : Google Scholar : PubMed/NCBI

20 

Ornitz DM and Itoh N: The Fibroblast Growth Factor signaling pathway. Wiley Interdiscip Rev Dev Biol. 4:215–266. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Flanders KC, Lüdecke G, Renzing J, Hamm C, Cissel DS and Unsicker K: Effects of TGF-betas and bFGF on astroglial cell growth and gene expression in vitro. Mol Cell Neurosci. 4:406–417. 1993. View Article : Google Scholar : PubMed/NCBI

22 

Hou YJ, Yu AC, Garcia JM, Aotaki-Keen A, Lee YL, Eng LF, Hjelmeland LJ and Menon VK: Astrogliosis in culture. IV. Effects of basic fibroblast growth factor. J Neurosci Res. 40:359–370. 1995. View Article : Google Scholar : PubMed/NCBI

23 

Reuss B, Dono R and Unsicker K: Functions of fibroblast growth factor (FGF)-2 and FGF-5 in astroglial differentiation and blood-brain barrier permeability: Evidence from mouse mutants. J Neurosci. 23:6404–6412. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Loilome W, Joshi AD, ap Rhys CM, Piccirillo S, Vescovi AL, Gallia GL and Riggins GJ: Glioblastoma cell growth is suppressed by disruption of Fibroblast Growth Factor pathway signaling. J Neurooncol. 94:359–366. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Stan AC, Nemati MN, Pietsch T, Walter GF and Dietz H: In vivo inhibition of angiogenesis and growth of the human U-87 malignant glial tumor by treatment with an antibody against basic fibroblast growth factor. J Neurosurg. 82:1044–1052. 1995. View Article : Google Scholar : PubMed/NCBI

26 

Gasser E, Sancar G, Downes M and Evans RM: Metabolic Messengers: Fibroblast growth factor 1. Nat Metab. 4:663–671. 2022. View Article : Google Scholar : PubMed/NCBI

27 

Sancar G, Liu S, Gasser E, Alvarez JG, Moutos C, Kim K, van Zutphen T, Wang Y, Huddy TF, Ross B and Dai Y: FGF1 and insulin control lipolysis by convergent pathways. Cell Metab. 34:171–183.e6. 2022. View Article : Google Scholar : PubMed/NCBI

28 

Uriel S, Brey EM and Greisler HP: Sustained low levels of fibroblast growth factor-1 promote persistent microvascular network formation. Am J Surg. 192:604–609. 2006. View Article : Google Scholar : PubMed/NCBI

29 

El-Hariry I, Pignatelli M and Lemoine NR: FGF-1 and FGF-2 regulate the expression of E-cadherin and catenins in pancreatic adenocarcinoma. Int J Cancer. 94:652–661. 2001. View Article : Google Scholar : PubMed/NCBI

30 

Zhao D, Lu Y, Yang C, Zhou X and Xu Z: Activation of FGF receptor signaling promotes invasion of non-small-cell lung cancer. Tumour Biol. 36:3637–3642. 2015. View Article : Google Scholar : PubMed/NCBI

31 

King ML, Lindberg ME, Stodden GR, Okuda H, Ebers SD, Johnson A, Montag A, Lengyel E, MacLean Ii JA and Hayashi K: WNT7A/β-catenin signaling induces FGF1 and influences sensitivity to niclosamide in ovarian cancer. Oncogene. 34:3452–3462. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Zubor P, Hatok J, Moricova P, Kajo K, Kapustova I, Mendelova A, Racay P and Danko J: Gene expression abnormalities in histologically normal breast epithelium from patients with luminal type of breast cancer. Mol Biol Rep. 42:977–988. 2015. View Article : Google Scholar : PubMed/NCBI

33 

Ke J, Yao YL, Zheng J, Wang P, Liu YH, Ma J, Li Z, Liu XB, Li ZQ, Wang ZH and Xue YX: Knockdown of long non-coding RNA HOTAIR inhibits malignant biological behaviors of human glioma cells via modulation of miR-326. Oncotarget. 6:21934–2149. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Katoh M and Katoh M: Comparative genomics on FGF8, FGF17, and FGF18 orthologs. Int J Mol Med. 16:493–496. 2005.PubMed/NCBI

35 

Iram T, Kern F, Kaur A, Myneni S, Morningstar AR, Shin H, Garcia MA, Yerra L, Palovics R, Yang AC, et al: Young CSF restores oligodendrogenesis and memory in aged mice via Fgf17. Nature. 605:509–515. 2022. View Article : Google Scholar : PubMed/NCBI

36 

Heer R, Douglas D, Mathers ME, Robson CN and Leung HY: Fibroblast growth factor 17 is over-expressed in human prostate cancer. J Pathol. 204:578–586. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Chen J, Wang X, Hu J, Du J, Dordoe C, Zhou Q, Huang W, Guo R, Han F, Guo K, et al: FGF20 Protected Against BBB disruption after traumatic brain injury by upregulating junction protein expression and inhibiting the inflammatory response. Front Pharmacol. 11:5906692021. View Article : Google Scholar : PubMed/NCBI

38 

Boshoff EL, Fletcher EJR and Duty S: Fibroblast growth factor 20 is protective towards dopaminergic neurons in vivo in a paracrine manner. Neuropharmacology. 137:156–163. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Dono R: Fibroblast growth factors as regulators of central nervous system development and function. Am J Physiol Regul Integr Comp Physiol. 284:R867–R881. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Niu J, Xie J, Guo K, Zhang X, Xia F, Zhao X, Song L, Zhuge D, Li X, Zhao Y and Huang Z: Efficient treatment of Parkinson's disease using ultrasonography-guided rhFGF20 proteoliposomes. Drug Deliv. 25:1560–1569. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Cai X, Tao W and Li L: Glioma cell-derived FGF20 suppresses macrophage function by activating β-catenin. Cell Signal. 89:1101812022. View Article : Google Scholar : PubMed/NCBI

42 

Dabrowski A, Terauchi A, Strong C and Umemori H: Distinct sets of FGF receptors sculpt excitatory and inhibitory synaptogenesis. Development. 142:1818–1830. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Pasaoglu T and Schikorski T: Presynaptic size of associational/commissural CA3 synapses is controlled by fibroblast growth factor 22 in adult mice. Hippocampus. 26:151–160. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Aljović A, Jacobi A, Marcantoni M, Kagerer F, Loy K, Kendirli A, Bräutigam J, Fabbio L, Van Steenbergen V, Pleśniar K, et al: Synaptogenic gene therapy with FGF22 improves circuit plasticity and functional recovery following spinal cord injury. EMBO Mol Med. 15:e161112023. View Article : Google Scholar : PubMed/NCBI

45 

Jacobi A, Loy K, Schmalz AM, Hellsten M, Umemori H, Kerschensteiner M and Bareyre FM: FGF22 signaling regulates synapse formation during post-injury remodeling of the spinal cord. EMBO J. 34:1231–1243. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2024
Volume 28 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
Zhao K, Xu J and Zhao B: Panoramic RNA expression of fibroblast growth factors in human glioblastoma tissues and the impact on the survival of patients. Oncol Lett 28: 317, 2024
APA
Zhao, K., Xu, J., & Zhao, B. (2024). Panoramic RNA expression of fibroblast growth factors in human glioblastoma tissues and the impact on the survival of patients. Oncology Letters, 28, 317. https://doi.org/10.3892/ol.2024.14450
MLA
Zhao, K., Xu, J., Zhao, B."Panoramic RNA expression of fibroblast growth factors in human glioblastoma tissues and the impact on the survival of patients". Oncology Letters 28.1 (2024): 317.
Chicago
Zhao, K., Xu, J., Zhao, B."Panoramic RNA expression of fibroblast growth factors in human glioblastoma tissues and the impact on the survival of patients". Oncology Letters 28, no. 1 (2024): 317. https://doi.org/10.3892/ol.2024.14450