Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-x4r87 Total loading time: 0 Render date: 2024-04-28T23:36:32.672Z Has data issue: false hasContentIssue false

16 - The FGF signaling axis in prostate tumorigenesis

from Part 2.1 - Molecular pathways underlying carcinogenesis: signal transduction

Published online by Cambridge University Press:  05 February 2015

Fen Wang
Affiliation:
Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX,USA
Yongde Luo
Affiliation:
Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
Wallace L. McKeehan
Affiliation:
Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

The prostate and prostate cancer

The prostate is an acorn-shaped accessory gland of the male reproductive system that secretes a variety of products into semen. Human prostate has three distinct zones: the peripheral zone, the transition zone, and the central zone. In Western societies, prostate cancer is the most frequently diagnosed neoplasm in males and the second leading cause of cancer mortality (1). About 5, 10, and 85% of human prostate cancer cases originate from the central zone, transition zone, and peripheral zone, respectively (1). Early prostate lesions are organ-confined, androgen-responsive, and are often alleviated by surgery (2). Over time and progression to advanced stages, however, the lesions frequently metastasize to bones, lymph nodes, and other organs, and usually become androgen-insensitive and lethal. To date, there is still no effective therapy for advanced prostate cancer. Understanding the mechanisms underlying the onset, progression to malignancy, and metastasis of prostate cancer is needed for developing new diagnostic, preventive, and therapeutic approaches.

The progression of prostate cancer is a slow and multi-step process. Although prostate cancer originates as focal lesions at early ages, clinically detectable prostate cancers are usually manifested in men after age 60. Animal models that mimic human prostate cancer initiation, progression, and metastasis are needed to understand the etiology and development of prostate cancer, and to test new prevention and intervention strategies for prostate cancer. Rodents have a strikingly different prostate morphology than humans and seldom develop spontaneous prostate tumors. Rare spontaneous and induced transplantable tumors from rats such as the Dunning tumor model that recapitulates some aspects of human prostate cancer progression in respect to stroma and epithelial compartments has been widely productive in understanding the general mechanisms underlying prostate tumor progression and castration resistance (3). However, the infrequent spontaneity of tumors in rodents and limitations in genetic manipulation of rats has made genetic manipulation in mice the avenue for developing new prostate tumor models. The mouse prostate consists of four distinct pairs of lobes: the anterior lobes (AP, also known as the coagulating gland), dorsal lobes (DP), lateral lobes (LP), and ventral lobe (VP; 4,5). The dorsal and lateral lobes are often referred to as the dorsolateral lobe (DLP). Despite the difference in organ morphology, both human and mouse prostates are composed of a complex ductal network that has general epithelial and stromal compartments comprised of diverse subcellular types within each (6).

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 190 - 203
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Jemal, A, Siegel, R, Ward, E, et al. Cancer statistics. CA: A cancer journal for clinicians 2009;59:225–49.
Denmeade, SR, Isaacs, JT. A history of prostate cancer treatment. Nature Reviews Cancer 2002;2:389–96.CrossRef
Tennant, TR, Kim, H, Sokoloff, M, Rinker-Schaeffer, CW.The Dunning model. The Prostate 2000;43:295–302.3.0.CO;2-W>CrossRefGoogle ScholarPubMed
Hayashi, N, Sugimura, Y, Kawamura, J, Donjacour, AA, Cunha, GR. Morphological and functional heterogeneity in the rat prostatic gland. Biology of Reproduction 1991;45:308–21.CrossRef
Sugimura, Y, Cunha, GR, Donjacour, AA. Morphogenesis of ductal networks in the mouse prostate. Biology of Reproduction 1986;34:961–71.CrossRef
Cunha, GR, Ricke, W, Thomson, A, et al. Hormonal, cellular, and molecular regulation of normal and neoplastic prostatic development. Journal of Steroid Biochemistry and Molecular Biology 2004;92:221–36.CrossRefGoogle ScholarPubMed
Shappell, SB, Thomas, GV, Roberts, RL, et al. Prostate pathology of genetically engineered mice: definitions and classification. The consensus report from the Bar Harbor meeting of the Mouse Models of Human Cancer Consortium Prostate Pathology Committee. Cancer Research 2004;64:2270–305.CrossRef
Portela-Gomes, GM, Lukinius, A, Grimelius, L.Synaptic vesicle protein 2: a new neuroendocrine cell marker. American Journal of Pathology 2000;157:1299–309.CrossRefGoogle ScholarPubMed
McKeehan, WL, Wang, F, Kan, M. The heparan sulfate-fibroblast growth factor family: diversity of structure and function. Progress in Nucleic Acid Research and Molecular Biology 1998;59:135–76.CrossRef
Foster, BA, Kaplan, PJ, Greenberg, NM. Peptide growth factors and prostate cancer: new models, new opportunities. Cancer Metastasis Reviews 1998;17:317–24.CrossRef
Culig, Z, Hobisch, A, Cronauer, MV, et al. Regulation of prostatic growth and function by peptide growth factors. The Prostate 1996;28:392–405.3.0.CO;2-C>CrossRef
Byrne, RL, Leung, H, Neal, DE.Peptide growth factors in the prostate as mediators of stromal epithelial interaction. British Journal of Urology 1996;77:627–33.CrossRefGoogle ScholarPubMed
Story, MT.Regulation of prostate growth by fibroblast growth factors. World Journal of Urology 1995;13:297–305.CrossRefGoogle ScholarPubMed
Di Silverio, F, Sciarra, A, Di Nicola, S, Di Chiro, C. Peptide growth factors: clinical and therapeutic strategies. Minerva Urolica e Nefrologica 1997;49:63–72.
Thomson, AA. Role of androgens and fibroblast growth factors in prostatic development. Reproduction (Cambridge, England) 2001;121:187–95.CrossRef
Yan, G, Fukabori, Y, Nikolaropoulos, S, Wang, F, McKeehan, WL. Heparin-binding keratinocyte growth factor is a candidate stromal-to- epithelial-cell andromedin. Molecular Endocrinology (Baltimore, MD) 1992;6:2123–8.
Yan, G, Fukabori, Y, McBride, G, Nikolaropolous, S, McKeehan, WL. Exon switching and activation of stromal and embryonic fibroblast growth factor (FGF)-FGF receptor genes in prostate epithelial cells accompany stromal independence and malignancy. Molecular and Cellular Biology 1993;13:4513–22.CrossRef
Nakano, K, Fukabori, Y, Itoh, N, et al. Androgen-stimulated human prostate epithelial growth mediated by stromal-derived fibroblast growth factor-10. Endocrinology Journal 1999;46:405–13.
Matsubara, A, Kan, M, Feng, S, McKeehan, WL. Inhibition of growth of malignant rat prostate tumor cells by restoration of fibroblast growth factor receptor 2. Cancer Research 1998;58:1509–14.
Lu, W, Luo, Y, Kan, M, McKeehan, WL.Fibroblast growth factor-10. A second candidate stromal to epithelial cell andromedin in prostate. Journal of Biological Chemistry 1999;274:12 827–34.CrossRefGoogle ScholarPubMed
Feng, S, Wang, F, Matsubara, A, Kan, M, McKeehan, WL. Fibroblast growth factor receptor 2 limits and receptor 1 accelerates tumorigenicity of prostate epithelial cells. Cancer Research 1997;57:5369–78.
Faham, S, Linhardt, RJ, Rees, DC. Diversity does make a difference: fibroblast growth factor-heparin interactions. Current Opinion in Structural Biology 1998;8:578–86.CrossRef
Klint, P, Claesson-Welsh, L. Signal transduction by fibroblast growth factor receptors. Frontiers in Bioscience 1999;4:D165–77.
Ornitz, DM. FGFs, heparan sulfate and FGFRs: complex interactions essential for development. Bioessays 2000;22:108–12.3.0.CO;2-M>CrossRef
Powers, CJ, McLeskey, SW, Wellstein, A. Fibroblast growth factors, their receptors and signaling. Endocrine-Related Cancer 2000;7:165–97.CrossRef
Liu, C, Dib-Hajj, SD, Waxman, SG.Fibroblast growth factor homologous factor 1B binds to the C terminus of the tetrodotoxin-resistant sodium channel rNav1.9a (NaN). Journal of Biological Chemistry 2001;276:18 925–33.CrossRefGoogle Scholar
Olsen, SK, Garbi, M, Zampieri, N, et al. Fibroblast growth factor (FGF) homologous factors share structural but not functional homology with FGFs. Journal of Biological Chemistry 2003;278:34 226–36.CrossRefGoogle Scholar
Itoh, N, Ornitz, DM. Functional evolutionary history of the mouse Fgf gene family. Developmental Dynamics 2008;237:18–27.CrossRef
Goldfarb, M, Schoorlemmer, J, Williams, A, et al. Fibroblast growth factor homologous factors control neuronal excitability through modulation of voltage-gated sodium channels. Neuron 2007;55:449–63.CrossRef
Laezza, F, Lampert, A, Kozel, MA, et al. FGF14 N-terminal splice variants differentially modulate Nav1.2 and Nav1.6-encoded sodium channels. Molecular and Cellular Neuroscience 2009;42:90–101.
Bernfield, M, Gotte, M, Park, PW, et al. Functions of cell surface heparan sulfate proteoglycans. Annual Review of Biochemistry 1999;68:729–77.CrossRef
Esko, JD, Lindahl, U.Molecular diversity of heparan sulfate. Journal of Clinical Investigation 2001;108:169–73.CrossRefGoogle ScholarPubMed
Park, PW, Reizes, O, Bernfield, M.Cell surface heparan sulfate proteoglycans: selective regulators of ligand-receptor encounters. Journal of Biological Chemistry 2000;275:29 923–6.CrossRefGoogle ScholarPubMed
Ye, S, Luo, Y, Lu, W, et al. Structural basis for interaction of FGF-1, FGF-2, and FGF-7 with different heparan sulfate motifs. Biochemistry 2001;40:14 429–39.
Kan, M, Wu, X, Wang, F, McKeehan, WL.Specificity for fibroblast growth factors determined by heparan sulfate in a binary complex with the receptor kinase. Journal of Biological Chemistry 1999;274:15 947–52.CrossRefGoogle Scholar
Kreuger, J, Salmivirta, M, Sturiale, L, Gimenez-Gallego, G, Lindahl, U.Sequence analysis of heparan sulfate epitopes with graded affinities for fibroblast growth factors 1 and 2. Journal of Biological Chemistry 2001;276:30 744–52.CrossRefGoogle ScholarPubMed
Guimond, SE, Turnbull, JE. Fibroblast growth factor receptor signalling is dictated by specific heparan sulphate saccharides. Current Biology 1999;9:1343–6.CrossRef
Ostrovsky, O, Berman, B, Gallagher, J, et al. Differential effects of heparin saccharides on the formation of specific fibroblast growth factor (FGF) and FGF receptor complexes. Journal of Biological Chemistry 2002;277:2444–53.CrossRefGoogle ScholarPubMed
Powell, AK, Fernig, DG, Turnbull, JE.Fibroblast growth factor receptors 1 and 2 interact differently with heparin/heparan sulfate. Implications for dynamic assembly of a ternary signaling complex. Journal of Biological Chemistry 2002;277:28 554–63.CrossRefGoogle ScholarPubMed
Luo, Y, Ye, S, Kan, M, McKeehan, WL.Control of fibroblast growth factor (FGF) 7- and FGF1-induced mitogenesis and downstream signaling by distinct heparin octasaccharide motifs. Journal of Biological Chemistry 2006;281:21 052–61.CrossRefGoogle ScholarPubMed
Luo, Y, Ye, S, Kan, M, McKeehan, WL.Structural specificity in a FGF7-affinity purified heparin octasaccharide required for formation of a complex with FGF7 and FGFR2IIIb. Journal of Cellular Biochemistry 2006;97:1241–58.CrossRefGoogle Scholar
Uematsu, F, Kan, M, Wang, F, et al. Ligand binding properties of binary complexes of heparin and immunoglobulin-like modules of FGF receptor 2. Biochemical and Biophysical Research Communications 2000;272:830–6.CrossRef
Burgar, HR, Burns, HD, Elsden, JL, Lalioti, MD, Heath, JK.Association of the signaling adaptor FRS2 with fibroblast growth factor receptor 1 (Fgfr1) is mediated by alternative splicing of the juxtamembrane domain. Journal of Biological Chemistry 2002;277:4018–23.CrossRefGoogle ScholarPubMed
Paterno, GD, Ryan, PJ, Kao, KR, Gillespie, LL. The VT+ and VT− isoforms of the fibroblast growth factor receptor type 1 are differentially expressed in the presumptive mesoderm of Xenopus embryos and differ in their ability to mediate mesoderm formation. Journal of Biological Chemistry 2000;275:9581–6.CrossRef
Zhang, X, Ibrahimi, OA, Olsen, SK, et al. Receptor specificity of the fibroblast growth factor family: the complete mammalian FGF family. Journal of Biological Chemistry 2006;281:15 694–700.CrossRefGoogle ScholarPubMed
Luo, Y, Lu, W, Mohamedali, KA, et al. The glycine box: a determinant of specificity for fibroblast growth factor. Biochemistry 1998;37:16 506–15.
Carstens, RP, Eaton, JV, Krigman, HR, Walther, PJ, Garcia-Blanco, MA. Alternative splicing of fibroblast growth factor receptor 2 (FGF-R2) in human prostate cancer. Oncogene 1997;15:3059–65.CrossRef
Carstens, RP, McKeehan, WL, Garcia-Blanco, MA. An intronic sequence element mediates both activation and repression of rat fibroblast growth factor receptor 2 pre-mRNA splicing. Molecular and Cellular Biology 1998;18:2205–17.CrossRef
Carstens, RP, Wagner, EJ, Garcia-Blanco, MA. An intronic splicing silencer causes skipping of the IIIb exon of fibroblast growth factor receptor 2 through involvement of polypyrimidine tract binding protein. Molecular and Cellular Biology 2000;20:7388–400.CrossRef
Jones, RB, Wang, F, Luo, Y, et al. The nonsense-mediated decay pathway and mutually exclusive expression of alternatively spliced FGFR2IIIb and -IIIc mRNAs. Journal of Biological Chemistry 2001;276:4158–67.CrossRefGoogle ScholarPubMed
Jones, RB, Carstens, RP, Luo, Y, McKeehan, WL. 5ʹ- and 3ʹ-terminal nucleotides in the FGFR2 ISAR splicing element core have overlapping roles in exon IIIb activation and exon IIIc repression. Nucleic Acids Research 2001;29:3557–65.CrossRef
Ricol, D, Cappellen, D, El Marjou, A, et al. Tumour suppressive properties of fibroblast growth factor receptor 2-IIIb in human bladder cancer. Oncogene 1999;18:7234–43.CrossRef
Wang, F, McKeehan, K, Yu, C, Ittmann, M, McKeehan, WL. Chronic activity of ectopic type 1 fibroblast growth factor receptor tyrosine kinase in prostate epithelium results in hyperplasia accompanied by intraepithelial neoplasia. The Prostate 2004;58:1–12.CrossRef
Jin, C, McKeehan, K, Guo, W, et al. Cooperation between ectopic FGFR1 and depression of FGFR2 in induction of prostatic intraepithelial neoplasia in the mouse prostate. Cancer Research 2003;63:8784–90.
Acevedo, VD, Gangula, RD, Freeman, KW, et al. Inducible FGFR-1 activation leads to irreversible prostate adenocarcinoma and an epithelial-to-mesenchymal transition. Cancer Cell 2007;12:559–71.CrossRef
Freeman, KW, Gangula, RD, Welm, BE, et al. Conditional activation of fibroblast growth factor receptor (FGFR) 1, but not FGFR2, in prostate cancer cells leads to increased osteopontin induction, extracellular signal-regulated kinase activation, and in vivo proliferation. Cancer Research 2003;63:6237–43.
Winter, SF, Acevedo, VD, Gangula, RD, et al. Conditional activation of FGFR1 in the prostate epithelium induces angiogenesis with concomitant differential regulation of Ang-1 and Ang-2. Oncogene 2007;26:4897–907.CrossRef
Memarzadeh, S, Xin, L, Mulholland, DJ, et al. Enhanced paracrine FGF10 expression promotes formation of multifocal prostate adenocarcinoma and an increase in epithelial androgen receptor. Cancer Cell 2007;12:572–85.CrossRef
McIntosh, I, Bellus, GA, Jab, EW. The pleiotropic effects of fibroblast growth factor receptors in mammalian development. Cell Structure and Function 2000;25:85–96.CrossRef
Xu, X, Weinstein, M, Li, C, Deng, C. Fibroblast growth factor receptors (FGFRs) and their roles in limb development. Cell Tissue Research 1999;296:33–43.CrossRef
Kannan, K, Givol, D. FGF receptor mutations: dimerization syndromes, cell growth suppression, and animal models. IUBMB Life 2000;49:197–205.CrossRef
Kato, S, Sekine, K. FGF-FGFR signaling in vertebrate organogenesis. Cellular and Molecular Biology (Noisy-le-grand) 1999;45:631–8.
Burke, D, Wilkes, D, Blundell, TL, Malcolm, S. Fibroblast growth factor receptors: lessons from the genes. Trends in Biochemical Sciences 1998;23:59–62.CrossRef
Webster, MK, Donoghue, DJ. FGFR activation in skeletal disorders: too much of a good thing. Trends in Genetics 1997;13:178–82.CrossRef
Muenke, M, Schell, U. Fibroblast-growth-factor receptor mutations in human skeletal disorders. Trends in Genetics 1995;11:308–13.CrossRef
Kwabi-Addo, B, Ropiquet, F, Giri, D, Ittmann, M. Alternative splicing of fibroblast growth factor receptors in human prostate cancer. The Prostate 2001;46:163–72.3.0.CO;2-T>CrossRef
Sahadevan, K, Darby, S, Leung, H, et al. Selective over-expression of fibroblast growth factor receptors 1 and 4 in clinical prostate cancer. Journal of Pathology 2007;213:82–90.CrossRefGoogle ScholarPubMed
Gowardhan, B, Douglas, DA, Mathers, ME, et al. Evaluation of the fibroblast growth factor system as a potential target for therapy in human prostate cancer. British Journal of Cancer 2005;92:320–7.CrossRefGoogle ScholarPubMed
Pirtskhalaishvili, G, Nelson, JB. Endothelium-derived factors as paracrine mediators of prostate cancer progression. The Prostate 2000;44: 77–87.3.0.CO;2-G>CrossRef
Yang, J, Meyer, M, Muller, AK, et al. Fibroblast growth factor receptors 1 and 2 in keratinocytes control the epidermal barrier and cutaneous homeostasis. Journal of Cell Biology 2010;188:935–52.CrossRefGoogle ScholarPubMed
Donjacour, AA, Thomson, AA, Cunha, GR. FGF-10 plays an essential role in the growth of the fetal prostate. Developmental Biology 2003;261:39–54.CrossRef
Ozen, M, Giri, D, Ropiquet, F, Mansukhani, A, Ittmann, M.Role of fibroblast growth factor receptor signaling in prostate cancer cell survival. Journal of the National Cancer Institute 2001;93:1783–90.CrossRefGoogle ScholarPubMed
Boget, S, Cereser, C, Parvaz, P, Leriche, A, Revol, A.Fibroblast growth factor receptor 1 (FGFR1) is over-expressed in benign prostatic hyperplasia whereas FGFR2-IIIc and FGFR3 are not. European Journal of Endocrinology 2001;145:303–10.CrossRefGoogle ScholarPubMed
Sibley, K, Stern, P, Knowles, MA. Frequency of fibroblast growth factor receptor 3 mutations in sporadic tumours. Oncogene 2001;20:4416–18.CrossRef
Ropiquet, F, Giri, D, Kwabi-Addo, B, Mansukhani, A, Ittmann, M. Increased expression of fibroblast growth factor 6 in human prostatic intraepithelial neoplasia and prostate cancer. Cancer Research 2000;60:4245–50.
Davol, PA, Frackelton, AR Targeting human prostatic carcinoma through basic fibroblast growth factor receptors in an animal model: characterizing and circumventing mechanisms of tumor resistance. The Prostate 1999;40:178–91.3.0.CO;2-I>CrossRef
Saez, C, Gonzalez-Baena, AC, Japon, MA, et al. Expression of basic fibroblast growth factor and its receptors FGFR1 and FGFR2 in human benign prostatic hyperplasia treated with finasteride. The Prostate 1999;40:83–8.3.0.CO;2-N>CrossRef
Giri, D, Ropiquet, F, Ittmann, M.FGF9 is an autocrine and paracrine prostatic growth factor expressed by prostatic stromal cells. Journal of Cell Physiology 1999;180:53–60.3.0.CO;2-P>CrossRefGoogle ScholarPubMed
Fidler, I. J, Kumar, R, Bielenberg, DR, Ellis, LM. Molecular determinants of angiogenesis in cancer metastasis. The Cancer Journal from Scientific American 1998;4 Suppl 1:S58–66.
Ropiquet, F, Giri, D, Lamb, DJ, Ittmann, M.FGF7 and FGF2 are increased in benign prostatic hyperplasia and are associated with increased proliferation. Journal of Urology 1999;162:595–9.CrossRefGoogle ScholarPubMed
Dorkin, TJ, Robinson, MC, Marsh, C, et al. FGF8 over-expression in prostate cancer is associated with decreased patient survival and persists in androgen independent disease. Oncogene 1999;18:2755–61.CrossRef
Wang, Q, Stamp, GW, Powell, S, et al. Correlation between androgen receptor expression and FGF8 mRNA levels in patients with prostate cancer and benign prostatic hypertrophy. Journal of Clinical Pathology 1999;52:29–34.CrossRefGoogle ScholarPubMed
Gnanapragasam, VJ, Robson, CN, Neal, DE, Leung HY. Regulation of FGF8 expression by the androgen receptor in human prostate cancer. Oncogene 2002;21:5069–80.CrossRef
Song, Z, Wu, X, Powell, WC, et al. Fibroblast growth factor 8 isoform B overexpression in prostate epithelium: a new mouse model for prostatic intraepithelial neoplasia. Cancer Research 2002;62: 5096–105.
Polnaszek, N, Kwabi-Addo, B, Wang, J, Ittmann, M. FGF17 is an autocrine prostatic epithelial growth factor and is upregulated in benign prostatic hyperplasia. The Prostate 2004;60:18–24.CrossRef
Valve, EM, Nevalainen, MT, Nurmi, MJ, et al. Increased expression of FGF-8 isoforms and FGF receptors in human premalignant prostatic intraepithelial neoplasia lesions and prostate cancer. Laboratory Investigation 2001;81:815–26.CrossRef
Valve, E, Martikainen, P, Seppanen, J, et al. Expression of fibroblast growth factor (FGF)-8 isoforms and FGF receptors in human ovarian tumors. International Journal of Cancer 2000;88:718–25.3.0.CO;2-F>CrossRefGoogle ScholarPubMed
Tanaka, A, Furuya, A, Yamasaki, M, et al. High frequency of fibroblast growth factor (FGF) 8 expression in clinical prostate cancers and breast tissues, immunohistochemically demonstrated by a newly established neutralizing monoclonal antibody against FGF 8. Cancer Research 1998;58:2053–6.
Schmitt, JF, Hearn, MT, Risbridger, GP.Expression of fibroblast growth factor-8 in adult rat tissues and human prostate carcinoma cells. Journal of Steroid Biochemistry and Molecular Biology 1996;57:173–8.CrossRefGoogle ScholarPubMed
Leung, HY, Dickson, C, Robson, CN, Neal DE. Over-expression of fibroblast growth factor-8 in human prostate cancer. Oncogene 1996;12:1833–5.
Saric, T, Shain, SA. Androgen regulation of prostate cancer cell FGF-1, FGF-2, and FGF-8: preferential down-regulation of FGF-2 transcripts. Growth Factors 1998;16:69–87.CrossRef
Luo, Y, Yang, C, Jin, C, et al. Novel phosphotyrosine targets of FGFR2IIIb signaling. Cellular Signalling 2009;21:1370–8.CrossRef
Xian, W, Schwertfeger, KL, Rosen, JM. Distinct roles of fibroblast growth factor receptor 1 and 2 in regulating cell survival and epithelial-mesenchymal transition. Molecular Endocrinology (Baltimore, MD) 2007;21:987–1000.CrossRef
Shi, E, Kan, M, Xu, J, et al. Control of fibroblast growth factor receptor kinase signal transduction by heterodimerization of combinatorial splice variants. Molecular and Cellular Biology 1993;13(7):3907–18.CrossRef
Hou, J, McKeehan, K, Kan, M, et al. Identification of tyrosines 154 and 307 in the extracellular domain and 653 and 766 in the intracellular domain as phosphorylation sites in the heparin-binding fibroblast growth factor receptor tyrosine kinase (flg). Protein Science 1993;2:86–92.
Pellegrini, L, Burke, DF, von Delft, F, Mulloy, B, Blundell, TL. Crystal structure of fibroblast growth factor receptor ectodomain bound to ligand and heparin. Nature 2000;407:1029–34.CrossRef
Plotnikov, AN, Hubbard, SR, Schlessinger, J, Mohammadi, M. Crystal structures of two FGF-FGFR complexes reveal the determinants of ligand-receptor specificity. Cell 2000;101:413–24.CrossRef
Schlessinger, J, Plotnikov, AN, Ibrahimi, OA, et al. Crystal structure of a ternary FGF-FGFR-heparin complex reveals a dual role for heparin in FGFR binding and dimerization. Molecular Cell 2000;6:743–50.CrossRef
Mohammadi, M, Dikic, I, Sorokin, A, et al. Identification of six novel autophosphorylation sites on fibroblast growth factor receptor 1 and elucidation of their importance in receptor activation and signal transduction. Molecular and Cellular Biology 1996;16:977–89.CrossRef
Lew, ED, Furdui, CM, Anderson, KS, Schlessinger, J. The precise sequence of FGF receptor autophosphorylation is kinetically driven and is disrupted by oncogenic mutations. Science Signaling 2009;2: ra6.
Mohammadi, M, Dionne, CA, Li, W, et al. Point mutation in FGF receptor eliminates phosphatidylinositol hydrolysis without affecting mitogenesis. Nature 1992;358:681–4.CrossRef
Peters, KG, Marie, J, Wilson, E, et al. Point mutation of an FGF receptor abolishes phosphatidylinositol turnover and Ca2+ flux but not mitogenesis. Nature 1992;358:678–81.CrossRef
Larsson, H, Klint, P, Landgren, E, Claesson-Welsh, L. Fibroblast growth factor receptor-1-mediated endothelial cell proliferation is dependent on the Src homology (SH) 2/SH3 domain-containing adaptor protein Crk. Journal of Biological Chemistry 1999;274:25 726–34.CrossRefGoogle ScholarPubMed
Klint, P, Claesson-Welsh, L. Signal transduction by fibroblast growth factor receptors. Frontiers in Bioscience 1999;4:D165–77.
Seo, JH, Suenaga, A, Hatakeyama, M, Taiji, M, Imamoto, A. Structural and functional basis of a role for CRKL in a fibroblast growth factor 8-induced feed-forward loop. Molecular and Cellular Biology 2009;29:3076–87.CrossRef
Hinsby, AM, Lundfald, L, Ditlevsen, DK, et al. ShcA regulates neurite outgrowth stimulated by neural cell adhesion molecule but not by fibroblast growth factor 2: evidence for a distinct fibroblast growth factor receptor response to neural cell adhesion molecule activation. Journal of Neurochemistry 2004;91:694–703.CrossRefGoogle Scholar
Guy, GR, Wong, ES, Yusoff, P, et al. Sprouty: how does the branch manager work?Journal of Cell Science 2003;116(Pt 15):3061–8.CrossRefGoogle ScholarPubMed
Greenberg, NM, DeMayo, FJ, Sheppard, PC, et al. The rat probasin gene promoter directs hormonally and developmentally regulated expression of a heterologous gene specifically to the prostate in transgenic mice. Molecular Endocrinology (Baltimore, MD) 1994;8:230–9.
Zhang, J, Gao, N, Kasper, S, et al. An androgen-dependent upstream enhancer is essential for high levels of probasin gene expression. Endocrinology 2004;145:134–48.CrossRef
Zhang, J, Thomas, TZ, Kasper, S, Matusik, RJ. A small composite probasin promoter confers high levels of prostate-specific gene expression through regulation by androgens and glucocorticoids in vitro and in vivo. Endocrinology 2000;141:4698–710.CrossRef
Capecchi, MR. Targeted gene replacement. Scientific American 1994;270:52–9.CrossRef
Sternberg, N, Hamilton, D.Bacteriophage P1 site-specific recombination. I. Recombination between loxP sites. Journal of Molecular Biology 1981;150:467–86.CrossRefGoogle ScholarPubMed
Golic, KG, Lindquist, S. The FLP recombinase of yeast catalyzes site-specific recombination in the Drosophila genome. Cell 1989;59:499–509.CrossRef
Sadowski, PD.The Flp recombinase of the 2-microns plasmid ofSaccharomyces cerevisiae. Progress in Nucleic Acid Research and Molecular Biology 1995;51:53–91.Google Scholar
St-Onge, L, Furth, PA, Gruss, P. Temporal control of the Cre recombinase in transgenic mice by a tetracycline responsive promoter. Nucleic Acids Research 1996;24:3875–7.CrossRef
Hayashi, S, McMahon, AP. Efficient recombination in diverse tissues by a tamoxifen-inducible form of Cre: a tool for temporally regulated gene activation/inactivation in the mouse. Developmental Biology 2002;244:305–18.CrossRef
Song, Z, Wu, X, Powell, WC, et al. Fibroblast growth factor 8 isoform b overexpression in prostate epithelium: a new mouse model for prostatic intraepithelial neoplasia. Cancer Research 2002;62:5096–105.
Zhong, C, Saribekyan, G, Liao, CP, Cohen, MB, Roy-Burman, P. Cooperation between FGF8b overexpression and PTEN deficiency in prostate tumorigenesis. Cancer Research 2006;66:2188–94.CrossRef
Foster, BA, Evangelou, A, Gingrich, JR, et al. Enforced expression of FGF-7 promotes epithelial hyperplasia whereas a dominant negative FGFR2iiib promotes the emergence of neuroendocrine phenotype in prostate glands of transgenic mice. Differentiation 2002;70:624–32.CrossRef
Chua, SS, Ma, ZQ, Gong, L, et al. Ectopic expression of FGF-3 results in abnormal prostate and Wolffian duct development. Oncogene 2002;21:1899–908.CrossRef
Konno-Takahashi, N, Takeuchi, T, Nishimatsu, H, et al. Engineered FGF-2 expression induces glandular epithelial hyperplasia in the murine prostatic dorsal lobe. European Urology 2004;46:126–32.CrossRef
Wang, F, McKeehan, K, Yu, C, McKeehan, WL. Fibroblast growth factor receptor 1 phosphotyrosine 766: molecular target for prevention of progression of prostate tumors to malignancy. Cancer Research 2002;62:1898–903.
Freeman, KW, Welm, BE, Gangula, RD, et al. Inducible prostate intraepithelial neoplasia with reversible hyperplasia in conditional FGFR1-expressing mice. Cancer Research 2003;63:8256–63.
Gingrich, JR, Barrios, RJ, Morton, RA, et al. Metastatic prostate cancer in a transgenic mouse. Cancer Research 1996;56:4096–102.
Skalnik, DG, Dorfman, DM, Williams, DA, Orkin, SH. Restriction of neuroblastoma to the prostate gland in transgenic mice. Molecular and Cellular Biology 1991;11:4518–27.CrossRef
Greenberg, NM, DeMayo, F, Finegold, MJ, et al. Prostate cancer in a transgenic mouse. Proceedings of the National Academy of Sciences USA 1995;92:3439–43.CrossRef
Masumori, N, Thomas, TZ, Chaurand, P, et al. A probasin-large T antigen transgenic mouse line develops prostate adenocarcinoma and neuroendocrine carcinoma with metastatic potential. Cancer Research 2001;61:2239–49.
Kasper, S, Sheppard, PC, Yan, Y, et al. Development, progression, and androgen-dependence of prostate tumors in probasin-large T antigen transgenic mice: a model for prostate cancer. Laboratory Investigation 1998;78:i–xv.
Giri, D, Ropiquet, F, Ittmann, M. Alterations in expression of basic fibroblast growth factor (FGF) 2 and its receptor FGFR-1 in human prostate cancer. Clinical Cancer Research 1999;5:1063–71.
Polnaszek, N, Kwabi-Addo, B, Peterson, LE, et al. Fibroblast growth factor 2 promotes tumor progression in an autochthonous mouse model of prostate cancer. Cancer Research 2003;63:5754–60.
Gotoh, N. Regulation of growth factor signaling by FRS2 family docking/scaffold adaptor proteins. Cancer Science 2008;99:1319–25.CrossRef
Xu, H, Lee, KW, Goldfarb, M.Novel recognition motif on fibroblast growth factor receptor mediates direct association and activation of SNT adapter proteins. Journal of Biological Chemistry 1998;273:17 987–90.CrossRefGoogle ScholarPubMed
Kouhara, H, Hadari, YR, Spivak-Kroizman, T, et al. A lipid-anchored Grb2-binding protein that links FGF-receptor activation to the Ras/MAPK signaling pathway. Cell 1997;89:693–702.CrossRef
Ong, SH, Goh, KC, Lim, YP, et al. Suc1-associated neurotrophic factor target (SNT) protein is a major FGF- stimulated tyrosine phosphorylated 90-kDa protein which binds to the SH2 domain of GRB2. Biochemical and Biophysical Research Communications 1996;225:1021–6.CrossRef
Ong, SH, Lim, YP, Low, BC, Guy, GR. SHP2 associates directly with tyrosine phosphorylated p90 (SNT) protein in FGF-stimulated cells. Biochemical and Biophysical Research Communications 1997;238: 261–6.CrossRef
Rabin, SJ, Cleghon, V, Kaplan, DR. SNT, a differentiation-specific target of neurotrophic factor-induced tyrosine kinase activity in neurons and PC12 cells. Molecular and Cellular Biology 1993;13:2203–13.CrossRef
Gotoh, N, Ito, M, Yamamoto, S, et al. Tyrosine phosphorylation sites on FRS2alpha responsible for Shp2 recruitment are critical for induction of lens and retina. Proceedings of the National Academy of Sciences USA 2004;101:17 144–9.
McDougall, K, Kubu, C, Verdi, JM, Meakin, SO. Developmental expression patterns of the signaling adapters FRS-2 and FRS-3 during early embryogenesis. Mechanisms of Development 2001;103:145–8.CrossRef
Hadari, YR, Gotoh, N, Kouhara, H, Lax, I, Schlessinger, J. Critical role for the docking-protein FRS2 alpha in FGF receptor-mediated signal transduction pathways. Proceedings of the National Academy of Sciences USA 2001;98:8578–83.CrossRef
Lin, Y, Liu, G, Zhang, Y, 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;134:723–34.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • The FGF signaling axis in prostate tumorigenesis
    • By Fen Wang, Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX,USA, Yongde Luo, Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA, Wallace L. McKeehan, Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.017
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • The FGF signaling axis in prostate tumorigenesis
    • By Fen Wang, Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX,USA, Yongde Luo, Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA, Wallace L. McKeehan, Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.017
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • The FGF signaling axis in prostate tumorigenesis
    • By Fen Wang, Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX,USA, Yongde Luo, Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA, Wallace L. McKeehan, Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, TX, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.017
Available formats
×