Next Article in Journal
A Randomized Controlled Trial on the Effects of Aerobic and Coordinative Training on Neural Correlates of Inhibitory Control in Children
Next Article in Special Issue
Obesity, Inflammation, and Prostate Cancer
Previous Article in Journal
The Homuncular Jigsaw: Investigations of Phantom Limb and Body Awareness Following Brachial Plexus Block or Avulsion
Previous Article in Special Issue
Pirfenidone, an Anti-Fibrotic Drug, Suppresses the Growth of Human Prostate Cancer Cells by Inducing G1 Cell Cycle Arrest
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Fibroblast Growth Factor Family in the Progression of Prostate Cancer

Department of Urology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2019, 8(2), 183; https://doi.org/10.3390/jcm8020183
Submission received: 10 January 2019 / Revised: 23 January 2019 / Accepted: 31 January 2019 / Published: 4 February 2019

Abstract

:
Fibroblast growth factors (FGFs) and FGF receptors (FGFRs) play an important role in the maintenance of tissue homeostasis and the development and differentiation of prostate tissue through epithelial-stromal interactions. Aberrations of this signaling are linked to the development and progression of prostate cancer (PCa). The FGF family includes two subfamilies, paracrine FGFs and endocrine FGFs. Paracrine FGFs directly bind the extracellular domain of FGFRs and act as a growth factor through the activation of tyrosine kinase signaling. Endocrine FGFs have a low affinity of heparin/heparan sulfate and are easy to circulate in serum. Their biological function is exerted as both a growth factor binding FGFRs with co-receptors and as an endocrine molecule. Many studies have demonstrated the significance of these FGFs and FGFRs in the development and progression of PCa. Herein, we discuss the current knowledge regarding the role of FGFs and FGFRs—including paracrine FGFs, endocrine FGFs, and FGFRs—in the development and progression of PCa, focusing on the representative molecules in each subfamily.

1. Introduction

Prostate cancer (PCa) is one of the most common hormone-dependent cancers. Androgen-deprived therapy (ADT) has been the standard option for PCa. It is initially effective in most PCa cases; however, PCa becomes refractory for ADT in spite of the castration level of serum testosterone, which is called “castration-resistant prostate cancer” (CRPC). There have been multiple studies on the efficacy of various agents that include androgen-receptor (AR) targeted agents and anticancer drugs.
Many studies have demonstrated the aberrant activation of fibroblast growth factor (FGF)/FGF receptor (FGFR) signaling in several cancers, including head and neck, lung, breast, endometrial, bladder, and prostate cancer [1,2]. Herein, we review the FGF family’s involvement in the development and progression of prostate cancer. We mainly discuss the representative molecules in each subfamily: FGF9 as classic FGFs, FGF19 as endocrine FGFs, and FGFR2IIIb.

2. FGFs and FGFRs

The human Fgf gene family consists of 22 members, and they are classified into seven subfamilies based on phylogenetic analysis (Figure 1) [3]. FGFs are also classified into three subfamilies (intracrine, paracrine, and endocrine FGFs) based on their mechanism of action. Intracrine FGFs are not typical and does not bind FGFR. Their function is not mediated by a receptor. Therefore, we herein focused two subfamilies, paracrine FGFs and endocrine FGFs. Paracrine FGFs—which consist of 15 members—exert their biological function through binding to an extracellular domain of FGFRs with heparin/ heparan sulfate and activating tyrosine kinase signaling of FGFRs.
Endocrine FGFs consist of FGF19, FGF21, and FGF23. These molecules have a low affinity to heparin/heparan sulfate in contrast to paracrine FGFs. They have to form complexes with co-receptors, α/β-Klotho, to bind to the extracellular domain of FGFR. Because the endocrine FGFs’ affinity to FGFRs changes depending on the existence of α/β-Klotho, they have hormone-like activity beyond functioning as a growth factor (Figure 2). Several studies have demonstrated the physiological function of endocrine FGFs (Table 1) [4]. Among endocrine FGFs, FGF15 (the mouse orthologue of human FGF19) was the first molecule to be identified. The physiological activity of FGF19, including the regulation of glucose and bile acid metabolism, is exerted through the formation of a complex with FGFR4 and beta-klotho and follows the activation of a signaling cascade by recruiting adaptor proteins in cytoplasm [5,6,7]. FGF21 mainly acts as a metabolic regulator in the liver, adipose tissue, and the pancreas. The tissue-specific metabolic action of FGF21 depends on its specificity to the receptor. FGF21 binds with beta-klotho and FGFR1c [6,8,9]. FGF23 is a bone-derived endocrine hormone. Expression of FGF23 is induced by activation of the vitamin D receptor (VDR) with 1,25 dihydroxyvitamin D (1,25D), and FGF23 is a suppressor of 1,25D. So FGF23 and 1,25D are linked by mutual regulation. A signaling cascade of FGF23 activates through formation of a complex with alfa-klotho and FGFR1 for the kidney and FGFR3c for the parathyroid gland. FGF23 regulates phosphate and vitamin D metabolism in the kidney and inhibits parathyroid hormone secretion and vitamin D synthesis in the parathyroid gland [10,11].
Four FGFRs, FGFR1-4, contain an extracellular ligand binding domain with three immunoglobulin (Ig)-like domains (I–III), a transmembrane domain, and a split intracellular tyrosine kinase domain. FGFR1-3 have two kinds of Ig-like III domains, IIIb and IIIc, which are generated by alternative splicing. The Ig-like domain is crucial for determining ligand-binding specificity, and as a result, seven FGFR with different ligand-binding specificities are derived from four Fgfr genes [12].

3. FGF-FGFR Signaling in Epithelial-Stromal Interaction in Prostate Tissue

Epithelial-stromal interaction plays an important role in maintaining the homeostasis in normal prostatic tissue [13,14]. Stromal tissues secrete paracrine factors that include FGF ligands, and they lead to stimulation of epithelial maintenance and growth. Huang et al. reported the significance of FGFR2 signaling for preserving stemness and preventing differentiation of prostate stem cells [15]. FGFR2IIIb, a splicing variant of the FGFR2, is a resident form of FGFRs expressed in normal prostate epithelial cells. FGFR2IIIb is specific to FGF7, and FGF7-FGFR2IIIb contributes epithelial-stromal interaction [16]. The loss of FGFR2 isoforms is shown in human PCa tissues, and the loss of FGFR2IIIb is associated with the characteristics of castration-resistant prostate cancer (CRPC) in particular [17].
The expression pattern of FGFR is different in each PCa cell line. For instance, FGFR1 expression is detectable and FGFR2IIIb expression is undetectable in PC3 cells that show androgen-independent growth and high potential of cell proliferation. On the other hand, FGFR2IIIb expression is detectable in LNCap cells that show expression of androgen receptor (AR), androgen-dependent growth, and relatively low potential of cell proliferation [18]. In addition to FGFs and FGFRs, FGF receptor substrate 2alpha (FRS2alpha), an FGFR interactive adaptor protein, involves multiple signaling pathways to the activated FGFR kinase. FRS2 alfa is required for prostate development and tumorigenesis [19], as well as in angiogenesis [20].

4. Effects of the Restoration of FGFR2IIIb in Prostate Cancer Cells

Many studies have demonstrated the association of aberrant FGFR signaling with the development and progression of PCa [21,22]. Binding ligands, FGFRs form functional dimerization and lead transphosphorylation and activation of downstream signaling pathways such as Ras, Src, PKCγ, MAPK, PI3K-AKT, and STAT [23,24,25]. The involvement of FGF signaling in various molecular mechanisms has been reported in PCa. Shao et al. reported that FGF-FGFR signaling plays an important role in transformation induced by the loss of a PTEN tumor suppressor when combined with the expression of the TMPRSS2/ERG fusion gene [26], and activation of FGF-FGFR signaling by FGF8b overexpression in PTEN deficiency is reported to be associated with prostate tumorigenesis [27]. FGF-FGFR signaling is also related to the induction of an inflammatory response in PCa tissues [28]. The involvement of aberrant FGFR1 signaling in the progression of PCa in particular was demonstrated in several studies. FGFR1 signaling promotes the reprogramming of energy metabolism from oxidative phosphorylation to aerobic glycolysis by regulating the expression of an LDH isoenzyme [29]. It also promotes an inflammatory response through activation of NF-κB signaling [30]. Furthermore, activation of FGFR1 signaling promotes epithelial to mesenchymal transition and androgen independency in PCa cells [31,32]. Loss of FGFR2IIIb and enhancement of ectopic expression of FGFR1 PCa progression have been reported as common events in the progression of PCa [21]. As FGFR2IIIb plays an important role in the maintenance and its disorder is found in PCa cell lines and tissues, several investigators have reported the effects of restoring FGFR2IIIb. In animal models and PCa cell lines, FGFR2IIIb’s restoration also restored responsiveness to stroma and significantly reduced in vivo tumorigenesis. In castration-resistant human PCa cells, restoration of FGFR2IIIb showed the inhibition of cell proliferation, the induction of differentiation, and the enhancement of apoptosis in a ligand-independent manner [21,33,34]. In addition, in PCa cells overexpressing FGFR2IIIb, clonogenic cell death increased in concurrence with enhanced apoptosis and cell cycle arrest in the G2/M-phase and radiosensitivity by gamma-irradiation [35]. Another study reported the effect FGFR2IIIb’s restoration had on the chemosensitivity in PCa cells. Restoration of FGFR2IIIb led to the enhanced chemosensitivity of several agents, especially docetaxel. The expression of N-cadherin, vimentin, survivin, and XIAP were induced by restoring FGFR2IIIb [36]. This data indicates that PCa cell lines are induced to a more differentiated phenotype when changing the pattern of gene expression that became sensitive to radiation and chemotherapy when FGFR2IIIb was restored.

5. Involvement of FGFs in the Development and Progression of Prostate Cancer

5.1. Paracrine FGFs

Upregulated expression of FGF1, FGF2, FGF8, FGF9, and FGF10 were shown in human PCa [37]. Murine studies demonstrated the epithelial and mesenchymal interaction using a FGF/FGFR complex in PCa [38]. Pecqueux et al. demonstrated the association between strong expression of FGF2 in tumor stroma and a high postoperative recurrence rate and that exogenous FGF2 can drive genomic instability to promote PCa progression through enhancement of DNA damage [39]. Cuevas et al. reported the linkage between altered micro-environmental signaling that includes FGF2 overexpression and mitotic instability [40]. In the study focused on a bone metastatic site, FGF2 was upregulated in osteoblast and promoted the proliferation of PCa cells under the loss of TGFβ signaling [41]. These reports indicated the significance of FGF/FGFR signaling through paracrine FGFs (especially FGF2) in a cancer micro-environment for the development and progression of PCa.
FGF9 is an abundant molecule in nervous tissue and soft tissue, while it has been reported to be a key molecule of epithelial-stromal interaction [42]. Several studies have demonstrated that FGF9 was associated with the proliferation of glia [43], regulation of the differentiation of astrocytes [44] and oligodendrocytes [45], and the regulation of joint formations [46,47]. FGF9’s involvement in malignant neoplasms has been reported in glioma [48], ovarian cancer [49], and lung cancer [50]. In prostate epithelial cells, overexpression of FGF9 lead the augmentation of reactive stroma formation and promoted initiation and progression of PCa [51]. In our study, cell viability and invasion of LNCaP was significantly enhanced, and expression of MMP2, N-cadherin, and VEGF-A were induced in LNCaP incubated in medium with FGF9. In immuno-histochemical staining, the prevalence of both VEGF-A and N-cadherin-positive cells was significantly higher compared to FGF9-negative cases [52]. The biochemical relapse-free survival (bRFS) rate in cases with FGF9-positive cases was significantly lower than that in FGF9-negative cases [53]. “FGF9-positive” in this study was determined based on the findings of immunohistochemistry staining on just one representative section. And even in FGF9-positive cases, just a small population of FGF9-positive cells with very aggressive pathological features was present. In other words, FGF9 was only positive in especially high-grade cancer cells in cases with localized PCa. Furthermore, several studies have reported FGF9 in PCa at an advanced stage. Accumulation of FGF9 to the region of bone metastasis formed by AR-negative PCa cells indicated that FGF9 was a key factor in formation of bone metastasis [54]. In AR-negative CRPC cases, neuroendocrine differentiation (NE) is one of the most representative phenotypes. In a further dedifferentiated phenotype, “AR-negative and NE-negative”, expression of FGF1, FGF8, and FGF9 increased, and MAPK pathway was activated [55]. The results of these studies indicated that FGF9 might be a key molecule for an advanced PCa that include CRPC rather than a localized one.

5.2. Endocrine FGFs

Endocrine FGFs have two different characteristics, a growth factor and a metabolic regulator. Investigating endocrine FGFs might clarify the molecular link between the progression of cancer and metabolism. Among endocrine FGFs, the association between FGF19 and malignant diseases such as liver cancer, colon cancer, and prostate cancer was demonstrated [56,57,58,59]. FGF19 induces the expression of markers of epithelial mesenchymal transition in hormone-sensitive prostate cancer cells [60,61]. Consistent with the results, accumulation of FGF19 in cytoplasm was shown in poorly differentiated prostate cancer cells in human prostate cancer tissues derived from radical prostatectomy, and the presence of FGF19-positive tissues correlated with positive immuno-histochemical staining with N-cadherin in prostate cancer tissues [60]. The bRFS rate in cases with FGF19-positive tissues was significantly lower than in cases with FGF19-negative tissues [60]. In prostate cancer cells, FGF19 stimulates cell proliferation and cell invasion through activation of MAP kinase and AKT pathways [62]. Expression of FGF19 increased in castration-resistant cell lines compared with castration-sensitive ones or immortalized normal prostate cells [62]. According to these reports, FGF19 might be associated with the risk of post-operative recurrence by enhancement of cell proliferation and epithelial-mesenchymal transition of PCa cells.
Since endocrine FGFs that include FGF19 act as circulating hormones related to several metabolic diseases, the impact of their serum concentration for metabolic diseases was investigated. These molecules are expected to be a potential serum biomarker for PCa. We measured serum FGF19 and beta-klotho level in cases with PCa. While there was no relationship between the serum klotho level and pathological findings, the results showed that patients with a high Gleason score had higher serum FGF19 levels than those with a low Gleason score [60]. This data indicates that FGF19 might be a potential serum biomarker in PCa. One limitation to using endocrine FGFs that include FGF19 as a serum biomarker is the change of serum concentration according to dietary conditions and blood sugar level because endocrine FGFs act as a metabolic regulator and their level is also regulated by a feedback mechanism. Further study will clarify the optimal timing and conditions for measuring endocrine FGFs to apply them as a serum biomarker for PCa. Besides FGF19, the increased expression of FGF23 in many PCa cell lines and PCa tissues was reported. FGF23 enhances proliferation, invasion through activation of AKT, and the MAPK pathway in PCa cell lines. These findings indicate that it can promote PCa progression in an autocrine, paracrine, and/or endocrine manner [63].

6. Conclusions

In this article, we described the impact of FGF-FGFR abnormalities on the development and/or the progression of PCa. Since we found the heterogeneity of PCa, it is important to clarify and understand various molecular mechanisms of PCa in order to determine the most appropriate therapeutic strategies.

Author Contributions

Conceptualization, J.T. and A.M.; methodology, J.T.; software, J.T.; formal analysis, K.G., R.Y., H.S., K.S. and H.N.; investigation, J.T., T.H., K.G., K.S. and H.N.; writing—original draft preparation, J.T.; writing—review and editing, J.T. and T.H.; visualization, S.I. and Y.S.; supervision, K.G., T.H., S.I., Y.S. and A.M.; funding acquisition, J.T. and A.M.

Funding

This research was supported by Grants-in-Aid for Scientific Research(C), grant number 18K09134 from the Japan Society for the Promotion of Science.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Corn, P.G.; Wang, F.; McKeehan, W.L.; Navone, N. Targeting fibroblast growth factor pathways in prostate cancer. Clin. Cancer. Res. 2013, 19, 5856–5866. [Google Scholar] [CrossRef] [PubMed]
  2. Wesche, J.; Haglund, K.; Haugsten, E.M. Fibroblast growth factors and their receptors in cancer. Biochem. J. 2011, 437, 199–213. [Google Scholar] [CrossRef] [Green Version]
  3. Itoh, N.; Ornitz, D.M. Fibroblast growth factors: From molecular evolution to roles in development, metabolism and disease. J. Biochem. 2011, 149, 121–130. [Google Scholar] [CrossRef] [PubMed]
  4. Degirolamo, C.; Sabbà, C.; Moschetta, A. Therapeutic potential of the endocrine fibroblast growth factors FGF19, FGF21 and FGF23. Nat. Rev. Drug Discov. 2016, 15, 51–69. [Google Scholar] [CrossRef] [PubMed]
  5. Lin, B.C.; Wang, M.; Blackmore, C.; Desnoyers, L.R. Liver-specific activities of FGF19 require Klotho beta. J. Biol. Chem. 2007, 282, 27277–27284. [Google Scholar] [CrossRef] [PubMed]
  6. Kurosu, H.; Choi, M.; Ogawa, Y.; Dickson, A.S.; Goetz, R.; Eliseenkova, A.V.; Mohammadi, M.; Rosenblatt, K.P.; Kliewer, S.A.; Kuro-o, M. Tissue-specific expression of betaKlotho and fibroblast growth factor (FGF) receptor isoforms determines metabolic activity of FGF19 and FGF21. J. Biol. Chem. 2007, 282, 26687–26695. [Google Scholar] [CrossRef] [PubMed]
  7. Xie, M.H.; Holcomb, I.; Deuel, B.; Dowd, P.; Huang, A.; Vagts, A.; Foster, J.; Liang, J.; Brush, J.; Gu, Q.; et al. FGF-19, a novel fibroblast growth factor with unique specificity for FGFR4. Cytokine 1999, 11, 729–735. [Google Scholar] [CrossRef]
  8. Ogawa, Y.; Kurosu, H.; Yamamoto, M.; Nandi, A.; Rosenblatt, K.P.; Goetz, R.; Eliseenkova, A.V.; Mohammadi, M.; Kuro-o, M. β-Klotho is required for metabolic activity of fibroblast growth factor 21. Proc. Natl. Acad. Sci. USA 2007, 104, 7432–7437. [Google Scholar] [CrossRef]
  9. Ding, X.; Boney-Montoya, J.; Owen, B.M.; Bookout, A.L.; Coate, K.C.; Mangelsdorf, D.J.; Kliewer, S.A. β-Klotho is required for fibroblast growth factor 21 effects on growth and metabolism. Cell Metab. 2012, 16, 387–393. [Google Scholar] [CrossRef]
  10. Kolek, O.I.; Hines, E.R.; Jones, M.D.; LeSueur, L.K.; Lipko, M.A.; Kiela, P.R.; Collins, J.F.; Haussler, M.R.; Ghishan, F.K. 1alpha, 25-Dihydroxyvitamin D3 upregulates FGF23 gene expression in bone: The final link in a renal-gastrointestinal-skeletal axis that controls phosphate transport. Am. J. Physiol. Gastrointest. Liver. Physiol. 2005, 289, G1036–G1042. [Google Scholar] [CrossRef]
  11. Yu, X.; Sabbagh, Y.; Davis, S.I.; Demay, M.B.; White, K.E. Genetic dissection of phosphate- and vitamin D-mediated regulation of circulating Fgf23 concentrations. Bone 2005, 36, 971–977. [Google Scholar] [CrossRef] [PubMed]
  12. Zhang, X.; Ibrahimi, O.A.; Olsen, S.K.; Umemori, H.; Mohammadi, M.; Ornitz, D.M. Receptor specificity of the fibroblast growth factor family. The complete mammalian FGF family. J. Biol. Chem. 2006, 281, 15694–15700. [Google Scholar] [CrossRef] [PubMed]
  13. Lin, Y.; Wang, F. FGF signaling in prostate development, tissue homoeostasis and tumorigenesis. Biosci. Rep. 2010, 30, 285–291. [Google Scholar] [CrossRef] [PubMed]
  14. Cotton, L.M.; O’Bryan, M.K.; Hinton, B.T. Cellular signaling by fibroblast growth factors (FGFs) and their receptors (FGFRs) in male reproduction. Endocr. Rev. 2008, 29, 193–216. [Google Scholar] [CrossRef] [PubMed]
  15. Huang, Y.; Hamana, T.; Liu, J.; Wang, C.; An, L.; You, P.; Chang, J.Y.; Xu, J.; Jin, C.; Zhang, Z.; et al. Type 2 Fibroblast Growth Factor Receptor Signaling Preserves Stemness and Prevents Differentiation of Prostate Stem Cells from the Basal Compartment. J. Biol. Chem. 2015, 290, 17753–17761. [Google Scholar] [CrossRef] [PubMed]
  16. Lin, Y.; Liu, G.; Zhang, Y.; Hu, Y.P.; Yu, K.; Lin, C.; McKeehan, K.; Xuan, J.W.; Ornitz, D.M.; Shen, M.M.; et al. Fibroblast growth factor receptor 2 tyrosine kinase is required for prostatic morphogenesis and the acquisition of strict androgen dependency for adult tissue homeostasis. Development 2007, 134, 723–734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Matsubara, A.; Yasumoto, H.; Usui, T. Hormone Refractory Prostate Cancer and Fibroblast Growth Factor Receptor. Breast Cancer 1999, 6, 320–324. [Google Scholar] [CrossRef]
  18. Carstens, R.P.; Eaton, J.V.; Krigman, H.R.; Walther, P.J.; Garcia-Blanco, M.A. Alternative splicing of fibroblast growth factor receptor 2 (FGF-R2) in human prostate cancer. Oncogene 1997, 15, 3059–3065. [Google Scholar] [CrossRef] [Green Version]
  19. Zhang, Y.; Zhang, J.; Lin, Y.; Lan, Y.; Lin, C.; Xuan, J.W.; Shen, M.M.; McKeehan, W.L.; Greenberg, N.M.; Wang, F. Role of epithelial cell fibroblast growth factor receptor substrate 2alpha in prostate development, regeneration and tumorigenesis. Development 2008, 135, 775–784. [Google Scholar] [CrossRef]
  20. Liu, J.; You, P.; Chen, G.; Fu, X.; Zeng, X.; Wang, C.; Huang, Y.; An, L.; Wan, X.; Navone, N.; et al. Hyperactivated FRS2α-mediated signaling in prostate cancer cells promotes tumor angiogenesis and predicts poor clinical outcome of patients. Oncogene 2016, 35, 1750–1759. [Google Scholar] [CrossRef]
  21. Lu, W.; Luo, Y.; Kan, M.; McKeehan, W.L. Fibroblast growth factor-10. A second candidate stromal to epithelial cell andromedin in prostate. J. Biol. Chem. 1999, 274, 12827–12834. [Google Scholar] [CrossRef]
  22. Feng, S.; Wang, F.; Matsubara, A.; Kan, M.; McKeehan, W.L. Fibroblast growth factor receptor 2 limits and receptor 1 accelerates tumorigenicity of prostate epithelial cells. Cancer Res. 1997, 57, 5369–5378. [Google Scholar] [PubMed]
  23. Turner, N.; Grose, R. Fibroblast growth factor signaling: From development to cancer. Nat. Rev. Cancer 2010, 10, 116–129. [Google Scholar] [CrossRef] [PubMed]
  24. Beenken, A.; Mohammadi, M. The FGF family: Biology, pathophysiology and therapy. Nat. Rev. Drug Discov. 2009, 8, 235–253. [Google Scholar] [CrossRef] [PubMed]
  25. Li, Q.; Ingram, L.; Kim, S.; Beharry, Z.; Cooper, J.A.; Cai, H. Paracrine Fibroblast Growth Factor Initiates Oncogenic Synergy with Epithelial FGFR/Src Transformation in Prostate Tumor Progression. Neoplasia 2018, 20, 233–243. [Google Scholar] [CrossRef] [PubMed]
  26. Shao, L.; Wang, J.; Karatas, O.F.; Feng, S.; Zhang, Y.; Creighton, C.J.; Ittmann, M. Fibroblast growth factor receptor signaling plays a key role in transformation induced by the TMPRSS2/ERG fusion gene and decreased PTEN. Oncotarget 2018, 9, 14456–14471. [Google Scholar] [CrossRef] [PubMed]
  27. Zhong, C.; Saribekyan, G.; Liao, C.P.; Cohen, M.B.; Roy-Burman, P. Cooperation between FGF8b overexpression and PTEN deficiency in prostate tumorigenesis. Cancer Res. 2006, 66, 2188–2194. [Google Scholar] [CrossRef]
  28. Ronca, R.; Tamma, R.; Coltrini, D.; Ruggieri, S.; Presta, M.; Ribatti, D. Fibroblast growth factor modulates mast cell recruitment in a murine model of prostate cancer. Oncotarget 2017, 8, 82583–82592. [Google Scholar] [CrossRef]
  29. Liu, J.; Chen, G.; Liu, Z.; Liu, S.; Cai, Z.; You, P.; Ke, Y.; Lai, L.; Huang, Y.; Gao, H.; et al. Aberrant FGFR tyrosine kinase signaling enhances the Warburg effect by reprogramming LDH isoform expression and activity in prostate cancer. Cancer Res. 2018, 78, 4459–4470. [Google Scholar] [CrossRef]
  30. Wang, C.; Ke, Y.; Liu, S.; Pan, S.; Liu, Z.; Zhang, H.; Fan, Z.; Zhou, C.; Liu, J.; Wang, F. Ectopic fibroblast growth factor receptor 1 promotes inflammation by promoting nuclear factor-κB signaling in prostate cancer cells. J. Biol. Chem. 2018, 293, 14839–14849. [Google Scholar] [CrossRef]
  31. Acevedo, V.D.; Gangula, R.D.; Freeman, K.W.; Li, R.; Zhang, Y.; Wang, F.; Ayala, G.E.; Peterson, L.E.; Ittmann, M.; Spencer, D.M. Inducible FGFR-1 activation leads to irreversible prostate adenocarcinoma and an epithelial-to-mesenchymal transition. Cancer Cell. 2007, 12, 559–571. [Google Scholar] [CrossRef] [PubMed]
  32. Kobayashi, M.; Huang, Y.; Jin, C.; Luo, Y.; Okamoto, T.; Wang, F.; McKeehan, W.L. FGFR1 abrogates inhibitory effect of androgen receptor concurrent with induction of androgen-receptor variants in androgen receptor-negative prostate tumor epithelial cells. Prostate 2011, 71, 1691–1700. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Matsubara, A.; Kan, M.; Feng, S.; McKeehan, W.L. Inhibition of growth of malignant rat prostate tumor cells by restoration of fibroblast growth factor receptor 2. Cancer Res 1998, 58, 1509–1514. [Google Scholar] [PubMed]
  34. Yasumoto, H.; Matsubara, A.; Mutaguchi, K.; Usui, T.; McKeehan, W.L. Restoration of fibroblast growth factor receptor2 suppresses growth and tumorigenicity of malignant human prostate carcinoma PC-3 cells. Prostate 2004, 61, 236–242. [Google Scholar] [CrossRef] [PubMed]
  35. Matsubara, A.; Teishima, J.; Mirkhat, S.; Yasumoto, H.; Mochizuki, H.; Seki, M.; Mutaguchi, K.; Mckeehan, W.L.; Usui, T. Restoration of FGF receptor type 2 enhances radiosensitivity of hormone-refractory human prostate carcinoma PC-3 cells. Anticancer Res. 2008, 28, 2141–2146. [Google Scholar] [PubMed]
  36. Shoji, K.; Teishima, J.; Hayashi, T.; Ohara, S.; Mckeehan, W.L.; Matsubara, A. Restoration of fibroblast growth factor receptor 2IIIb enhances the chemosensitivity of human prostate cancer cells. Oncol. Rep. 2014, 32, 65–70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Ronca, R.; Giacomini, A.; Di Salle, E.; Coltrini, D.; Pagano, K.; Ragona, L.; Matarazzo, S.; Rezzola, S.; Maiolo, D.; Torrella, R.; et al. Long-Pentraxin 3 derivative as a small-molecule FGF trap for cancer therapy. Cancer Cell. 2015, 28, 225–239. [Google Scholar] [CrossRef]
  38. Memarzadeh, S.; Xin, L.; Mulholland, D.J.; Mansukhani, A.; Wu, H.; Teitell, M.A.; Witte, O.N. Enhanced paracrine FGF10 expression promotes formation of multifocal prostate adenocarcinoma and an increase in epithelial androgen receptor. Cancer Cell. 2007, 12, 572–585. [Google Scholar] [CrossRef]
  39. Pecqueux, C.; Arslan, A.; Heller, M.; Falkenstein, M.; Kaczorowski, A.; Tolstov, Y.; Sültmann, H.; Grüllich, C.; Herpel, E.; Duensing, A. FGF-2 is a driving force for chromosomal instability and a stromal factor associated with adverse clinico-pathological features in prostate cancer. Urol. Oncol. 2018, 36, e15–e365. [Google Scholar] [CrossRef]
  40. Cuevas, R.; Korzeniewski, N.; Tolstov, Y.; Hohenfellner, M.; Duensing, S. FGF-2 disrupts mitotic stability in prostate cancer through the intracellular trafficking protein CEP57. Cancer Res. 2013, 73, 1400–1410. [Google Scholar] [CrossRef]
  41. Meng, X.; Vander Ark, A.; Daft, P.; Woodford, E.; Wang, J.; Madaj, Z.; Li, X. Loss of TGF-β signaling in osteoblasts increases basic-FGF and promotes prostate cancer bone metastasis. Cancer Lett. 2018, 418, 109–118. [Google Scholar] [CrossRef] [PubMed]
  42. Jin, C.; Wang, F.; Wu, X.; Yu, C.; Luo, Y.; McKeehan, W.L. Directionally specific paracrine communication mediated by epithelial FGF9 to stromal FGFR3 in two-compartment premalignant prostate tumors. Cancer Res. 2004, 64, 4555–4562. [Google Scholar] [CrossRef] [PubMed]
  43. Reuss, B.; Hertel, M.; Werner, S.; Unsicker, K. Fibroblast growth factors-5 and -9 distinctly regulate expression and function of the gap junction protein connexin43 in cultured astroglial cells from different brain regions. Glia 2000, 30, 231–241. [Google Scholar] [CrossRef]
  44. Lum, M.; Turbic, A.; Mitrovic, B.; Turnley, A.M.J. Fibroblast growth factor-9 inhibits astrocyte differentiation of adult mouse neural progenitor cells. Neurosci. Res. 2009, 87, 2201–2210. [Google Scholar] [CrossRef] [PubMed]
  45. Cohen, R.I.; Chandross, K.J. Fibroblast growth factor-9 modulates the expression of myelin related proteins and multiple fibroblast growth factor receptors in developing oligodendrocytes. J. Neurosci. Res. 2000, 61, 273–287. [Google Scholar] [CrossRef]
  46. Harada, M.; Murakami, H.; Okawa, A.; Okimoto, N.; Hiraoka, S.; Nakahara, T.; Akasaka, R.; Shiraishi, Y.; Futatsugi, N.; Mizutani-Koseki, Y.; et al. FGF9 monomer/ dimer equilibrium regulates extracellular matrix affinity and tissue diffusion. Nat. Genet. 2009, 41, 289–298. [Google Scholar] [CrossRef] [PubMed]
  47. Kalinina, J.; Byron, S.A.; Makarenkova, H.P.; Olsen, S.K.; Eliseenkova, A.V.; Larochelle, W.J.; Dhanabal, M.; Blais, S.; Ornitz, D.M.; Day, L.A.; et al. Homodimerization controls the fibroblast growth factor 9 subfamily’s receptor binding and heparan sulfate-dependent diffusion in the extracellular matrix. Mol. Cell. Biol. 2009, 29, 4663–4678. [Google Scholar] [CrossRef]
  48. Miyagi, N.; Kato, S.; Terasaki, M.; Aoki, T.; Sugita, Y.; Yamaguchi, M.; Shigemori, M.; Morimatsu, M. Fibroblast growth factor-9 (glia-activating factor) stimulates proliferation and production of glial fibrillary acidic protein in human gliomas either in the presence or in the absence of the endogenous growth factor expression. Oncol. Rep. 1999, 6, 87–92. [Google Scholar] [CrossRef]
  49. Hendrix, N.D.; Wu, R.; Kuick, R.; Schwartz, D.R.; Fearon, E.R.; Cho, K.R. Fibroblast growth factor 9 has oncogenic activity and is a downstream target of Wnt signaling in ovarian endometrioid adenocarcinomas. Cancer Res. 2006, 66, 1354–1362. [Google Scholar] [CrossRef]
  50. Wang, C.K.; Chang, H.; Chen, P.H.; Chang, J.T.; Kuo, Y.C.; Ko, J.L.; Lin, P. Aryl hydrocarbon receptor activation and overexpression upregulated fibroblast growth factor-9 in human lung adenocarcinomas. Int. J. Cancer. 2009, 125, 807–815. [Google Scholar] [CrossRef] [Green Version]
  51. Huang, Y.; Jin, C.; Hamana, T.; Liu, J.; Wang, C.; An, L.; McKeehan, W.L.; Wang, F. Overexpression of FGF9 in prostate epithelial cells augments reactive stroma formation and promotes prostate cancer progression. Int. J. Biol. Sci. 2015, 11, 948–960. [Google Scholar] [CrossRef] [PubMed]
  52. Teishima, J.; Yano, S.; Shoji, K.; Hayashi, T.; Goto, K.; Kitano, H.; Oka, K.; Nagamatsu, H.; Matsubara, A. Accumulation of FGF9 in prostate cancer correlates with epithelial-to-mesenchymal transition and induction of VEGF-A expression. Anticancer Res. 2014, 34, 695–700. [Google Scholar]
  53. Teishima, J.; Shoji, K.; Hayashi, T.; Miyamoto, K.; Ohara, S.; Matsubara, A. Relationship between the localization of fibroblast growth factor 9 in prostate cancer cells and postoperative recurrence. Prostate Cancer Prostatic Dis. 2012, 15, 8–14. [Google Scholar] [CrossRef] [PubMed]
  54. Li, ZG.; Mathew, P.; Yang, J.; Starbuck, M.W.; Zurita, A.J.; Liu, J.; Sikes, C.; Multani, A.S.; Efstathiou, E.; Lopez, A.; et al. Androgen receptor-negative human prostate cancer cells induce osteogenesis in mice through FGF9-mediated mechanisms. J. Clin. Investig. 2008, 118, 2697–2710. [Google Scholar] [CrossRef] [PubMed]
  55. Bluemn, E.G.; Coleman, I.M.; Lucas, J.M.; Coleman, R.T.; Hernandez-Lopez, S.; Tharakan, R.; Bianchi-Frias, D.; Dumpit, R.F.; Kaipainen, A.; Corella, A.N.; et al. Androgen Receptor Pathway-Independent Prostate Cancer Is Sustained through FGF Signaling. Cancer Cell. 2017, 32, 474–489. [Google Scholar] [CrossRef] [PubMed]
  56. Nicholes, K.; Guillet, S.; Tomlinson, E.; Hillan, K.; Wright, B.; Frantz, G.D.; Pham, T.A.; Dillard-Telm, L.; Tsai, S.P.; Stephan, J.P.; et al. A mouse model of hepatocellular carcinoma: Ectopic expression of fibroblast growth factor 19 in skeletal muscle of transgenic mice. Am. J. Pathol. 2002, 160, 2295–2307. [Google Scholar] [CrossRef]
  57. Alvarez-Sola, G.; Uriarte, I.; Latasa, M.U.; Urtasun, R.; Bárcena-Varela, M.; Elizalde, M.; Jiménez, M.; Rodriguez-Ortigosa, C.M.; Corrales, F.J.; Fernández-Barrena, M.G.; et al. Fibroblast growth factor 15/19 in hepatocarcinogenesis. Dig. Dis. 2017, 35, 158–165. [Google Scholar] [CrossRef]
  58. Desnoyers, L.R.; Pai, R.; Ferrando, R.E.; Hötzel, K.; Le, T.; Ross, J.; Carano, R.; D’Souza, A.; Qing, J.; Mohtashemi, I.; et al. Targeting FGF19 inhibits tumor growth in colon cancer xenograft and FGF19 transgenic hepatocellular carcinoma models. Oncogene 2008, 27, 85–97. [Google Scholar] [CrossRef]
  59. Pai, R.; Dunlap, D.; Qing, J.; Mohtashemi, I.; Hotzel, K.; French, D.M. Inhibition of fibroblast growth factor 19 reduces tumor growth by modulating beta-catenin signaling. Cancer Res. 2008, 68, 5086–5095. [Google Scholar] [CrossRef]
  60. Nagamatsu, H.; Teishima, J.; Goto, K.; Shikuma, H.; Kitano, H.; Shoji, K.; Inoue, S.; Matsubara, A. FGF19 promotes progression of prostate cancer. Prostate 2015, 75, 1092–1101. [Google Scholar] [CrossRef]
  61. Liu, Z.; Zhang, H.; Ding, S.; Qi, S.; Liu, S.; Sun, D.; Dong, W.; Yin, L.; Li, M.; Zhao, X.; et al. βKlotho inhibits androgen/androgen receptor‑associated epithelial‑mesenchymal transition in prostate cancer through inactivation of ERK1/2 signaling. Oncol. Rep. 2018, 40, 217–225. [Google Scholar] [CrossRef] [PubMed]
  62. Feng, S.; Dakhova, O.; Creighton, C.J.; Ittmann, M. Endocrine fibroblast growth factor FGF19 promotes prostate cancer progression. Cancer Res. 2013, 73, 2551–2562. [Google Scholar] [CrossRef] [PubMed]
  63. Feng, S.; Wang, J.; Zhang, Y.; Creighton, C.J.; Ittmann, M. FGF23 promotes prostate cancer progression. Oncotarget 2015, 6, 17291–17301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Fgf genes consisting of 7 subfamilies and Fgfr genes. Asterisks indicates fibroblast growth factor (FGF)/ fibroblast growth factor receptor (FGFR) whose expression are enhanced in prostate cancer cells and/or tissues.
Figure 1. Fgf genes consisting of 7 subfamilies and Fgfr genes. Asterisks indicates fibroblast growth factor (FGF)/ fibroblast growth factor receptor (FGFR) whose expression are enhanced in prostate cancer cells and/or tissues.
Jcm 08 00183 g001
Figure 2. Difference in the mechanism in signal transduction between paracrine FGFs and endocrine FGFs.
Figure 2. Difference in the mechanism in signal transduction between paracrine FGFs and endocrine FGFs.
Jcm 08 00183 g002
Table 1. Pathophysiological activity of endocrine FGFs.
Table 1. Pathophysiological activity of endocrine FGFs.
Up-RegulationDown-RegulationIncreaseDecrease
FGF19
Glycogen synthesisBile acid synthesisExtrahepatic cholestasisIBD
GluconeogenesisChronic hemodialysisNAFLD
Primary bile acid malabsorption
Obesity
FGF21
Hepatic fatty acid oxidationOvulationType 2 diabetesAnorexia
KetogenesisGrowth hormone signalingMetabolic syndromeNervosa
Glucogenesis NAFLD
Thermogenesis Coronary heart disease
WAT browing
Growth hormone resistance
Weight loss
Ovulation
FGF23
Calcium secretionRenal phosphate absorptionADHRHemodialysis
Life spanBone and renal calcium reabsorptionXLH ricketsFamilial tumoral calcinosis
Vitamin D synthesisTIO
PTH secretionCardiac hypertrophy
ADHR, autosomal dominant hypophosphataemic rickets; FGF, fibroblast growth factor; IBD, irritable bowel disease; NAFLD, non-alcoholic fatty liver disease; PTH, parathyroid hormone; TIO, tumor-induced osteomalacia; WAT, white adipose tissue; and XLH, X-linked hypophosphataemic.

Share and Cite

MDPI and ACS Style

Teishima, J.; Hayashi, T.; Nagamatsu, H.; Shoji, K.; Shikuma, H.; Yamanaka, R.; Sekino, Y.; Goto, K.; Inoue, S.; Matsubara, A. Fibroblast Growth Factor Family in the Progression of Prostate Cancer. J. Clin. Med. 2019, 8, 183. https://doi.org/10.3390/jcm8020183

AMA Style

Teishima J, Hayashi T, Nagamatsu H, Shoji K, Shikuma H, Yamanaka R, Sekino Y, Goto K, Inoue S, Matsubara A. Fibroblast Growth Factor Family in the Progression of Prostate Cancer. Journal of Clinical Medicine. 2019; 8(2):183. https://doi.org/10.3390/jcm8020183

Chicago/Turabian Style

Teishima, Jun, Tetsutaro Hayashi, Hirotaka Nagamatsu, Koichi Shoji, Hiroyuki Shikuma, Ryoken Yamanaka, Yohei Sekino, Keisuke Goto, Shogo Inoue, and Akio Matsubara. 2019. "Fibroblast Growth Factor Family in the Progression of Prostate Cancer" Journal of Clinical Medicine 8, no. 2: 183. https://doi.org/10.3390/jcm8020183

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop