Get 20M+ Full-Text Papers For Less Than $1.50/day. Start a 14-Day Trial for You or Your Team.

Learn More →

A Network Map of FGF-1/FGFR Signaling System

A Network Map of FGF-1/FGFR Signaling System A Network Map of FGF-1/FGFR Signaling System div.banner_title_bkg div.trangle { border-color: #2D4917 transparent transparent transparent; opacity:0.8; /*new styles start*/ -ms-filter:"progid:DXImageTransform.Microsoft.Alpha(Opacity=80)" ;filter: alpha(opacity=80); /*new styles end*/ } div.banner_title_bkg_if div.trangle { border-color: transparent transparent #2D4917 transparent ; opacity:0.8; /*new styles start*/ -ms-filter:"progid:DXImageTransform.Microsoft.Alpha(Opacity=80)" ;filter: alpha(opacity=80); /*new styles end*/ } div.banner_title_bkg div.trangle { width: 292px; } #banner { background-image: url('http://images.hindawi.com/journals/jst/jst.banner.jpg'); background-position: 50% 0;} Hindawi Publishing Corporation Home Journals About Us Journal of Signal Transduction About this Journal Submit a Manuscript Table of Contents Journal Menu About this Journal · Abstracting and Indexing · Aims and Scope · Article Processing Charges · Articles in Press · Author Guidelines · Bibliographic Information · Citations to this Journal · Contact Information · Editorial Board · Editorial Workflow · Free eTOC Alerts · Publication Ethics · Reviewers Acknowledgment · Submit a Manuscript · Subscription Information · Table of Contents Open Special Issues · Published Special Issues · Special Issue Guidelines Abstract Full-Text PDF Full-Text HTML Full-Text ePUB Linked References How to Cite this Article Journal of Signal Transduction Volume 2014 (2014), Article ID 962962, 16 pages http://dx.doi.org/10.1155/2014/962962 Review Article A Network Map of FGF-1/FGFR Signaling System Rajesh Raju , 1 Shyam Mohan Palapetta , 1,2 Varot K. Sandhya , 1 Apeksha Sahu , 1,2 Abbas Alipoor , 3 Lavanya Balakrishnan , 1 Jayshree Advani , 1 Bijesh George , 1 K. Ramachandra Kini , 3 N. P. Geetha , 3 H. S. Prakash , 3 T. S. Keshava Prasad , 1 Yu-Jung Chang , 4 Linyi Chen , 4 Akhilesh Pandey , 5,6,7,8 and Harsha Gowda 1 1 Institute of Bioinformatics, International Tech Park, Bangalore 560066, India 2 Centre of Excellence in Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry 605014, India 3 Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysore 570006, India 4 Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan 5 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA 6 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA 7 Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA 8 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Received 20 November 2013; Accepted 3 March 2014; Published 16 April 2014 Academic Editor: Shoukat Dedhar Copyright © 2014 Rajesh Raju et al. This is an open access article distributed under the Creative Commons Attribution License , which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Fibroblast growth factor-1 (FGF-1) is a well characterized growth factor among the 22 members of the FGF superfamily in humans. It binds to all the four known FGF receptors and regulates a plethora of functions including cell growth, proliferation, migration, differentiation, and survival in different cell types. FGF-1 is involved in the regulation of diverse physiological processes such as development, angiogenesis, wound healing, adipogenesis, and neurogenesis. Deregulation of FGF-1 signaling is not only implicated in tumorigenesis but also is associated with tumor invasion and metastasis. Given the biomedical significance of FGFs and the fact that individual FGFs have different roles in diverse physiological processes, the analysis of signaling pathways induced by the binding of specific FGFs to their cognate receptors demands more focused efforts. Currently, there are no resources in the public domain that facilitate the analysis of signaling pathways induced by individual FGFs in the FGF/FGFR signaling system. Towards this, we have developed a resource of signaling reactions triggered by FGF-1/FGFR system in various cell types/tissues. The pathway data and the reaction map are made available for download in different community standard data exchange formats through NetPath and NetSlim signaling pathway resources. 1. Introduction Fibroblast growth factor (FGF) superfamily consists of structurally related polypeptides most of which function through its high affinity fibroblast growth factor receptors (FGFRs). In addition to FGFRs, they also bind to heparan sulfate proteoglycans (HPSGs) and their analog, heparin. These interactions influence the stability of FGFs in the extracellular matrix and also regulate their binding and activation of FGFRs [ 1 – 9 ]. In humans, FGFs are encoded by 22 genes, FGF-1-14 and FGF-16-23, and are divided into 7 subfamilies. FGFs 1–10 and 16–23 are FGFR ligands, while FGFs 11–14 are intracellular FGF homologous factors which act in a receptor-independent fashion [ 10 ]. Knock-out mice of different FGFs exhibit diverse developmental and physiological disorders [ 11 ]. For instance, FGF-9 is involved in the development of lung and testes [ 12 , 13 ], FGF-3 is critical for inner ear development [ 14 ], and FGF-18 is important in bone and lung development [ 15 – 17 ]. Moreover, knock-out of FGFs 4, 8, 9, 10, 15, 18, or 23 was found to be lethal in mice [ 18 ]. FGFs are also involved in wound healing, tissue repair [ 19 , 20 ], and angiogenesis [ 21 ]. Facilitating cell proliferation, migration, and differentiation [ 16 , 22 – 26 ], FGFs are implicated in diverse pathological conditions including cancer [ 27 ] as well as metabolic and developmental disorders [ 18 ]. Most FGFs have an N-terminal signal peptide and are thus secreted. FGFs 1, 2, 9, 16, and 20 do not have signal peptides. FGFs 9, 16, and 20 may be released through classical secretory pathway; however, FGF-1 and FGF-2 are released from damaged cells or through endoplasmic reticulum-golgi independent exocytotic pathway [ 10 ]. FGF-1 along with FGF-2 was initially isolated from bovine pituitary extracts based on their ability to induce proliferation in 3T3 fibroblasts [ 28 , 29 ]. Also known as acidic FGF, FGF-1 is a 155 amino acid long non-glycosylated polypeptide. FGF-1 is not released from the cells under normal physiological conditions, but it was secreted in response to stress conditions such as heat shock, hypoxia [ 30 , 31 ], serum starvation [ 32 ], and exposure to low-density lipoproteins [ 33 ]. Stress induces the release of inactive disulfide bond-linked homodimeric form of FGF-1, which is dependent on p40-Syt1, S100A13, and Cu 2+ ions [ 34 – 37 ]. FGF-1 has been shown to reduce apoptosis in vascular injury [ 38 – 40 ]. Administration of FGF-1 has shown promise as a therapeutic strategy against human cervical spinal cord injury [ 41 ] and ischemic conditions [ 42 – 44 ]. Increased expression of FGF-1 was observed in ovarian [ 45 ] and prostate cancers [ 46 ]. Taken together, FGF1 is involved in different cellular functions that are mediated through its interaction with the four FGF receptors [ 47 , 48 ]. A pathway resource representing these diverse functions and the underlying mechanisms that regulate these processes would be immensely useful. Curated pathway maps are invaluable resources for scientific community. Such comprehensive pathway datasets are being increasingly used in bioinformatics efforts directed towards analysis of high-throughput datasets from various disease contexts. Repositories including Pathway Interaction Database of the National Cancer Institute ( http://pid.nci.nih.gov/ ), Database of Cell Signaling ( http://stke.sciencemag.org/cm/ ), KEGG Pathway Database ( http://www.genome.jp/kegg/pathway.html ), and INOH Pathway Database ( http://inoh.org/ ) have cataloged basic components of FGF signaling. We have expanded the scope of this by providing a comprehensive representation of FGF1 signaling pathway and its diverse roles in regulating various cellular processes. 2. Methodology Documentation of specific pathway reactions scattered in the literature into an organized, user-friendly, query-enabled platform is primary to the analysis of signaling pathways. We used NCBI PubMed database to carry out an extensive literature search to retrieve research articles where molecular events triggered by the FGF-1/FGFR signaling system were studied. Specific molecular events screened include (a) physical associations between proteins, (b) posttranslational modifications (PTMs), (c) change in subcellular localization of proteins, (d) activation or inhibition of specific proteins, and (e) regulation of gene expression. Relevant information from research articles were manually documented using the curation tool, PathBuilder. To streamline and organize data collection from literature, we followed the previously described criteria for the inclusion/exclusion of pathway specific reactions [ 49 , 50 ]. The data accumulated was submitted to the NetPath signaling pathway resource developed by our group [ 51 ]. We then generated a signaling map for this pathway using PathVisio pathway visualization software. We also applied additional criteria to filter out low confidence reactions from the gathered data [ 52 ] and generated a NetSlim map. In addition to curation of molecular level information, we have also cataloged physiological effects brought about by FGF-1 in different cell types/tissues. 3. Results and Discussion Canonical FGF/FGFR signaling reactions have been documented in a few public repositories and review articles. Vast amount of literature in the last few years have revealed several novel pathway intermediates of FGF/FGFR signaling system. In order to generate a comprehensive view of FGF/FGFR signaling pathway, we carried out extensive literature search on PubMed for articles pertaining to FGF-1 signaling. Of a total of 3275 articles that were screened, 237 of them had molecular reactions reported downstream of FGF-1 in various cell types/tissues. Manual curation from these research articles revealed 109 molecules involved in FGF-1 induced physical associations, modulation by PTMs, activity, and subcellular or cell surface translocation events. Of the 42 physical associations that were cataloged, 29 were “binary” and 13 were “complex” interactions inclusive of the ligand/receptor interactors. We could record a total of 87 catalysis events, 15 activation/inhibition, and 21 translocation events. The 87 catalysis events include 19 events, where the enzymes directly catalyzing the reactions were studied and reported, and 68 events for which the enzymes which post-translationally modified the proteins are not studied under FGF-1 stimulation. Apart from these molecular reactions, we have also cataloged 117 genes whose expression is reported to be either upregulated or downregulated by FGF-1 treatment. However, only a total of 25 genes were reported to be differentially regulated at mRNA level by FGF-1 stimulation in different human cell types. A list of genes reported to be regulated by FGF-1 in different mammalian systems at the mRNA and/or the protein level is provided in Table 1 . After the annotation process, all the entries were reviewed and approved by internal reviewers. Internally reviewed pathways were further reviewed and approved by an external pathway authority (LC, who is an author in this paper). Table 1: List of genes that are reported to be transcriptionally and translationally regulated by FGF-1 in humans and other mammals. 3.1. Signaling Modules Activated by FGF-1 Signaling modules comprise a well-characterized group of molecules and their interactions downstream of activation of a receptor. We documented the following signaling modules to be activated upon stimulation with FGF-1. 3.1.1. Ras/Raf/Mek/Erk Pathway The Ras/Raf/Mek/Erk pathway has been implicated in cellular processes including cell growth, proliferation, and migration. Stimulation of different cell types with FGF-1 resulted in the formation of multiple complexes involving FRS2, GAB1, SOS1, PTPN11, SHC1, SH2B1, and GRB2 [ 53 – 60 ]. These complexes are critical to the subsequent activation of Ras [ 53 , 56 ]. Association of Ras with Raf kinase [ 53 ] induces autophosphorylation and activation of Raf. Activation of Raf leads to phosphorylation dependent activation of Map kinases 1/2 (MAP2K1/2) and subsequently Erk2/1 (MAPK1/3) [ 60 – 62 ]. In the context of FGF-1 signaling, this module was reported to be involved in a number of processes including neurogenesis, adipocyte differentiation, cell proliferation, cholesterogenesis, cardioprotection, and tumor invasion and metastasis [ 62 – 67 ]. 3.1.2. Pi3k/Akt Pathway The complexes mentioned above also lead to the activation of Pi3k/Akt pathway, another signaling module that regulates various processes including cell growth, survival, cell proliferation, and cell migration [ 68 ]. A number of studies have shown FGF-1 induced phosphorylation of Akt [ 63 , 64 , 69 ]. Pi3k inhibitor-based functional assays also proved the involvement of FGF-1 pathway in diverse physiological conditions including angiogenesis [ 70 ], lung development [ 71 ], maintenance of neuronal phenotype [ 72 ], neuroprotection [ 73 ], and ApoE-HDL secretion [ 69 ]. 3.1.3. Jnk and p38 Mapk Pathway The c-jun N-terminal kinase ( Jnk) pathway is implicated in the regulation of cell cycle, cell survival and apoptosis. FGF-1 stimulates the phosphorylation of p38 Mapk (MAPK14) as well as Jnk1/2 (MAPK8/9). The Jnk1/2 was also found to be crucial to neurogenesis and vascular remodeling [ 63 , 74 ]. The specific functions of FGF-1 signaling mediated by p38 Mapk include growth arrest, promotion of apoptosis in response to oxidative stress, and formation of actin stress fibers [ 75 – 77 ]. 3.1.4. STAT3 and Nf-kb Pathway FGF-1 also stimulates STATs (STAT1 and STAT3) and Nf-kB signaling modules. FGFR signaling is reported to be regulated through several downstream molecules including JAK2, SRC, SH2B1, MAPK1/3, MAPK8/9, and STAT3. This signaling axis is known to regulate various cellular processes including neurite outgrowth, cell proliferation, and increased cancer cell invasion [ 78 – 80 ]. In addition, FGF-1 is also reported to induce MMP9 expression in mammary adenocarcinoma cells through the Nf-kb pathway [ 81 ]. 3.2. Physiological Effects Mediated by FGF-1 FGF-1 was found to be involved in a number of biological processes. It is associated with the development of heart [ 82 ], lens [ 83 ], lung, and liver [ 84 – 86 ]. Its crucial roles in neurogenesis as well as adipogenesis [ 65 , 87 , 88 ] have also been reported. FGF-1 induces growth arrest and differentiation in chondrocytes [ 89 – 92 ]. It is implicated in angiogenesis [ 93 – 95 ] and wound healing [ 95 – 99 ]. Multiple studies have also shown the role of FGF-1 in cardioprotection [ 99 – 101 ] and neuroprotection [ 22 , 102 ]. FGF-1 also induces migration [ 103 – 105 ] and proliferation [ 106 – 108 ] in different types of cancer cells. It is also involved in the regulation of epithelial-to-mesenchymal transition [ 109 , 110 ], and tumorigenesis [ 111 ] as well as invasion and metastasis [ 64 , 112 ]. A list of functional effects of FGF-1 studied in different cell types/tissues is provided in Table 2 . Table 2: Functions of FGF-1 identified in diverse cell/tissue types of human and other mammalian origins. 3.3. Pathway Visualization, Data Formats, and Availability User-friendly visualization of pathways is an important aspect to provide a concise view. A number of tools are available for visualization and analysis of pathway data including Cytoscape [ 113 ], ChisioBioPAX Editor (ChiBE) [ 114 ], visualization and layout services for BioPAX pathway models (VISIBIOweb) [ 115 ], and ingenuity pathway analysis. These tools use pathway and molecular interaction data in different XML-based community standard data exchange formats as input. These standard formats, which include Proteomics Standards Initiative for Molecular Interaction (PSI-MI version 2.5), Biological Pathway eXchange (BioPAX level 3), and Systems Biology Markup Language (SBML version 2.1), enable easy data exchange and interoperability with multiple software. We have provided the annotated pathway data in the standard formats mentioned above. This data can be downloaded and used from NetPath [ 51 ], an open source resource for signal transduction pathways developed by our group ( http://www.netpath.org/index.html ). Additionally, we have drawn a map of FGF-1/FGFR signaling using the data accumulated in NetPath. This network map represents the molecules and their reactions organized by topology and excludes the molecules identified through phosphoproteomics approaches for which topology could not be assigned (Figure 1 ). The map was manually drawn using freely available software, PathVisio [ 116 ]. The topology of the molecules and their reactions in the pathway was arranged based on (i) inhibitor-based assays, (ii) mutation-based assays, (iii) knock-out studies, (iv) prior knowledge of canonical modules, and/or (v) with reference to multiple review articles. Another map, which incorporated high confidence reactions in accordance with NetSlim criteria [ 52 ], is submitted to the NetSlim database. These maps can be visualized and downloaded in gpml, GenMAPP, png, and pdf formats from http://www.netpath.org/netslim/FGF-1_pathway.html . Each node in the map is linked to their molecule page in NetPath, thereby to other pathways in NetPath, and to HPRD [ 117 ] and RefSeq protein accessions. In the “map with citation” option, the edges connecting the nodes are linked to the corresponding articles in PubMed that report the FGF-1 stimulated reaction(s). Direct reactions are represented by solid edges. Indirect reactions are represented with dashed edges. The edges which represent the protein-protein interactions, enzyme-substrate reactions and translocation events are distinguished by different colors. Figure 1: Network map of FGF-1 signaling. This map manually drawn using PathVisio [ 112 ] represents the reactions induced by FGF-1 through their receptors. Each node represents the molecules and the post-translationally modified states of proteins are also represented. Distinguished by color and continuous/dashed lines, the edges represent the specific information such as protein-protein interactions, enzyme-substrate reactions, reactions mediated through unknown/multiple steps, and protein translocations as provided in the legend. The biological processes that FGF-1 regulates through multiple signaling modules are also represented. A NetSlim [ 52 ] version of this map can be obtained from http://www.netpath.org/netslim/FGF-1_pathway.html . 4. Conclusions Availability of specific ligand-receptor mediated signaling data in community approved formats is crucial to the understanding of proteins and their reactions in diverse biological processes. Analysis of high-throughput data obtained from microarray- and mass spectrometry-based platforms essentially relies on enrichment of biological function or signaling pathways available in databases to obtain insights into their physiological functions. Although some resources have cataloged FGF signaling in general, this is the first attempt to provide a comprehensive view of FGF-1 signaling. This will be extended to other FGF ligands and/or specific FGFRs in the future to facilitate the analysis of differences between different FGFs and/or FGFRs. The pathway information has been made available through NetPath and NetSlim resources in multiple community standard data formats. The FGF-1 signaling pathway data will be periodically updated in NetPath. We have cataloged multiple signaling modules that are activated upon activation of FGFR and their implications in diverse physiological and pathophysiological processes. We believe that the data presented here will boost further research in this area and will help identify novel therapeutically important molecules that could be targeted in pathological conditions involving aberrant FGF-1 signaling. Abbreviations S100A13: S100 calcium binding protein A13 FRS2: Fibroblast growth factor receptor substrate 2 GAB1: GRB2-associated binding protein 1 SOS1: Son of sevenless homolog 1 PTPN11: Protein tyrosine phosphatase, non-receptor type 11 SHC1: Src homology 2 domain containing transforming protein 1 GRB2: Growth factor receptor-bound protein 2 Mapk: Mitogen activated protein kinase Pi3k: Phosphatidylinositide 3-kinase Akt: v-akt murine thymoma viral oncogene homolog HDL: High density lipoprotein Jnk: Jun N-terminal kinase STAT3: Signal transducer and activator of transcription 3. Conflict of Interests The authors have no conflict of interests. Authors’ Contribution Shyam Mohan Palapetta, Varot K. Sandhya, and Apeksha Sahu contributed equally to the paper. Acknowledgments The authors thank the Department of Biotechnology (DBT), Government of India, for research support to the Institute of Bioinformatics, Bangalore. Shyam Mohan Palapetta is supported by a Senior Research Fellowship from the Council of Scientific and Industrial Research (CSIR), India. Varot K. Sandhya is a recipient of Inspire Fellowship from the Department of Science and Technology (DST), Government of India. Harsha Gowda is a Wellcome Trust/DBT India Alliance Early Career Fellow. References N. J. Harmer, “Insights into the role of heparan sulphate in fibroblast growth factor signalling,” Biochemical Society Transactions , vol. 34, no. 3, pp. 442–445, 2006. View at Publisher · View at Google Scholar · View at Scopus O. A. Ibrahimi, F. Zhang, S. C. L. Hrstka, M. Mohammadi, and R. J. Linhardt, “Kinetic model for FGF, FGFR, and proteoglycan signal transduction complex assembly,” Biochemistry , vol. 43, no. 16, pp. 4724–4730, 2004. View at Publisher · View at Google Scholar · View at Scopus D. M. Ornitz, A. B. Herr, M. Nilsson, J. Westman, C.-M. Svahn, and G. Waksman, “FGF binding and FGF receptor activation by synthetic heparan-derived di- and trisaccharides,” Science , vol. 268, no. 5209, pp. 432–436, 1995. View at Scopus M. W. Pantoliano, “Multivalent ligand-receptor binding interactions in the fibroblast growth factor system produce a cooperative growth factor and heparin mechanism for receptor dimerization,” Biochemistry , vol. 33, no. 34, pp. 10229–10248, 1994. View at Scopus D. M. Ornitz and P. Leder, “Ligand specificity and heparin dependence of fibroblast growth factor receptors 1 and 3,” The Journal of Biological Chemistry , vol. 267, no. 23, pp. 16305–16311, 1992. View at Scopus D. M. Ornitz, A. Yayon, J. G. Flanagan, C. M. Svahn, E. Levi, and P. Leder, “Heparin is required for cell-free binding of basic fibroblast growth factor to a soluble receptor and for mitogenesis in whole cells,” Molecular and Cellular Biology , vol. 12, no. 1, pp. 240–247, 1992. View at Scopus A. Yayon, M. Klagsbrun, J. D. Esko, P. Leder, and D. M. Ornitz, “Cell surface, heparin-like molecules are required for binding of basic fibroblast growth factor to its high affinity receptor,” Cell , vol. 64, no. 4, pp. 841–848, 1991. View at Scopus O. Saksela, D. Moscatelli, A. Sommer, and D. B. Rifkin, “Endothelial cell-derived heparan sulfate binds basic fibroblast growth factor and protects it from proteolytic degradation,” Journal of Cell Biology , vol. 107, no. 2, pp. 743–751, 1988. View at Scopus D. Gospodarowicz and J. Cheng, “Heparin protects basic and acidic FGF from inactivation,” Journal of Cellular Physiology , vol. 128, no. 3, pp. 475–484, 1986. View at Scopus N. Itoh and D. M. Ornitz, “Evolution of the Fgf and Fgfr gene families,” Trends in Genetics , vol. 20, no. 11, pp. 563–569, 2004. View at Publisher · View at Google Scholar · View at Scopus A. Beenken and M. Mohammadi, “The FGF family: biology, pathophysiology and therapy,” Nature Reviews Drug Discovery , vol. 8, no. 3, pp. 235–253, 2009. View at Publisher · View at Google Scholar · View at Scopus J. S. Colvin, A. C. white, S. J. Pratt, and D. M. Ornitz, “Lung hypoplasia and neonatal death in Fgf9-null mice identify this gene as an essential regulator of lung mesenchyme,” Development , vol. 128, no. 11, pp. 2095–2106, 2001. View at Scopus J. S. Colvin, R. P. Green, J. Schmahl, B. Capel, and D. M. Ornitz, “Male-to-female sex reversal in mice lacking fibroblast growth factor 9,” Cell , vol. 104, no. 6, pp. 875–889, 2001. View at Publisher · View at Google Scholar · View at Scopus M. Tekin, B. Ö. Hişmi, S. Fitoz et al., “Homozygous mutations in fibroblast growth factor 3 are associated with a new form of syndromic deafness characterized by inner ear agenesis, microtia, and microdontia,” American Journal of Human Genetics , vol. 80, no. 2, pp. 338–344, 2007. View at Publisher · View at Google Scholar · View at Scopus H. Usui, M. Shibayama, N. Ohbayashi, M. Konishi, S. Takada, and N. Itoh, “Fgf18 is required for embryonic lung alveolar development,” Biochemical and Biophysical Research Communications , vol. 322, no. 3, pp. 887–892, 2004. View at Publisher · View at Google Scholar · View at Scopus N. Ohbayashi, M. Shibayama, Y. Kurotaki et al., “FGF18 is required for normal cell proliferation and differentiation during osteogenesis and chondrogenesis,” Genes and Development , vol. 16, no. 7, pp. 870–879, 2002. View at Publisher · View at Google Scholar · View at Scopus Z. Liu, J. Xu, J. S. Colvin, and D. M. Ornitz, “Coordination of chondrogenesis and osteogenesis by fibroblast growth factor 18,” Genes and Development , vol. 16, no. 7, pp. 859–869, 2002. View at Publisher · View at Google Scholar · View at Scopus N. Itoh and D. M. Ornitz, “Fibroblast growth factors: from molecular evolution to roles in development, metabolism and disease,” Journal of Biochemistry , vol. 149, no. 2, pp. 121–130, 2011. View at Publisher · View at Google Scholar · View at Scopus S. Werner, K. G. Peters, M. T. Longaker, F. Fuller-Pace, M. J. Banda, and L. T. Williams, “Large induction of keratinocyte growth factor expression in the dermis during wound healing,” Proceedings of the National Academy of Sciences of the United States of America , vol. 89, no. 15, pp. 6896–6900, 1992. View at Publisher · View at Google Scholar · View at Scopus P. Rieck, M. Assouline, M. Savoldelli et al., “Recombinant human basic fibroblast growth factor (Rh-bFGF) in three different wound models in rabbits: corneal wound healing effect and pharmacology,” Experimental Eye Research , vol. 54, no. 6, pp. 987–998, 1992. View at Publisher · View at Google Scholar · View at Scopus J. Slavin, “Fibroblast growth factors: at the heart of angiogenesis,” Cell Biology International , vol. 19, no. 5, pp. 431–444, 1995. View at Publisher · View at Google Scholar · View at Scopus W. A. Hossain and D. K. Morest, “Fibroblast growth factors (FGF-1, FGF-2) promote migration and neurite growth of mouse cochlear ganglion cells in vitro: immunohistochemistry and antibody perturbation,” Journal of Neuroscience Research , vol. 62, no. 1, pp. 40–55, 2000. S. Tanaka, T. Kunath, A.-K. Hadjantonakis, A. Nagy, and J. Rossant, “Promotion to trophoblast stem cell proliferation by FGF4,” Science , vol. 282, no. 5396, pp. 2072–2075, 1998. View at Publisher · View at Google Scholar · View at Scopus S. E. Webb, K. K. Lee, M. K. Tang, et al., “Fibroblast growth factors 2 and 4 stimulate migration of mouse embryonic limb myogenic cells,” Developmental Dynamics , vol. 209, no. 2, pp. 206–216, 1997. S. Werner, W. Weinberg, X. Liao et al., “Targeted expression of a dominant-negative FGF receptor mutant in the epidermis of transgenic mice reveals a role of FGF in keratinocyte organization and differentiation,” EMBO Journal , vol. 12, no. 7, pp. 2635–2643, 1993. View at Scopus M. Murphy, J. Drago, and P. F. Bartlett, “Fibroblast growth factor stimulates the proliferation and differentiation of neural precursor cells in vitro,” Journal of Neuroscience Research , vol. 25, no. 4, pp. 463–475, 1990. View at Scopus N. Turner and R. Grose, “Fibroblast growth factor signalling: from development to cancer,” Nature Reviews Cancer , vol. 10, no. 2, pp. 116–129, 2010. View at Publisher · View at Google Scholar · View at Scopus D. Gospodarowicz, “Localisation of a fibroblast growth factor and its effect along and with hydrocortisone on 3T3 cell growth,” Nature , vol. 249, no. 5453, pp. 123–127, 1974. View at Scopus H. A. Armelin, “Pituitary extracts and steroid hormones in the control of 3T3 cell growth,” Proceedings of the National Academy of Sciences of the United States of America , vol. 70, no. 9, pp. 2702–2706, 1973. View at Scopus C. M. Carreira, M. Landriscina, S. Bellum, I. Prudovsky, and T. Maciag, “The comparative release of FGF1 by hypoxia and temperature stress,” Growth Factors , vol. 18, no. 4, pp. 277–285, 2001. View at Scopus A. Jackson, S. Friedman, X. Zhan, K. A. Engleka, R. Forough, and T. Maciag, “Heat shock induces the release of fibroblast growth factor 1 from NIH 3T3 cells,” Proceedings of the National Academy of Sciences of the United States of America , vol. 89, no. 22, pp. 10691–10695, 1992. View at Publisher · View at Google Scholar · View at Scopus J. T. Shin, S. R. Opalenik, J. N. Wehby et al., “Serum-starvation induces the extracellular appearance of FGF-1,” Biochimica et Biophysica Acta, Molecular Cell Research , vol. 1312, no. 1, pp. 27–38, 1996. View at Publisher · View at Google Scholar · View at Scopus N. M. Ananyeva, A. V. Tjurmin, J. A. Berliner, et al., “Oxidized LDL mediates the release of fibroblast growth factor-1,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 17, no. 3, pp. 445–453, 1997. View at Publisher · View at Google Scholar S. K. Mohan, S. G. Rani, S. M. Kumar, and C. Yu, “S100A13-C2A binary complex structure-a key component in the acidic fibroblast growth factor for the non-classical pathway,” Biochemical and Biophysical Research Communications , vol. 380, no. 3, pp. 514–519, 2009. View at Publisher · View at Google Scholar · View at Scopus M. Landriscina, C. Bagalá, A. Mandinova et al., “Copper induces the assembly of a multiprotein aggregate implicated in the release of fibroblast growth factor 1 in response to stress,” The Journal of Biological Chemistry , vol. 276, no. 27, pp. 25549–25557, 2001. View at Publisher · View at Google Scholar · View at Scopus C. M. Carreira, T. M. LaVallee, F. Tarantini et al., “S100A13 is involved in the regulation of fibroblast growth factor-1 and p40 synaptotagmin-1 release in vitro,” The Journal of Biological Chemistry , vol. 273, no. 35, pp. 22224–22231, 1998. View at Publisher · View at Google Scholar · View at Scopus F. Tarantini, T. Lavallee, A. Jackson et al., “The extravesicular domain of synaptotagmin-1 is released with the latent fibroblast growth factor-1 homodimer in response to heat shock,” The Journal of Biological Chemistry , vol. 273, no. 35, pp. 22209–22216, 1998. View at Publisher · View at Google Scholar · View at Scopus S. Uriel, E. M. Brey, and H. P. Greisler, “Sustained low levels of fibroblast growth factor-1 promote persistent microvascular network formation,” American Journal of Surgery , vol. 192, no. 5, pp. 604–609, 2006. View at Publisher · View at Google Scholar · View at Scopus A. Iwakura, M. Fujita, M. Ikemoto et al., “Myocardial ischemia enhances the expression of acidic fibroblast growth factor in human pericardial fluid,” Heart and Vessels , vol. 15, no. 3, pp. 112–116, 2000. View at Scopus P. Cuevas, D. Reimers, F. Carceller et al., “Fibroblast growth factor-1 prevents myocardial apoptosis triggered by ischemia reperfusion injury,” European Journal of Medical Research , vol. 2, no. 11, pp. 465–468, 1997. View at Scopus J.-C. Wu, W.-C. Huang, Y.-A. Tsai, Y.-C. Chen, and H. Cheng, “Nerve repair using acidic fibroblast growth factor in human cervical spinal cord injury: a preliminary Phase I clinical study,” Journal of Neurosurgery: Spine , vol. 8, no. 3, pp. 208–214, 2008. View at Publisher · View at Google Scholar · View at Scopus X. Xia, J. P. Babcock, S. I. Blaber, et al., “Pharmacokinetic properties of 2nd-generation fibroblast growth factor-1 mutants for therapeutic application,” PLoS ONE , vol. 7, no. 11, Article ID e48210, 2012. View at Publisher · View at Google Scholar J. Belch, W. R. Hiatt, I. Baumgartner et al., “Effect of fibroblast growth factor NV1FGF on amputation and death: a randomised placebo-controlled trial of gene therapy in critical limb ischaemia,” The Lancet , vol. 377, no. 9781, pp. 1929–1937, 2011. View at Publisher · View at Google Scholar · View at Scopus A. J. Comerota, R. C. Throm, K. A. Miller et al., “Naked plasmid DNA encoding fibroblast growth factor type 1 for the treatment of end-stage unreconstructible lower extremity ischemia: preliminary results of a phase I trial,” Journal of Vascular Surgery , vol. 35, no. 5, pp. 930–936, 2002. View at Publisher · View at Google Scholar · View at Scopus B. Birrer, “Whole genome oligonucleotide-based array comparative genomic hybridization analysis identified fibroblast growth factor 1 as a prognostic marker for advanced-stage serous ovarian adenocarcinomas,” Journal of Clinical Oncology , vol. 25, no. 21, p. 3184, 2007. View at Publisher · View at Google Scholar · View at Scopus B. Kwabi-Addo, M. Ozen, and M. Ittmann, “The role of fibroblast growth factors and their receptors in prostate cancer,” Endocrine-Related Cancer , vol. 11, no. 4, pp. 709–724, 2004. View at Publisher · View at Google Scholar · View at Scopus V. P. Eswarakumar, I. Lax, and J. Schlessinger, “Cellular signaling by fibroblast growth factor receptors,” Cytokine and Growth Factor Reviews , vol. 16, no. 2, pp. 139–149, 2005. View at Publisher · View at Google Scholar · View at Scopus D. M. Ornitz and N. Itoh, “Fibroblast growth factors,” Genome Biology , vol. 2, no. 3, article 3005, 2001. View at Scopus M. Bhattacharjee, R. Raju, A. Radhakrishnan, et al., “A bioinformatics resource for TWEAK-Fn14 signaling pathway,” Journal of Signal Transduction , vol. 2012, Article ID 376470, 10 pages, 2012. View at Publisher · View at Google Scholar D. Telikicherla, A. Ambekar, S. Palapetta et al., “A comprehensive curated resource for follicle stimulating hormone signaling,” BMC Research Notes , vol. 4, article 408, 2011. View at Publisher · View at Google Scholar · View at Scopus K. Kandasamy, S. Sujatha Mohan, R. Raju et al., “NetPath: A public resource of curated signal transduction pathways,” Genome Biology , vol. 11, no. 1, article r3, 2010. View at Publisher · View at Google Scholar · View at Scopus R. Raju, V. Nanjappa, L. Balakrishnan et al., “NetSlim: high-confidence curated signaling maps,” The Journal of Biological Databases and Curation , vol. 2011, p. bar032, 2011. View at Scopus M. Manuvakhova, J. V. Thottassery, S. Hays et al., “Expression of the SNT-1/FRS2 phosphotyrosine binding domain inhibits activation of MAP kinase and PI3-kinase pathways and antiestrogen resistant growth induced by FGF-1 in human breast carcinoma cells,” Oncogene , vol. 25, no. 44, pp. 6003–6014, 2006. View at Publisher · View at Google Scholar · View at Scopus S. H. Ong, Y. R. Hadari, N. Gotoh, G. R. Guy, J. Schlessinger, and I. Lax, “Stimulation of phosphatidylinositol 3-kinase by fibroblast growth factor receptors is mediated by coordinated recruitment of multiple docking proteins,” Proceedings of the National Academy of Sciences of the United States of America , vol. 98, no. 11, pp. 6074–6079, 2001. View at Publisher · View at Google Scholar · View at Scopus Y. R. Hadari, H. Kouhara, I. Lax, and J. Schlessinger, “Binding of Shp2 tyrosine phosphatase to FRS2 is essential for fibroblast growth factor-induced PC12 cell differentiation,” Molecular and Cellular Biology , vol. 18, no. 7, pp. 3966–3973, 1998. View at Scopus H. Kouhara, Y. R. Hadari, T. Spivak-Kroizman et al., “A lipid-anchored Grb2-binding protein that links FGF-receptor activation to the Ras/MAPK signaling pathway,” Cell , vol. 89, no. 5, pp. 693–702, 1997. View at Scopus M. Kanai, M. Göke, S. Tsunekawa, and D. K. Podolsky, “Signal transduction pathway of human fibroblast growth factor receptor 3. Identification of a novel 66-kDa phosphoprotein,” The Journal of Biological Chemistry , vol. 272, no. 10, pp. 6621–6628, 1997. View at Publisher · View at Google Scholar · View at Scopus Y. R. Hadari, H. Kouhara, I. Lax, and J. Schlessinger, “Binding of Shp2 tyrosine phosphatase to FRS2 is essential for fibroblast growth factor-induced PC12 cell differentiation,” Molecular and Cellular Biology , vol. 18, no. 7, pp. 3966–3973, 1998. View at Scopus W.-F. Lin, C.-J. Chen, Y.-J. Chang, S.-L. Chen, I.-M. Chiu, and L. Chen, “SH2B1 β enhances fibroblast growth factor 1 (FGF1)-induced neurite outgrowth through MEK-ERK1/2-STAT3-Egr1 pathway,” Cellular Signalling , vol. 21, no. 7, pp. 1060–1072, 2009. View at Publisher · View at Google Scholar · View at Scopus M. Mohammadi, I. Dikic, A. Sorokin, W. H. Burgess, M. Jaye, and J. Schlessinger, “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 , vol. 16, no. 3, pp. 977–989, 1996. View at Scopus A. Willems-Widyastuti, B. M. Vanaudenaerde, R. Vos et al., “Azithromycin attenuates fibroblast growth factors induced vascular endothelial growth factor Via p38MAPK signaling in human airway smooth muscle cells,” Cell Biochemistry and Biophysics , vol. 67, no. 2, pp. 331–339, 2013. View at Publisher · View at Google Scholar · View at Scopus T. Nishida, J.-I. Ito, Y. Nagayasu, and S. Yokoyama, “FGF-1-induced reactions for biogenesis of apoE-HDL are mediated by Src in rat astrocytes,” Journal of Biochemistry , vol. 146, no. 6, pp. 881–886, 2009. View at Publisher · View at Google Scholar · View at Scopus C. W. Chen, C. S. Liu, I. M. Chiu, et al., “The signals of FGFs on the neurogenesis of embryonic stem cells,” Journal of Biomedical Science , vol. 17, p. 33, 2010. View at Publisher · View at Google Scholar G. Lungu, L. Covaleda, O. Mendes, H. Martini-Stoica, and G. Stoica, “FGF-1-induced matrix metalloproteinase-9 expression in breast cancer cells is mediated by increased activities of NF-kappa;B and activating protein-1,” Molecular Carcinogenesis , vol. 47, no. 6, pp. 424–435, 2008. View at Publisher · View at Google Scholar · View at Scopus F. S. Newell, H. Su, H. Tornqvist, J. P. Whitehead, J. B. Prins, and L. J. Hutley, “Characterization of the transcriptional and functional effects of fibroblast growth factor-1 on human preadipocyte differentiation,” The FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology , vol. 20, no. 14, pp. 2615–2617, 2006. View at Publisher · View at Google Scholar · View at Scopus D. R. Newman, C.-M. Li, R. Simmons, J. Khosla, and P. L. Sannes, “Heparin affects signaling pathways stimulated by fibroblast growth factor-1 and -2 in type II cells,” American Journal of Physiology, Lung Cellular and Molecular Physiology , vol. 287, no. 1, pp. L191–L200, 2004. View at Publisher · View at Google Scholar · View at Scopus A. Buehlera, A. Martirea, C. Strohma, et al., “Angiogenesis-independent cardioprotection in FGF-1 transgenic mice,” Cardiovascular Research , vol. 55, no. 4, pp. 768–777, 2002. View at Publisher · View at Google Scholar C. A. Castaneda, H. Cortes-Funes, H. L. Gomez, and E. M. Ciruelos, “The phosphatidyl inositol 3-kinase/AKT signaling pathway in breast cancer,” Cancer Metastasis Reviews , vol. 29, no. 4, pp. 751–759, 2010. View at Scopus J.-I. Ito, Y. Nagayasu, K. Okumura-Noji et al., “Mechanism for FGF-1 to regulate biogenesis of apoE-HDL in astrocytes,” Journal of Lipid Research , vol. 48, no. 9, pp. 2020–2027, 2007. View at Publisher · View at Google Scholar · View at Scopus R. Forough, B. Weylie, C. Patel, S. Ambrus, U. S. Singh, and J. Zhu, “Role of AKT/PKB signaling in fibroblast growth factor-1 (FGF-1)-induced angiogenesis in the chicken chorioallantoic membrane (CAM),” Journal of Cellular Biochemistry , vol. 94, no. 1, pp. 109–116, 2005. View at Publisher · View at Google Scholar · View at Scopus J. Wang, T. Ito, N. Udaka, K. Okudela, T. Yazawa, and H. Kitamura, “PI3K-AKT pathway mediates growth and survival signals during development of fetal mouse lung,” Tissue and Cell , vol. 37, no. 1, pp. 25–35, 2005. View at Publisher · View at Google Scholar · View at Scopus W.-F. Lin, C.-J. Chen, Y.-J. Chang, S.-L. Chen, I.-M. Chiu, and L. Chen, “SH2B1 β enhances fibroblast growth factor 1 (FGF1)-induced neurite outgrowth through MEK-ERK1/2-STAT3-Egr1 pathway,” Cellular Signalling , vol. 21, no. 7, pp. 1060–1072, 2009. View at Publisher · View at Google Scholar · View at Scopus M. A. Hossain, J. C. Russell, R. Gomes, and J. Laterra, “Neuroprotection by scatter factor/hepatocyte growth factor and FGF-1 in cerebellar granule neurons is phosphatidylinositol 3-kinase/Akt-dependent and MAPK/CREB-independent,” Journal of Neurochemistry , vol. 81, no. 2, pp. 365–378, 2002. View at Publisher · View at Google Scholar · View at Scopus P. Li, S. Oparil, W. Feng, and Y.-F. Chen, “Hypoxia-responsive growth factors upregulate periostin and osteopontin expression via distinct signaling pathways in rat pulmonary arterial smooth muscle cells,” Journal of Applied Physiology , vol. 97, no. 4, pp. 1550–1558, 2004. View at Publisher · View at Google Scholar · View at Scopus A. Raucci, E. Laplantine, A. Mansukhani, and C. Basilico, “Activation of the ERK1/2 and p38 mitogen-activated protein kinase pathways mediates fibroblast growth factor-induced growth arrest of chondrocytes,” The Journal of Biological Chemistry , vol. 279, no. 3, pp. 1747–1756, 2004. View at Publisher · View at Google Scholar · View at Scopus J. Jiao, J. S. Greendorfer, P. Zhang, K. R. Zinn, C. A. Diglio, and J. A. Thompson, “Alternatively spliced FGFR-1 isoform signaling differentially modulates endothelial cell responses to peroxynitrite,” Archives of Biochemistry and Biophysics , vol. 410, no. 2, pp. 187–200, 2003. View at Publisher · View at Google Scholar · View at Scopus P. B. Mehta, C. N. Robson, D. E. Neal, and H. Y. Leung, “Keratinocyte growth factor activates p38 MAPK to induce stress fibre formation in human prostate DU145 cells,” Oncogene , vol. 20, no. 38, pp. 5359–5365, 2001. View at Publisher · View at Google Scholar · View at Scopus Y. J. Chang, K. W. Chen, C. J. Chen, et al., “SH2B1 β interacts with STAT3 and enhances fibroblast growth factor 1-induced gene expression during neuronal differentiation,” Molecular and Cellular Biology , vol. 34, no. 6, pp. 1003–1019, 2014. A. A. Dudka, S. M. M. Sweet, and J. K. Heath, “Signal transducers and activators of transcription-3 binding to the fibroblast growth factor receptor is activated by receptor amplification,” Cancer Research , vol. 70, no. 8, pp. 3391–3401, 2010. View at Publisher · View at Google Scholar · View at Scopus W.-F. Lin, C.-J. Chen, Y.-J. Chang, S.-L. Chen, I.-M. Chiu, and L. Chen, “SH2B1 β enhances fibroblast growth factor 1 (FGF1)-induced neurite outgrowth through MEK-ERK1/2-STAT3-Egr1 pathway,” Cellular Signalling , vol. 21, no. 7, pp. 1060–1072, 2009. View at Publisher · View at Google Scholar · View at Scopus G. Lungu, L. Covaleda, O. Mendes, H. Martini-Stoica, and G. Stoica, “FGF-1-induced matrix metalloproteinase-9 expression in breast cancer cells is mediated by increased activities of NF-kappa;B and activating protein-1,” Molecular Carcinogenesis , vol. 47, no. 6, pp. 424–435, 2008. View at Publisher · View at Google Scholar · View at Scopus X. Zhu, J. Sasse, D. McAllister, et al., “Evidence that fibroblast growth factors 1 and 4 participate in regulation of cardiogenesis,” Developmental Dynamics , vol. 207, no. 4, pp. 429–438, 1996. X. Qu, K. Hertzler, Y. Pan, K. Grobe, M. L. Robinson, and X. Zhang, “Genetic epistasis between heparan sulfate and FGF-Ras signaling controls lens development,” Developmental Biology , vol. 355, no. 1, pp. 12–20, 2011. View at Publisher · View at Google Scholar · View at Scopus A. E. Serls, S. Doherty, P. Parvatiyar, J. M. Wells, and G. H. Deutsch, “Different thresholds of fibroblast growth factors pattern the ventral foregut into liver and lung,” Development , vol. 132, no. 1, pp. 35–47, 2005. View at Publisher · View at Google Scholar · View at Scopus D. Lebeche, S. Malpel, and W. V. Cardoso, “Fibroblast growth factor interactions in the developing lung,” Mechanisms of Development , vol. 86, no. 1-2, pp. 125–136, 1999. View at Publisher · View at Google Scholar · View at Scopus J. Jung, M. Zheng, M. Goldfarb, and K. S. Zaret, “Initiation of mammalian liver development from endoderm by fibroblast growth factors,” Science , vol. 284, no. 5422, pp. 1998–2003, 1999. View at Publisher · View at Google Scholar · View at Scopus X. Luo, L. J. Hutley, J. A. Webster et al., “Identification of BMP and activin membrane-bound inhibitor (BAMBI) as a potent negative regulator of adipogenesis and modulator of autocrine/paracrine adipogenic factors,” Diabetes , vol. 61, no. 1, pp. 124–136, 2012. View at Publisher · View at Google Scholar · View at Scopus L. Hutley, W. Shurety, F. Newell et al., “Fibroblast growth factor 1: a key regulator of human adipogenesis,” Diabetes , vol. 53, no. 12, pp. 3097–3106, 2004. View at Publisher · View at Google Scholar · View at Scopus T. Tran, V. Kolupaeva, and C. Basilico, “FGF inhibits the activity of the cyclin B1/CDK1 kinase to induce a transient G 2 arrest in RCS chondrocytes,” Cell Cycle , vol. 9, no. 21, pp. 4379–4386, 2010. View at Publisher · View at Google Scholar · View at Scopus V. Kolupaeva, E. Laplantine, and C. Basilico, “PP2A-mediated dephosphorylation of p107 plays a critical role in chondrocyte cell cycle arrest by FGF,” PLoS ONE , vol. 3, no. 10, Article ID e3447, 2008. View at Publisher · View at Google Scholar · View at Scopus R. Priore, L. Dailey, and C. Basilico, “Downregulation of Akt activity contributes to the growth arrest induced by FGF in chondrocytes,” Journal of Cellular Physiology , vol. 207, no. 3, pp. 800–808, 2006. View at Publisher · View at Google Scholar · View at Scopus L. Dailey, E. Laplantine, R. Priore, and C. Basilico, “A network of transcriptional and signaling events is activated by FGF to induce chondrocyte growth arrest and differentiation,” Journal of Cell Biology , vol. 161, no. 6, pp. 1053–1066, 2003. View at Publisher · View at Google Scholar · View at Scopus P. Zhang, J. S. Greendorfer, J. Jiao, S. C. Kelpke, and J. A. Thompson, “Alternatively spliced FGFR-1 isoforms differentially modulate endothelial cell activation of c-YES,” Archives of Biochemistry and Biophysics , vol. 450, no. 1, pp. 50–62, 2006. View at Publisher · View at Google Scholar · View at Scopus B. Fernandez, A. Buehler, S. Wolfram et al., “Transgenic myocardial overexpression of fibroblast growth factor-1 increases coronary artery density and branching,” Circulation Research , vol. 87, no. 3, pp. 207–213, 2000. View at Scopus B. Schumacher, P. Peecher, B. U. Von Specht, and T. Stegmann, “Induction of neoangiogenesis in ischemic myocardium by human growth factors: first clinical results of a new treatment of coronary heart disease,” Circulation , vol. 97, no. 7, pp. 645–650, 1998. View at Scopus J.-I. Ito, Y. Nagayasu, R. Lu, A. Kheirollah, M. Hayashi, and S. Yokoyama, “Astrocytes produce and secrete FGF-1, which promotes the production of apoE-HDL in a manner of autocrine action,” Journal of Lipid Research , vol. 46, no. 4, pp. 679–686, 2005. View at Publisher · View at Google Scholar · View at Scopus L. Sun, L. Xu, H. Chang et al., “Transfection with aFGF cDNA improves wound healing,” Journal of Investigative Dermatology , vol. 108, no. 3, pp. 313–318, 1997. View at Scopus U. Pirvola, Y. Cao, C. Oellig, Z. Suoqiang, R. F. Pettersson, and J. Ylikoski, “The site of action of neuronal acidic fibroblast growth factor is the organ of corti of the rat cochlea,” Proceedings of the National Academy of Sciences of the United States of America , vol. 92, no. 20, pp. 9269–9273, 1995. View at Publisher · View at Google Scholar · View at Scopus M. Palmen, M. J. A. P. Daemen, L. J. De Windt et al., “Fibroblast growth factor-1 improves cardiac functional recovery and enhances cell survival after ischemia and reperfusion: a fibroblast growth factor receptor, protein kinase C, and tyrosine kinase-dependent mechanism,” Journal of the American College of Cardiology , vol. 44, no. 5, pp. 1113–1123, 2004. View at Publisher · View at Google Scholar · View at Scopus A. Buehler, A. Martire, C. Strohm et al., “Angiogenesis-independent cardioprotection in FGF-1 transgenic mice,” Cardiovascular Research , vol. 55, no. 4, pp. 768–777, 2002. View at Publisher · View at Google Scholar · View at Scopus P. Htun, W. D. Ito, I. E. Hoefer, J. Schaper, and W. Schaper, “Intramyocardial infusion of FGF-1 mimics ischemic preconditioning in pig myocardium,” Journal of Molecular and Cellular Cardiology , vol. 30, no. 4, pp. 867–877, 1998. View at Publisher · View at Google Scholar · View at Scopus M. Hashimoto, Y. Sagara, D. Langford et al., “Fibroblast growth factor 1 regulates signaling via the glycogen synthase kinase-3 β pathway. Implications for neuroprotection,” The Journal of Biological Chemistry , vol. 277, no. 36, pp. 32985–32991, 2002. View at Publisher · View at Google Scholar · View at Scopus J. Taeger, C. Moser, C. Hellerbrand et al., “Targeting FGFR/PDGFR/VEGFR impairs tumor growth, angiogenesis, and metastasis by effects on tumor cells, endothelial cells, and pericytes in pancreatic cancer,” Molecular Cancer Therapeutics , vol. 10, no. 11, pp. 2157–2167, 2011. View at Publisher · View at Google Scholar · View at Scopus E. M. Haugsten, M. Zakrzewska, A. Brech et al., “Clathrin- and dynamin-independent endocytosis of fgfr3—implications for signalling,” PLoS ONE , vol. 6, no. 7, Article ID e21708, 2011. View at Publisher · View at Google Scholar · View at Scopus C. Bonneton, J. B. Sibarita, and J. P. Thiery, “Relationship between cell migration and cell cycle during the initiation of epithelial to fibroblastoid transition,” Cell Motility and the Cytoskeleton , vol. 43, no. 4, pp. 288–295, 1999. Z. Liu, Y. E. Hartman, J. M. Warram et al., “Fibroblast growth factor receptor mediates fibroblast-dependent growth in EMMPRIN-depleted head and neck cancer tumor cells,” Molecular Cancer Research , vol. 9, no. 8, pp. 1008–1017, 2011. View at Publisher · View at Google Scholar · View at Scopus N. R. Estes II, J. V. Thottassery, L. Westbrook, and F. G. Kern, “MEK ablation in MCF-7 cells blocks DNA synthesis induced by serum, but not by estradiol or growth factors,” International Journal of Oncology , vol. 29, no. 6, pp. 1573–1580, 2006. View at Scopus O. Klingenberg, A. Wiçdłocha, A. Rapak, R. Muñoz, P. Ø. Falnes, and S. Olsnes, “Inability of the acidic fibroblast growth factor mutant K132E to stimulate DNA synthesis after translocation into cells,” The Journal of Biological Chemistry , vol. 273, no. 18, pp. 11164–11172, 1998. View at Publisher · View at Google Scholar · View at Scopus J.-M. Rodier, A. M. Valles, M. Denoyelle, J. P. Thiery, and B. Boyer, “pp60(c-src) Is a positive regulator of growth factor-induced cell scattering in a rat bladder carcinoma cell line,” Journal of Cell Biology , vol. 131, no. 3, pp. 761–773, 1995. View at Publisher · View at Google Scholar · View at Scopus A. M. Valles, B. Boyer, J. Badet, G. C. Tucker, D. Barritault, and J. P. Thiery, “Acidic fibroblast growth factor is a modulator of epithelial plasticity in a rat bladder carcinoma cell line,” Proceedings of the National Academy of Sciences of the United States of America , vol. 87, no. 3, pp. 1124–1128, 1990. View at Scopus J. Jouanneau, J. Plouet, G. Moens, and J. P. Thiery, “FGF-2 and FGF-1 expressed in rat bladder carcinoma cells have similar angiogenic potential but different tumorigenic properties in vivo,” Oncogene , vol. 14, no. 6, pp. 671–676, 1997. View at Scopus J. Jouanneau, J. Gavrilovic, D. Caruelle et al., “Secreted or nonsecreted forms of acidic fibroblast growth factor produced by transfected epithelial cells influence cell morphology, motility, and invasive potential,” Proceedings of the National Academy of Sciences of the United States of America , vol. 88, no. 7, pp. 2893–2897, 1991. View at Scopus P. Shannon, A. Markiel, O. Ozier et al., “Cytoscape: a software environment for integrated models of biomolecular interaction networks,” Genome Research , vol. 13, no. 11, pp. 2498–2504, 2003. View at Publisher · View at Google Scholar · View at Scopus O. Babur, U. Dogrusoz, E. Demir, and C. Sander, “ChiBE: interactive visualization and manipulation of BioPAX pathway models,” Bioinformatics , vol. 26, no. 3, pp. 429–431, 2010. View at Scopus A. Dilek, M. E. Belviranli, and U. Dogrusoz, “VISIBIOweb: visualization and layout services for BioPAX pathway models,” Nucleic Acids Research , vol. 38, no. 2, Article ID gkq352, pp. W150–W154, 2010. View at Publisher · View at Google Scholar · View at Scopus M. P. van Iersel, T. Kelder, A. R. Pico et al., “Presenting and exploring biological pathways with PathVisio,” BMC Bioinformatics , vol. 9, article 399, 2008. View at Publisher · View at Google Scholar · View at Scopus T. S. K. Prasad, R. Goel, K. Kandasamy et al., “Human protein reference database—2009 update,” Nucleic Acids Research , vol. 37, no. 1, pp. D767–D772, 2009. View at Publisher · View at Google Scholar · View at Scopus var _gaq = _gaq || []; _gaq.push(['_setAccount', 'UA-8578054-2']); _gaq.push(['_trackPageview']); (function () { var ga = document.createElement('script'); ga.type = 'text/javascript'; ga.async = true; ga.src = ('https:' == document.location.protocol ? 'https://ssl' : 'http://www') + '.google-analytics.com/ga.js'; var s = document.getElementsByTagName('script')[0]; s.parentNode.insertBefore(ga, s); })(); http://www.deepdyve.com/assets/images/DeepDyve-Logo-lg.png Journal of Signal Transduction Hindawi Publishing Corporation

Loading next page...
 
/lp/hindawi-publishing-corporation/a-network-map-of-fgf-1-fgfr-signaling-system-wgGpHKvWvN

References

References for this paper are not available at this time. We will be adding them shortly, thank you for your patience.

Publisher
Hindawi Publishing Corporation
Copyright
Copyright © 2014 Rajesh Raju et al.
ISSN
2090-1739
eISSN
2090-1747
Publisher site
See Article on Publisher Site

Abstract

A Network Map of FGF-1/FGFR Signaling System div.banner_title_bkg div.trangle { border-color: #2D4917 transparent transparent transparent; opacity:0.8; /*new styles start*/ -ms-filter:"progid:DXImageTransform.Microsoft.Alpha(Opacity=80)" ;filter: alpha(opacity=80); /*new styles end*/ } div.banner_title_bkg_if div.trangle { border-color: transparent transparent #2D4917 transparent ; opacity:0.8; /*new styles start*/ -ms-filter:"progid:DXImageTransform.Microsoft.Alpha(Opacity=80)" ;filter: alpha(opacity=80); /*new styles end*/ } div.banner_title_bkg div.trangle { width: 292px; } #banner { background-image: url('http://images.hindawi.com/journals/jst/jst.banner.jpg'); background-position: 50% 0;} Hindawi Publishing Corporation Home Journals About Us Journal of Signal Transduction About this Journal Submit a Manuscript Table of Contents Journal Menu About this Journal · Abstracting and Indexing · Aims and Scope · Article Processing Charges · Articles in Press · Author Guidelines · Bibliographic Information · Citations to this Journal · Contact Information · Editorial Board · Editorial Workflow · Free eTOC Alerts · Publication Ethics · Reviewers Acknowledgment · Submit a Manuscript · Subscription Information · Table of Contents Open Special Issues · Published Special Issues · Special Issue Guidelines Abstract Full-Text PDF Full-Text HTML Full-Text ePUB Linked References How to Cite this Article Journal of Signal Transduction Volume 2014 (2014), Article ID 962962, 16 pages http://dx.doi.org/10.1155/2014/962962 Review Article A Network Map of FGF-1/FGFR Signaling System Rajesh Raju , 1 Shyam Mohan Palapetta , 1,2 Varot K. Sandhya , 1 Apeksha Sahu , 1,2 Abbas Alipoor , 3 Lavanya Balakrishnan , 1 Jayshree Advani , 1 Bijesh George , 1 K. Ramachandra Kini , 3 N. P. Geetha , 3 H. S. Prakash , 3 T. S. Keshava Prasad , 1 Yu-Jung Chang , 4 Linyi Chen , 4 Akhilesh Pandey , 5,6,7,8 and Harsha Gowda 1 1 Institute of Bioinformatics, International Tech Park, Bangalore 560066, India 2 Centre of Excellence in Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry 605014, India 3 Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysore 570006, India 4 Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan 5 McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA 6 Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA 7 Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA 8 Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA Received 20 November 2013; Accepted 3 March 2014; Published 16 April 2014 Academic Editor: Shoukat Dedhar Copyright © 2014 Rajesh Raju et al. This is an open access article distributed under the Creative Commons Attribution License , which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Abstract Fibroblast growth factor-1 (FGF-1) is a well characterized growth factor among the 22 members of the FGF superfamily in humans. It binds to all the four known FGF receptors and regulates a plethora of functions including cell growth, proliferation, migration, differentiation, and survival in different cell types. FGF-1 is involved in the regulation of diverse physiological processes such as development, angiogenesis, wound healing, adipogenesis, and neurogenesis. Deregulation of FGF-1 signaling is not only implicated in tumorigenesis but also is associated with tumor invasion and metastasis. Given the biomedical significance of FGFs and the fact that individual FGFs have different roles in diverse physiological processes, the analysis of signaling pathways induced by the binding of specific FGFs to their cognate receptors demands more focused efforts. Currently, there are no resources in the public domain that facilitate the analysis of signaling pathways induced by individual FGFs in the FGF/FGFR signaling system. Towards this, we have developed a resource of signaling reactions triggered by FGF-1/FGFR system in various cell types/tissues. The pathway data and the reaction map are made available for download in different community standard data exchange formats through NetPath and NetSlim signaling pathway resources. 1. Introduction Fibroblast growth factor (FGF) superfamily consists of structurally related polypeptides most of which function through its high affinity fibroblast growth factor receptors (FGFRs). In addition to FGFRs, they also bind to heparan sulfate proteoglycans (HPSGs) and their analog, heparin. These interactions influence the stability of FGFs in the extracellular matrix and also regulate their binding and activation of FGFRs [ 1 – 9 ]. In humans, FGFs are encoded by 22 genes, FGF-1-14 and FGF-16-23, and are divided into 7 subfamilies. FGFs 1–10 and 16–23 are FGFR ligands, while FGFs 11–14 are intracellular FGF homologous factors which act in a receptor-independent fashion [ 10 ]. Knock-out mice of different FGFs exhibit diverse developmental and physiological disorders [ 11 ]. For instance, FGF-9 is involved in the development of lung and testes [ 12 , 13 ], FGF-3 is critical for inner ear development [ 14 ], and FGF-18 is important in bone and lung development [ 15 – 17 ]. Moreover, knock-out of FGFs 4, 8, 9, 10, 15, 18, or 23 was found to be lethal in mice [ 18 ]. FGFs are also involved in wound healing, tissue repair [ 19 , 20 ], and angiogenesis [ 21 ]. Facilitating cell proliferation, migration, and differentiation [ 16 , 22 – 26 ], FGFs are implicated in diverse pathological conditions including cancer [ 27 ] as well as metabolic and developmental disorders [ 18 ]. Most FGFs have an N-terminal signal peptide and are thus secreted. FGFs 1, 2, 9, 16, and 20 do not have signal peptides. FGFs 9, 16, and 20 may be released through classical secretory pathway; however, FGF-1 and FGF-2 are released from damaged cells or through endoplasmic reticulum-golgi independent exocytotic pathway [ 10 ]. FGF-1 along with FGF-2 was initially isolated from bovine pituitary extracts based on their ability to induce proliferation in 3T3 fibroblasts [ 28 , 29 ]. Also known as acidic FGF, FGF-1 is a 155 amino acid long non-glycosylated polypeptide. FGF-1 is not released from the cells under normal physiological conditions, but it was secreted in response to stress conditions such as heat shock, hypoxia [ 30 , 31 ], serum starvation [ 32 ], and exposure to low-density lipoproteins [ 33 ]. Stress induces the release of inactive disulfide bond-linked homodimeric form of FGF-1, which is dependent on p40-Syt1, S100A13, and Cu 2+ ions [ 34 – 37 ]. FGF-1 has been shown to reduce apoptosis in vascular injury [ 38 – 40 ]. Administration of FGF-1 has shown promise as a therapeutic strategy against human cervical spinal cord injury [ 41 ] and ischemic conditions [ 42 – 44 ]. Increased expression of FGF-1 was observed in ovarian [ 45 ] and prostate cancers [ 46 ]. Taken together, FGF1 is involved in different cellular functions that are mediated through its interaction with the four FGF receptors [ 47 , 48 ]. A pathway resource representing these diverse functions and the underlying mechanisms that regulate these processes would be immensely useful. Curated pathway maps are invaluable resources for scientific community. Such comprehensive pathway datasets are being increasingly used in bioinformatics efforts directed towards analysis of high-throughput datasets from various disease contexts. Repositories including Pathway Interaction Database of the National Cancer Institute ( http://pid.nci.nih.gov/ ), Database of Cell Signaling ( http://stke.sciencemag.org/cm/ ), KEGG Pathway Database ( http://www.genome.jp/kegg/pathway.html ), and INOH Pathway Database ( http://inoh.org/ ) have cataloged basic components of FGF signaling. We have expanded the scope of this by providing a comprehensive representation of FGF1 signaling pathway and its diverse roles in regulating various cellular processes. 2. Methodology Documentation of specific pathway reactions scattered in the literature into an organized, user-friendly, query-enabled platform is primary to the analysis of signaling pathways. We used NCBI PubMed database to carry out an extensive literature search to retrieve research articles where molecular events triggered by the FGF-1/FGFR signaling system were studied. Specific molecular events screened include (a) physical associations between proteins, (b) posttranslational modifications (PTMs), (c) change in subcellular localization of proteins, (d) activation or inhibition of specific proteins, and (e) regulation of gene expression. Relevant information from research articles were manually documented using the curation tool, PathBuilder. To streamline and organize data collection from literature, we followed the previously described criteria for the inclusion/exclusion of pathway specific reactions [ 49 , 50 ]. The data accumulated was submitted to the NetPath signaling pathway resource developed by our group [ 51 ]. We then generated a signaling map for this pathway using PathVisio pathway visualization software. We also applied additional criteria to filter out low confidence reactions from the gathered data [ 52 ] and generated a NetSlim map. In addition to curation of molecular level information, we have also cataloged physiological effects brought about by FGF-1 in different cell types/tissues. 3. Results and Discussion Canonical FGF/FGFR signaling reactions have been documented in a few public repositories and review articles. Vast amount of literature in the last few years have revealed several novel pathway intermediates of FGF/FGFR signaling system. In order to generate a comprehensive view of FGF/FGFR signaling pathway, we carried out extensive literature search on PubMed for articles pertaining to FGF-1 signaling. Of a total of 3275 articles that were screened, 237 of them had molecular reactions reported downstream of FGF-1 in various cell types/tissues. Manual curation from these research articles revealed 109 molecules involved in FGF-1 induced physical associations, modulation by PTMs, activity, and subcellular or cell surface translocation events. Of the 42 physical associations that were cataloged, 29 were “binary” and 13 were “complex” interactions inclusive of the ligand/receptor interactors. We could record a total of 87 catalysis events, 15 activation/inhibition, and 21 translocation events. The 87 catalysis events include 19 events, where the enzymes directly catalyzing the reactions were studied and reported, and 68 events for which the enzymes which post-translationally modified the proteins are not studied under FGF-1 stimulation. Apart from these molecular reactions, we have also cataloged 117 genes whose expression is reported to be either upregulated or downregulated by FGF-1 treatment. However, only a total of 25 genes were reported to be differentially regulated at mRNA level by FGF-1 stimulation in different human cell types. A list of genes reported to be regulated by FGF-1 in different mammalian systems at the mRNA and/or the protein level is provided in Table 1 . After the annotation process, all the entries were reviewed and approved by internal reviewers. Internally reviewed pathways were further reviewed and approved by an external pathway authority (LC, who is an author in this paper). Table 1: List of genes that are reported to be transcriptionally and translationally regulated by FGF-1 in humans and other mammals. 3.1. Signaling Modules Activated by FGF-1 Signaling modules comprise a well-characterized group of molecules and their interactions downstream of activation of a receptor. We documented the following signaling modules to be activated upon stimulation with FGF-1. 3.1.1. Ras/Raf/Mek/Erk Pathway The Ras/Raf/Mek/Erk pathway has been implicated in cellular processes including cell growth, proliferation, and migration. Stimulation of different cell types with FGF-1 resulted in the formation of multiple complexes involving FRS2, GAB1, SOS1, PTPN11, SHC1, SH2B1, and GRB2 [ 53 – 60 ]. These complexes are critical to the subsequent activation of Ras [ 53 , 56 ]. Association of Ras with Raf kinase [ 53 ] induces autophosphorylation and activation of Raf. Activation of Raf leads to phosphorylation dependent activation of Map kinases 1/2 (MAP2K1/2) and subsequently Erk2/1 (MAPK1/3) [ 60 – 62 ]. In the context of FGF-1 signaling, this module was reported to be involved in a number of processes including neurogenesis, adipocyte differentiation, cell proliferation, cholesterogenesis, cardioprotection, and tumor invasion and metastasis [ 62 – 67 ]. 3.1.2. Pi3k/Akt Pathway The complexes mentioned above also lead to the activation of Pi3k/Akt pathway, another signaling module that regulates various processes including cell growth, survival, cell proliferation, and cell migration [ 68 ]. A number of studies have shown FGF-1 induced phosphorylation of Akt [ 63 , 64 , 69 ]. Pi3k inhibitor-based functional assays also proved the involvement of FGF-1 pathway in diverse physiological conditions including angiogenesis [ 70 ], lung development [ 71 ], maintenance of neuronal phenotype [ 72 ], neuroprotection [ 73 ], and ApoE-HDL secretion [ 69 ]. 3.1.3. Jnk and p38 Mapk Pathway The c-jun N-terminal kinase ( Jnk) pathway is implicated in the regulation of cell cycle, cell survival and apoptosis. FGF-1 stimulates the phosphorylation of p38 Mapk (MAPK14) as well as Jnk1/2 (MAPK8/9). The Jnk1/2 was also found to be crucial to neurogenesis and vascular remodeling [ 63 , 74 ]. The specific functions of FGF-1 signaling mediated by p38 Mapk include growth arrest, promotion of apoptosis in response to oxidative stress, and formation of actin stress fibers [ 75 – 77 ]. 3.1.4. STAT3 and Nf-kb Pathway FGF-1 also stimulates STATs (STAT1 and STAT3) and Nf-kB signaling modules. FGFR signaling is reported to be regulated through several downstream molecules including JAK2, SRC, SH2B1, MAPK1/3, MAPK8/9, and STAT3. This signaling axis is known to regulate various cellular processes including neurite outgrowth, cell proliferation, and increased cancer cell invasion [ 78 – 80 ]. In addition, FGF-1 is also reported to induce MMP9 expression in mammary adenocarcinoma cells through the Nf-kb pathway [ 81 ]. 3.2. Physiological Effects Mediated by FGF-1 FGF-1 was found to be involved in a number of biological processes. It is associated with the development of heart [ 82 ], lens [ 83 ], lung, and liver [ 84 – 86 ]. Its crucial roles in neurogenesis as well as adipogenesis [ 65 , 87 , 88 ] have also been reported. FGF-1 induces growth arrest and differentiation in chondrocytes [ 89 – 92 ]. It is implicated in angiogenesis [ 93 – 95 ] and wound healing [ 95 – 99 ]. Multiple studies have also shown the role of FGF-1 in cardioprotection [ 99 – 101 ] and neuroprotection [ 22 , 102 ]. FGF-1 also induces migration [ 103 – 105 ] and proliferation [ 106 – 108 ] in different types of cancer cells. It is also involved in the regulation of epithelial-to-mesenchymal transition [ 109 , 110 ], and tumorigenesis [ 111 ] as well as invasion and metastasis [ 64 , 112 ]. A list of functional effects of FGF-1 studied in different cell types/tissues is provided in Table 2 . Table 2: Functions of FGF-1 identified in diverse cell/tissue types of human and other mammalian origins. 3.3. Pathway Visualization, Data Formats, and Availability User-friendly visualization of pathways is an important aspect to provide a concise view. A number of tools are available for visualization and analysis of pathway data including Cytoscape [ 113 ], ChisioBioPAX Editor (ChiBE) [ 114 ], visualization and layout services for BioPAX pathway models (VISIBIOweb) [ 115 ], and ingenuity pathway analysis. These tools use pathway and molecular interaction data in different XML-based community standard data exchange formats as input. These standard formats, which include Proteomics Standards Initiative for Molecular Interaction (PSI-MI version 2.5), Biological Pathway eXchange (BioPAX level 3), and Systems Biology Markup Language (SBML version 2.1), enable easy data exchange and interoperability with multiple software. We have provided the annotated pathway data in the standard formats mentioned above. This data can be downloaded and used from NetPath [ 51 ], an open source resource for signal transduction pathways developed by our group ( http://www.netpath.org/index.html ). Additionally, we have drawn a map of FGF-1/FGFR signaling using the data accumulated in NetPath. This network map represents the molecules and their reactions organized by topology and excludes the molecules identified through phosphoproteomics approaches for which topology could not be assigned (Figure 1 ). The map was manually drawn using freely available software, PathVisio [ 116 ]. The topology of the molecules and their reactions in the pathway was arranged based on (i) inhibitor-based assays, (ii) mutation-based assays, (iii) knock-out studies, (iv) prior knowledge of canonical modules, and/or (v) with reference to multiple review articles. Another map, which incorporated high confidence reactions in accordance with NetSlim criteria [ 52 ], is submitted to the NetSlim database. These maps can be visualized and downloaded in gpml, GenMAPP, png, and pdf formats from http://www.netpath.org/netslim/FGF-1_pathway.html . Each node in the map is linked to their molecule page in NetPath, thereby to other pathways in NetPath, and to HPRD [ 117 ] and RefSeq protein accessions. In the “map with citation” option, the edges connecting the nodes are linked to the corresponding articles in PubMed that report the FGF-1 stimulated reaction(s). Direct reactions are represented by solid edges. Indirect reactions are represented with dashed edges. The edges which represent the protein-protein interactions, enzyme-substrate reactions and translocation events are distinguished by different colors. Figure 1: Network map of FGF-1 signaling. This map manually drawn using PathVisio [ 112 ] represents the reactions induced by FGF-1 through their receptors. Each node represents the molecules and the post-translationally modified states of proteins are also represented. Distinguished by color and continuous/dashed lines, the edges represent the specific information such as protein-protein interactions, enzyme-substrate reactions, reactions mediated through unknown/multiple steps, and protein translocations as provided in the legend. The biological processes that FGF-1 regulates through multiple signaling modules are also represented. A NetSlim [ 52 ] version of this map can be obtained from http://www.netpath.org/netslim/FGF-1_pathway.html . 4. Conclusions Availability of specific ligand-receptor mediated signaling data in community approved formats is crucial to the understanding of proteins and their reactions in diverse biological processes. Analysis of high-throughput data obtained from microarray- and mass spectrometry-based platforms essentially relies on enrichment of biological function or signaling pathways available in databases to obtain insights into their physiological functions. Although some resources have cataloged FGF signaling in general, this is the first attempt to provide a comprehensive view of FGF-1 signaling. This will be extended to other FGF ligands and/or specific FGFRs in the future to facilitate the analysis of differences between different FGFs and/or FGFRs. The pathway information has been made available through NetPath and NetSlim resources in multiple community standard data formats. The FGF-1 signaling pathway data will be periodically updated in NetPath. We have cataloged multiple signaling modules that are activated upon activation of FGFR and their implications in diverse physiological and pathophysiological processes. We believe that the data presented here will boost further research in this area and will help identify novel therapeutically important molecules that could be targeted in pathological conditions involving aberrant FGF-1 signaling. Abbreviations S100A13: S100 calcium binding protein A13 FRS2: Fibroblast growth factor receptor substrate 2 GAB1: GRB2-associated binding protein 1 SOS1: Son of sevenless homolog 1 PTPN11: Protein tyrosine phosphatase, non-receptor type 11 SHC1: Src homology 2 domain containing transforming protein 1 GRB2: Growth factor receptor-bound protein 2 Mapk: Mitogen activated protein kinase Pi3k: Phosphatidylinositide 3-kinase Akt: v-akt murine thymoma viral oncogene homolog HDL: High density lipoprotein Jnk: Jun N-terminal kinase STAT3: Signal transducer and activator of transcription 3. Conflict of Interests The authors have no conflict of interests. Authors’ Contribution Shyam Mohan Palapetta, Varot K. Sandhya, and Apeksha Sahu contributed equally to the paper. Acknowledgments The authors thank the Department of Biotechnology (DBT), Government of India, for research support to the Institute of Bioinformatics, Bangalore. Shyam Mohan Palapetta is supported by a Senior Research Fellowship from the Council of Scientific and Industrial Research (CSIR), India. Varot K. Sandhya is a recipient of Inspire Fellowship from the Department of Science and Technology (DST), Government of India. Harsha Gowda is a Wellcome Trust/DBT India Alliance Early Career Fellow. References N. J. Harmer, “Insights into the role of heparan sulphate in fibroblast growth factor signalling,” Biochemical Society Transactions , vol. 34, no. 3, pp. 442–445, 2006. View at Publisher · View at Google Scholar · View at Scopus O. A. Ibrahimi, F. Zhang, S. C. L. Hrstka, M. Mohammadi, and R. J. Linhardt, “Kinetic model for FGF, FGFR, and proteoglycan signal transduction complex assembly,” Biochemistry , vol. 43, no. 16, pp. 4724–4730, 2004. View at Publisher · View at Google Scholar · View at Scopus D. M. Ornitz, A. B. Herr, M. Nilsson, J. Westman, C.-M. Svahn, and G. Waksman, “FGF binding and FGF receptor activation by synthetic heparan-derived di- and trisaccharides,” Science , vol. 268, no. 5209, pp. 432–436, 1995. View at Scopus M. W. Pantoliano, “Multivalent ligand-receptor binding interactions in the fibroblast growth factor system produce a cooperative growth factor and heparin mechanism for receptor dimerization,” Biochemistry , vol. 33, no. 34, pp. 10229–10248, 1994. View at Scopus D. M. Ornitz and P. Leder, “Ligand specificity and heparin dependence of fibroblast growth factor receptors 1 and 3,” The Journal of Biological Chemistry , vol. 267, no. 23, pp. 16305–16311, 1992. View at Scopus D. M. Ornitz, A. Yayon, J. G. Flanagan, C. M. Svahn, E. Levi, and P. Leder, “Heparin is required for cell-free binding of basic fibroblast growth factor to a soluble receptor and for mitogenesis in whole cells,” Molecular and Cellular Biology , vol. 12, no. 1, pp. 240–247, 1992. View at Scopus A. Yayon, M. Klagsbrun, J. D. Esko, P. Leder, and D. M. Ornitz, “Cell surface, heparin-like molecules are required for binding of basic fibroblast growth factor to its high affinity receptor,” Cell , vol. 64, no. 4, pp. 841–848, 1991. View at Scopus O. Saksela, D. Moscatelli, A. Sommer, and D. B. Rifkin, “Endothelial cell-derived heparan sulfate binds basic fibroblast growth factor and protects it from proteolytic degradation,” Journal of Cell Biology , vol. 107, no. 2, pp. 743–751, 1988. View at Scopus D. Gospodarowicz and J. Cheng, “Heparin protects basic and acidic FGF from inactivation,” Journal of Cellular Physiology , vol. 128, no. 3, pp. 475–484, 1986. View at Scopus N. Itoh and D. M. Ornitz, “Evolution of the Fgf and Fgfr gene families,” Trends in Genetics , vol. 20, no. 11, pp. 563–569, 2004. View at Publisher · View at Google Scholar · View at Scopus A. Beenken and M. Mohammadi, “The FGF family: biology, pathophysiology and therapy,” Nature Reviews Drug Discovery , vol. 8, no. 3, pp. 235–253, 2009. View at Publisher · View at Google Scholar · View at Scopus J. S. Colvin, A. C. white, S. J. Pratt, and D. M. Ornitz, “Lung hypoplasia and neonatal death in Fgf9-null mice identify this gene as an essential regulator of lung mesenchyme,” Development , vol. 128, no. 11, pp. 2095–2106, 2001. View at Scopus J. S. Colvin, R. P. Green, J. Schmahl, B. Capel, and D. M. Ornitz, “Male-to-female sex reversal in mice lacking fibroblast growth factor 9,” Cell , vol. 104, no. 6, pp. 875–889, 2001. View at Publisher · View at Google Scholar · View at Scopus M. Tekin, B. Ö. Hişmi, S. Fitoz et al., “Homozygous mutations in fibroblast growth factor 3 are associated with a new form of syndromic deafness characterized by inner ear agenesis, microtia, and microdontia,” American Journal of Human Genetics , vol. 80, no. 2, pp. 338–344, 2007. View at Publisher · View at Google Scholar · View at Scopus H. Usui, M. Shibayama, N. Ohbayashi, M. Konishi, S. Takada, and N. Itoh, “Fgf18 is required for embryonic lung alveolar development,” Biochemical and Biophysical Research Communications , vol. 322, no. 3, pp. 887–892, 2004. View at Publisher · View at Google Scholar · View at Scopus N. Ohbayashi, M. Shibayama, Y. Kurotaki et al., “FGF18 is required for normal cell proliferation and differentiation during osteogenesis and chondrogenesis,” Genes and Development , vol. 16, no. 7, pp. 870–879, 2002. View at Publisher · View at Google Scholar · View at Scopus Z. Liu, J. Xu, J. S. Colvin, and D. M. Ornitz, “Coordination of chondrogenesis and osteogenesis by fibroblast growth factor 18,” Genes and Development , vol. 16, no. 7, pp. 859–869, 2002. View at Publisher · View at Google Scholar · View at Scopus N. Itoh and D. M. Ornitz, “Fibroblast growth factors: from molecular evolution to roles in development, metabolism and disease,” Journal of Biochemistry , vol. 149, no. 2, pp. 121–130, 2011. View at Publisher · View at Google Scholar · View at Scopus S. Werner, K. G. Peters, M. T. Longaker, F. Fuller-Pace, M. J. Banda, and L. T. Williams, “Large induction of keratinocyte growth factor expression in the dermis during wound healing,” Proceedings of the National Academy of Sciences of the United States of America , vol. 89, no. 15, pp. 6896–6900, 1992. View at Publisher · View at Google Scholar · View at Scopus P. Rieck, M. Assouline, M. Savoldelli et al., “Recombinant human basic fibroblast growth factor (Rh-bFGF) in three different wound models in rabbits: corneal wound healing effect and pharmacology,” Experimental Eye Research , vol. 54, no. 6, pp. 987–998, 1992. View at Publisher · View at Google Scholar · View at Scopus J. Slavin, “Fibroblast growth factors: at the heart of angiogenesis,” Cell Biology International , vol. 19, no. 5, pp. 431–444, 1995. View at Publisher · View at Google Scholar · View at Scopus W. A. Hossain and D. K. Morest, “Fibroblast growth factors (FGF-1, FGF-2) promote migration and neurite growth of mouse cochlear ganglion cells in vitro: immunohistochemistry and antibody perturbation,” Journal of Neuroscience Research , vol. 62, no. 1, pp. 40–55, 2000. S. Tanaka, T. Kunath, A.-K. Hadjantonakis, A. Nagy, and J. Rossant, “Promotion to trophoblast stem cell proliferation by FGF4,” Science , vol. 282, no. 5396, pp. 2072–2075, 1998. View at Publisher · View at Google Scholar · View at Scopus S. E. Webb, K. K. Lee, M. K. Tang, et al., “Fibroblast growth factors 2 and 4 stimulate migration of mouse embryonic limb myogenic cells,” Developmental Dynamics , vol. 209, no. 2, pp. 206–216, 1997. S. Werner, W. Weinberg, X. Liao et al., “Targeted expression of a dominant-negative FGF receptor mutant in the epidermis of transgenic mice reveals a role of FGF in keratinocyte organization and differentiation,” EMBO Journal , vol. 12, no. 7, pp. 2635–2643, 1993. View at Scopus M. Murphy, J. Drago, and P. F. Bartlett, “Fibroblast growth factor stimulates the proliferation and differentiation of neural precursor cells in vitro,” Journal of Neuroscience Research , vol. 25, no. 4, pp. 463–475, 1990. View at Scopus N. Turner and R. Grose, “Fibroblast growth factor signalling: from development to cancer,” Nature Reviews Cancer , vol. 10, no. 2, pp. 116–129, 2010. View at Publisher · View at Google Scholar · View at Scopus D. Gospodarowicz, “Localisation of a fibroblast growth factor and its effect along and with hydrocortisone on 3T3 cell growth,” Nature , vol. 249, no. 5453, pp. 123–127, 1974. View at Scopus H. A. Armelin, “Pituitary extracts and steroid hormones in the control of 3T3 cell growth,” Proceedings of the National Academy of Sciences of the United States of America , vol. 70, no. 9, pp. 2702–2706, 1973. View at Scopus C. M. Carreira, M. Landriscina, S. Bellum, I. Prudovsky, and T. Maciag, “The comparative release of FGF1 by hypoxia and temperature stress,” Growth Factors , vol. 18, no. 4, pp. 277–285, 2001. View at Scopus A. Jackson, S. Friedman, X. Zhan, K. A. Engleka, R. Forough, and T. Maciag, “Heat shock induces the release of fibroblast growth factor 1 from NIH 3T3 cells,” Proceedings of the National Academy of Sciences of the United States of America , vol. 89, no. 22, pp. 10691–10695, 1992. View at Publisher · View at Google Scholar · View at Scopus J. T. Shin, S. R. Opalenik, J. N. Wehby et al., “Serum-starvation induces the extracellular appearance of FGF-1,” Biochimica et Biophysica Acta, Molecular Cell Research , vol. 1312, no. 1, pp. 27–38, 1996. View at Publisher · View at Google Scholar · View at Scopus N. M. Ananyeva, A. V. Tjurmin, J. A. Berliner, et al., “Oxidized LDL mediates the release of fibroblast growth factor-1,” Arteriosclerosis, Thrombosis, and Vascular Biology , vol. 17, no. 3, pp. 445–453, 1997. View at Publisher · View at Google Scholar S. K. Mohan, S. G. Rani, S. M. Kumar, and C. Yu, “S100A13-C2A binary complex structure-a key component in the acidic fibroblast growth factor for the non-classical pathway,” Biochemical and Biophysical Research Communications , vol. 380, no. 3, pp. 514–519, 2009. View at Publisher · View at Google Scholar · View at Scopus M. Landriscina, C. Bagalá, A. Mandinova et al., “Copper induces the assembly of a multiprotein aggregate implicated in the release of fibroblast growth factor 1 in response to stress,” The Journal of Biological Chemistry , vol. 276, no. 27, pp. 25549–25557, 2001. View at Publisher · View at Google Scholar · View at Scopus C. M. Carreira, T. M. LaVallee, F. Tarantini et al., “S100A13 is involved in the regulation of fibroblast growth factor-1 and p40 synaptotagmin-1 release in vitro,” The Journal of Biological Chemistry , vol. 273, no. 35, pp. 22224–22231, 1998. View at Publisher · View at Google Scholar · View at Scopus F. Tarantini, T. Lavallee, A. Jackson et al., “The extravesicular domain of synaptotagmin-1 is released with the latent fibroblast growth factor-1 homodimer in response to heat shock,” The Journal of Biological Chemistry , vol. 273, no. 35, pp. 22209–22216, 1998. View at Publisher · View at Google Scholar · View at Scopus S. Uriel, E. M. Brey, and H. P. Greisler, “Sustained low levels of fibroblast growth factor-1 promote persistent microvascular network formation,” American Journal of Surgery , vol. 192, no. 5, pp. 604–609, 2006. View at Publisher · View at Google Scholar · View at Scopus A. Iwakura, M. Fujita, M. Ikemoto et al., “Myocardial ischemia enhances the expression of acidic fibroblast growth factor in human pericardial fluid,” Heart and Vessels , vol. 15, no. 3, pp. 112–116, 2000. View at Scopus P. Cuevas, D. Reimers, F. Carceller et al., “Fibroblast growth factor-1 prevents myocardial apoptosis triggered by ischemia reperfusion injury,” European Journal of Medical Research , vol. 2, no. 11, pp. 465–468, 1997. View at Scopus J.-C. Wu, W.-C. Huang, Y.-A. Tsai, Y.-C. Chen, and H. Cheng, “Nerve repair using acidic fibroblast growth factor in human cervical spinal cord injury: a preliminary Phase I clinical study,” Journal of Neurosurgery: Spine , vol. 8, no. 3, pp. 208–214, 2008. View at Publisher · View at Google Scholar · View at Scopus X. Xia, J. P. Babcock, S. I. Blaber, et al., “Pharmacokinetic properties of 2nd-generation fibroblast growth factor-1 mutants for therapeutic application,” PLoS ONE , vol. 7, no. 11, Article ID e48210, 2012. View at Publisher · View at Google Scholar J. Belch, W. R. Hiatt, I. Baumgartner et al., “Effect of fibroblast growth factor NV1FGF on amputation and death: a randomised placebo-controlled trial of gene therapy in critical limb ischaemia,” The Lancet , vol. 377, no. 9781, pp. 1929–1937, 2011. View at Publisher · View at Google Scholar · View at Scopus A. J. Comerota, R. C. Throm, K. A. Miller et al., “Naked plasmid DNA encoding fibroblast growth factor type 1 for the treatment of end-stage unreconstructible lower extremity ischemia: preliminary results of a phase I trial,” Journal of Vascular Surgery , vol. 35, no. 5, pp. 930–936, 2002. View at Publisher · View at Google Scholar · View at Scopus B. Birrer, “Whole genome oligonucleotide-based array comparative genomic hybridization analysis identified fibroblast growth factor 1 as a prognostic marker for advanced-stage serous ovarian adenocarcinomas,” Journal of Clinical Oncology , vol. 25, no. 21, p. 3184, 2007. View at Publisher · View at Google Scholar · View at Scopus B. Kwabi-Addo, M. Ozen, and M. Ittmann, “The role of fibroblast growth factors and their receptors in prostate cancer,” Endocrine-Related Cancer , vol. 11, no. 4, pp. 709–724, 2004. View at Publisher · View at Google Scholar · View at Scopus V. P. Eswarakumar, I. Lax, and J. Schlessinger, “Cellular signaling by fibroblast growth factor receptors,” Cytokine and Growth Factor Reviews , vol. 16, no. 2, pp. 139–149, 2005. View at Publisher · View at Google Scholar · View at Scopus D. M. Ornitz and N. Itoh, “Fibroblast growth factors,” Genome Biology , vol. 2, no. 3, article 3005, 2001. View at Scopus M. Bhattacharjee, R. Raju, A. Radhakrishnan, et al., “A bioinformatics resource for TWEAK-Fn14 signaling pathway,” Journal of Signal Transduction , vol. 2012, Article ID 376470, 10 pages, 2012. View at Publisher · View at Google Scholar D. Telikicherla, A. Ambekar, S. Palapetta et al., “A comprehensive curated resource for follicle stimulating hormone signaling,” BMC Research Notes , vol. 4, article 408, 2011. View at Publisher · View at Google Scholar · View at Scopus K. Kandasamy, S. Sujatha Mohan, R. Raju et al., “NetPath: A public resource of curated signal transduction pathways,” Genome Biology , vol. 11, no. 1, article r3, 2010. View at Publisher · View at Google Scholar · View at Scopus R. Raju, V. Nanjappa, L. Balakrishnan et al., “NetSlim: high-confidence curated signaling maps,” The Journal of Biological Databases and Curation , vol. 2011, p. bar032, 2011. View at Scopus M. Manuvakhova, J. V. Thottassery, S. Hays et al., “Expression of the SNT-1/FRS2 phosphotyrosine binding domain inhibits activation of MAP kinase and PI3-kinase pathways and antiestrogen resistant growth induced by FGF-1 in human breast carcinoma cells,” Oncogene , vol. 25, no. 44, pp. 6003–6014, 2006. View at Publisher · View at Google Scholar · View at Scopus S. H. Ong, Y. R. Hadari, N. Gotoh, G. R. Guy, J. Schlessinger, and I. Lax, “Stimulation of phosphatidylinositol 3-kinase by fibroblast growth factor receptors is mediated by coordinated recruitment of multiple docking proteins,” Proceedings of the National Academy of Sciences of the United States of America , vol. 98, no. 11, pp. 6074–6079, 2001. View at Publisher · View at Google Scholar · View at Scopus Y. R. Hadari, H. Kouhara, I. Lax, and J. Schlessinger, “Binding of Shp2 tyrosine phosphatase to FRS2 is essential for fibroblast growth factor-induced PC12 cell differentiation,” Molecular and Cellular Biology , vol. 18, no. 7, pp. 3966–3973, 1998. View at Scopus H. Kouhara, Y. R. Hadari, T. Spivak-Kroizman et al., “A lipid-anchored Grb2-binding protein that links FGF-receptor activation to the Ras/MAPK signaling pathway,” Cell , vol. 89, no. 5, pp. 693–702, 1997. View at Scopus M. Kanai, M. Göke, S. Tsunekawa, and D. K. Podolsky, “Signal transduction pathway of human fibroblast growth factor receptor 3. Identification of a novel 66-kDa phosphoprotein,” The Journal of Biological Chemistry , vol. 272, no. 10, pp. 6621–6628, 1997. View at Publisher · View at Google Scholar · View at Scopus Y. R. Hadari, H. Kouhara, I. Lax, and J. Schlessinger, “Binding of Shp2 tyrosine phosphatase to FRS2 is essential for fibroblast growth factor-induced PC12 cell differentiation,” Molecular and Cellular Biology , vol. 18, no. 7, pp. 3966–3973, 1998. View at Scopus W.-F. Lin, C.-J. Chen, Y.-J. Chang, S.-L. Chen, I.-M. Chiu, and L. Chen, “SH2B1 β enhances fibroblast growth factor 1 (FGF1)-induced neurite outgrowth through MEK-ERK1/2-STAT3-Egr1 pathway,” Cellular Signalling , vol. 21, no. 7, pp. 1060–1072, 2009. View at Publisher · View at Google Scholar · View at Scopus M. Mohammadi, I. Dikic, A. Sorokin, W. H. Burgess, M. Jaye, and J. Schlessinger, “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 , vol. 16, no. 3, pp. 977–989, 1996. View at Scopus A. Willems-Widyastuti, B. M. Vanaudenaerde, R. Vos et al., “Azithromycin attenuates fibroblast growth factors induced vascular endothelial growth factor Via p38MAPK signaling in human airway smooth muscle cells,” Cell Biochemistry and Biophysics , vol. 67, no. 2, pp. 331–339, 2013. View at Publisher · View at Google Scholar · View at Scopus T. Nishida, J.-I. Ito, Y. Nagayasu, and S. Yokoyama, “FGF-1-induced reactions for biogenesis of apoE-HDL are mediated by Src in rat astrocytes,” Journal of Biochemistry , vol. 146, no. 6, pp. 881–886, 2009. View at Publisher · View at Google Scholar · View at Scopus C. W. Chen, C. S. Liu, I. M. Chiu, et al., “The signals of FGFs on the neurogenesis of embryonic stem cells,” Journal of Biomedical Science , vol. 17, p. 33, 2010. View at Publisher · View at Google Scholar G. Lungu, L. Covaleda, O. Mendes, H. Martini-Stoica, and G. Stoica, “FGF-1-induced matrix metalloproteinase-9 expression in breast cancer cells is mediated by increased activities of NF-kappa;B and activating protein-1,” Molecular Carcinogenesis , vol. 47, no. 6, pp. 424–435, 2008. View at Publisher · View at Google Scholar · View at Scopus F. S. Newell, H. Su, H. Tornqvist, J. P. Whitehead, J. B. Prins, and L. J. Hutley, “Characterization of the transcriptional and functional effects of fibroblast growth factor-1 on human preadipocyte differentiation,” The FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology , vol. 20, no. 14, pp. 2615–2617, 2006. View at Publisher · View at Google Scholar · View at Scopus D. R. Newman, C.-M. Li, R. Simmons, J. Khosla, and P. L. Sannes, “Heparin affects signaling pathways stimulated by fibroblast growth factor-1 and -2 in type II cells,” American Journal of Physiology, Lung Cellular and Molecular Physiology , vol. 287, no. 1, pp. L191–L200, 2004. View at Publisher · View at Google Scholar · View at Scopus A. Buehlera, A. Martirea, C. Strohma, et al., “Angiogenesis-independent cardioprotection in FGF-1 transgenic mice,” Cardiovascular Research , vol. 55, no. 4, pp. 768–777, 2002. View at Publisher · View at Google Scholar C. A. Castaneda, H. Cortes-Funes, H. L. Gomez, and E. M. Ciruelos, “The phosphatidyl inositol 3-kinase/AKT signaling pathway in breast cancer,” Cancer Metastasis Reviews , vol. 29, no. 4, pp. 751–759, 2010. View at Scopus J.-I. Ito, Y. Nagayasu, K. Okumura-Noji et al., “Mechanism for FGF-1 to regulate biogenesis of apoE-HDL in astrocytes,” Journal of Lipid Research , vol. 48, no. 9, pp. 2020–2027, 2007. View at Publisher · View at Google Scholar · View at Scopus R. Forough, B. Weylie, C. Patel, S. Ambrus, U. S. Singh, and J. Zhu, “Role of AKT/PKB signaling in fibroblast growth factor-1 (FGF-1)-induced angiogenesis in the chicken chorioallantoic membrane (CAM),” Journal of Cellular Biochemistry , vol. 94, no. 1, pp. 109–116, 2005. View at Publisher · View at Google Scholar · View at Scopus J. Wang, T. Ito, N. Udaka, K. Okudela, T. Yazawa, and H. Kitamura, “PI3K-AKT pathway mediates growth and survival signals during development of fetal mouse lung,” Tissue and Cell , vol. 37, no. 1, pp. 25–35, 2005. View at Publisher · View at Google Scholar · View at Scopus W.-F. Lin, C.-J. Chen, Y.-J. Chang, S.-L. Chen, I.-M. Chiu, and L. Chen, “SH2B1 β enhances fibroblast growth factor 1 (FGF1)-induced neurite outgrowth through MEK-ERK1/2-STAT3-Egr1 pathway,” Cellular Signalling , vol. 21, no. 7, pp. 1060–1072, 2009. View at Publisher · View at Google Scholar · View at Scopus M. A. Hossain, J. C. Russell, R. Gomes, and J. Laterra, “Neuroprotection by scatter factor/hepatocyte growth factor and FGF-1 in cerebellar granule neurons is phosphatidylinositol 3-kinase/Akt-dependent and MAPK/CREB-independent,” Journal of Neurochemistry , vol. 81, no. 2, pp. 365–378, 2002. View at Publisher · View at Google Scholar · View at Scopus P. Li, S. Oparil, W. Feng, and Y.-F. Chen, “Hypoxia-responsive growth factors upregulate periostin and osteopontin expression via distinct signaling pathways in rat pulmonary arterial smooth muscle cells,” Journal of Applied Physiology , vol. 97, no. 4, pp. 1550–1558, 2004. View at Publisher · View at Google Scholar · View at Scopus A. Raucci, E. Laplantine, A. Mansukhani, and C. Basilico, “Activation of the ERK1/2 and p38 mitogen-activated protein kinase pathways mediates fibroblast growth factor-induced growth arrest of chondrocytes,” The Journal of Biological Chemistry , vol. 279, no. 3, pp. 1747–1756, 2004. View at Publisher · View at Google Scholar · View at Scopus J. Jiao, J. S. Greendorfer, P. Zhang, K. R. Zinn, C. A. Diglio, and J. A. Thompson, “Alternatively spliced FGFR-1 isoform signaling differentially modulates endothelial cell responses to peroxynitrite,” Archives of Biochemistry and Biophysics , vol. 410, no. 2, pp. 187–200, 2003. View at Publisher · View at Google Scholar · View at Scopus P. B. Mehta, C. N. Robson, D. E. Neal, and H. Y. Leung, “Keratinocyte growth factor activates p38 MAPK to induce stress fibre formation in human prostate DU145 cells,” Oncogene , vol. 20, no. 38, pp. 5359–5365, 2001. View at Publisher · View at Google Scholar · View at Scopus Y. J. Chang, K. W. Chen, C. J. Chen, et al., “SH2B1 β interacts with STAT3 and enhances fibroblast growth factor 1-induced gene expression during neuronal differentiation,” Molecular and Cellular Biology , vol. 34, no. 6, pp. 1003–1019, 2014. A. A. Dudka, S. M. M. Sweet, and J. K. Heath, “Signal transducers and activators of transcription-3 binding to the fibroblast growth factor receptor is activated by receptor amplification,” Cancer Research , vol. 70, no. 8, pp. 3391–3401, 2010. View at Publisher · View at Google Scholar · View at Scopus W.-F. Lin, C.-J. Chen, Y.-J. Chang, S.-L. Chen, I.-M. Chiu, and L. Chen, “SH2B1 β enhances fibroblast growth factor 1 (FGF1)-induced neurite outgrowth through MEK-ERK1/2-STAT3-Egr1 pathway,” Cellular Signalling , vol. 21, no. 7, pp. 1060–1072, 2009. View at Publisher · View at Google Scholar · View at Scopus G. Lungu, L. Covaleda, O. Mendes, H. Martini-Stoica, and G. Stoica, “FGF-1-induced matrix metalloproteinase-9 expression in breast cancer cells is mediated by increased activities of NF-kappa;B and activating protein-1,” Molecular Carcinogenesis , vol. 47, no. 6, pp. 424–435, 2008. View at Publisher · View at Google Scholar · View at Scopus X. Zhu, J. Sasse, D. McAllister, et al., “Evidence that fibroblast growth factors 1 and 4 participate in regulation of cardiogenesis,” Developmental Dynamics , vol. 207, no. 4, pp. 429–438, 1996. X. Qu, K. Hertzler, Y. Pan, K. Grobe, M. L. Robinson, and X. Zhang, “Genetic epistasis between heparan sulfate and FGF-Ras signaling controls lens development,” Developmental Biology , vol. 355, no. 1, pp. 12–20, 2011. View at Publisher · View at Google Scholar · View at Scopus A. E. Serls, S. Doherty, P. Parvatiyar, J. M. Wells, and G. H. Deutsch, “Different thresholds of fibroblast growth factors pattern the ventral foregut into liver and lung,” Development , vol. 132, no. 1, pp. 35–47, 2005. View at Publisher · View at Google Scholar · View at Scopus D. Lebeche, S. Malpel, and W. V. Cardoso, “Fibroblast growth factor interactions in the developing lung,” Mechanisms of Development , vol. 86, no. 1-2, pp. 125–136, 1999. View at Publisher · View at Google Scholar · View at Scopus J. Jung, M. Zheng, M. Goldfarb, and K. S. Zaret, “Initiation of mammalian liver development from endoderm by fibroblast growth factors,” Science , vol. 284, no. 5422, pp. 1998–2003, 1999. View at Publisher · View at Google Scholar · View at Scopus X. Luo, L. J. Hutley, J. A. Webster et al., “Identification of BMP and activin membrane-bound inhibitor (BAMBI) as a potent negative regulator of adipogenesis and modulator of autocrine/paracrine adipogenic factors,” Diabetes , vol. 61, no. 1, pp. 124–136, 2012. View at Publisher · View at Google Scholar · View at Scopus L. Hutley, W. Shurety, F. Newell et al., “Fibroblast growth factor 1: a key regulator of human adipogenesis,” Diabetes , vol. 53, no. 12, pp. 3097–3106, 2004. View at Publisher · View at Google Scholar · View at Scopus T. Tran, V. Kolupaeva, and C. Basilico, “FGF inhibits the activity of the cyclin B1/CDK1 kinase to induce a transient G 2 arrest in RCS chondrocytes,” Cell Cycle , vol. 9, no. 21, pp. 4379–4386, 2010. View at Publisher · View at Google Scholar · View at Scopus V. Kolupaeva, E. Laplantine, and C. Basilico, “PP2A-mediated dephosphorylation of p107 plays a critical role in chondrocyte cell cycle arrest by FGF,” PLoS ONE , vol. 3, no. 10, Article ID e3447, 2008. View at Publisher · View at Google Scholar · View at Scopus R. Priore, L. Dailey, and C. Basilico, “Downregulation of Akt activity contributes to the growth arrest induced by FGF in chondrocytes,” Journal of Cellular Physiology , vol. 207, no. 3, pp. 800–808, 2006. View at Publisher · View at Google Scholar · View at Scopus L. Dailey, E. Laplantine, R. Priore, and C. Basilico, “A network of transcriptional and signaling events is activated by FGF to induce chondrocyte growth arrest and differentiation,” Journal of Cell Biology , vol. 161, no. 6, pp. 1053–1066, 2003. View at Publisher · View at Google Scholar · View at Scopus P. Zhang, J. S. Greendorfer, J. Jiao, S. C. Kelpke, and J. A. Thompson, “Alternatively spliced FGFR-1 isoforms differentially modulate endothelial cell activation of c-YES,” Archives of Biochemistry and Biophysics , vol. 450, no. 1, pp. 50–62, 2006. View at Publisher · View at Google Scholar · View at Scopus B. Fernandez, A. Buehler, S. Wolfram et al., “Transgenic myocardial overexpression of fibroblast growth factor-1 increases coronary artery density and branching,” Circulation Research , vol. 87, no. 3, pp. 207–213, 2000. View at Scopus B. Schumacher, P. Peecher, B. U. Von Specht, and T. Stegmann, “Induction of neoangiogenesis in ischemic myocardium by human growth factors: first clinical results of a new treatment of coronary heart disease,” Circulation , vol. 97, no. 7, pp. 645–650, 1998. View at Scopus J.-I. Ito, Y. Nagayasu, R. Lu, A. Kheirollah, M. Hayashi, and S. Yokoyama, “Astrocytes produce and secrete FGF-1, which promotes the production of apoE-HDL in a manner of autocrine action,” Journal of Lipid Research , vol. 46, no. 4, pp. 679–686, 2005. View at Publisher · View at Google Scholar · View at Scopus L. Sun, L. Xu, H. Chang et al., “Transfection with aFGF cDNA improves wound healing,” Journal of Investigative Dermatology , vol. 108, no. 3, pp. 313–318, 1997. View at Scopus U. Pirvola, Y. Cao, C. Oellig, Z. Suoqiang, R. F. Pettersson, and J. Ylikoski, “The site of action of neuronal acidic fibroblast growth factor is the organ of corti of the rat cochlea,” Proceedings of the National Academy of Sciences of the United States of America , vol. 92, no. 20, pp. 9269–9273, 1995. View at Publisher · View at Google Scholar · View at Scopus M. Palmen, M. J. A. P. Daemen, L. J. De Windt et al., “Fibroblast growth factor-1 improves cardiac functional recovery and enhances cell survival after ischemia and reperfusion: a fibroblast growth factor receptor, protein kinase C, and tyrosine kinase-dependent mechanism,” Journal of the American College of Cardiology , vol. 44, no. 5, pp. 1113–1123, 2004. View at Publisher · View at Google Scholar · View at Scopus A. Buehler, A. Martire, C. Strohm et al., “Angiogenesis-independent cardioprotection in FGF-1 transgenic mice,” Cardiovascular Research , vol. 55, no. 4, pp. 768–777, 2002. View at Publisher · View at Google Scholar · View at Scopus P. Htun, W. D. Ito, I. E. Hoefer, J. Schaper, and W. Schaper, “Intramyocardial infusion of FGF-1 mimics ischemic preconditioning in pig myocardium,” Journal of Molecular and Cellular Cardiology , vol. 30, no. 4, pp. 867–877, 1998. View at Publisher · View at Google Scholar · View at Scopus M. Hashimoto, Y. Sagara, D. Langford et al., “Fibroblast growth factor 1 regulates signaling via the glycogen synthase kinase-3 β pathway. Implications for neuroprotection,” The Journal of Biological Chemistry , vol. 277, no. 36, pp. 32985–32991, 2002. View at Publisher · View at Google Scholar · View at Scopus J. Taeger, C. Moser, C. Hellerbrand et al., “Targeting FGFR/PDGFR/VEGFR impairs tumor growth, angiogenesis, and metastasis by effects on tumor cells, endothelial cells, and pericytes in pancreatic cancer,” Molecular Cancer Therapeutics , vol. 10, no. 11, pp. 2157–2167, 2011. View at Publisher · View at Google Scholar · View at Scopus E. M. Haugsten, M. Zakrzewska, A. Brech et al., “Clathrin- and dynamin-independent endocytosis of fgfr3—implications for signalling,” PLoS ONE , vol. 6, no. 7, Article ID e21708, 2011. View at Publisher · View at Google Scholar · View at Scopus C. Bonneton, J. B. Sibarita, and J. P. Thiery, “Relationship between cell migration and cell cycle during the initiation of epithelial to fibroblastoid transition,” Cell Motility and the Cytoskeleton , vol. 43, no. 4, pp. 288–295, 1999. Z. Liu, Y. E. Hartman, J. M. Warram et al., “Fibroblast growth factor receptor mediates fibroblast-dependent growth in EMMPRIN-depleted head and neck cancer tumor cells,” Molecular Cancer Research , vol. 9, no. 8, pp. 1008–1017, 2011. View at Publisher · View at Google Scholar · View at Scopus N. R. Estes II, J. V. Thottassery, L. Westbrook, and F. G. Kern, “MEK ablation in MCF-7 cells blocks DNA synthesis induced by serum, but not by estradiol or growth factors,” International Journal of Oncology , vol. 29, no. 6, pp. 1573–1580, 2006. View at Scopus O. Klingenberg, A. Wiçdłocha, A. Rapak, R. Muñoz, P. Ø. Falnes, and S. Olsnes, “Inability of the acidic fibroblast growth factor mutant K132E to stimulate DNA synthesis after translocation into cells,” The Journal of Biological Chemistry , vol. 273, no. 18, pp. 11164–11172, 1998. View at Publisher · View at Google Scholar · View at Scopus J.-M. Rodier, A. M. Valles, M. Denoyelle, J. P. Thiery, and B. Boyer, “pp60(c-src) Is a positive regulator of growth factor-induced cell scattering in a rat bladder carcinoma cell line,” Journal of Cell Biology , vol. 131, no. 3, pp. 761–773, 1995. View at Publisher · View at Google Scholar · View at Scopus A. M. Valles, B. Boyer, J. Badet, G. C. Tucker, D. Barritault, and J. P. Thiery, “Acidic fibroblast growth factor is a modulator of epithelial plasticity in a rat bladder carcinoma cell line,” Proceedings of the National Academy of Sciences of the United States of America , vol. 87, no. 3, pp. 1124–1128, 1990. View at Scopus J. Jouanneau, J. Plouet, G. Moens, and J. P. Thiery, “FGF-2 and FGF-1 expressed in rat bladder carcinoma cells have similar angiogenic potential but different tumorigenic properties in vivo,” Oncogene , vol. 14, no. 6, pp. 671–676, 1997. View at Scopus J. Jouanneau, J. Gavrilovic, D. Caruelle et al., “Secreted or nonsecreted forms of acidic fibroblast growth factor produced by transfected epithelial cells influence cell morphology, motility, and invasive potential,” Proceedings of the National Academy of Sciences of the United States of America , vol. 88, no. 7, pp. 2893–2897, 1991. View at Scopus P. Shannon, A. Markiel, O. Ozier et al., “Cytoscape: a software environment for integrated models of biomolecular interaction networks,” Genome Research , vol. 13, no. 11, pp. 2498–2504, 2003. View at Publisher · View at Google Scholar · View at Scopus O. Babur, U. Dogrusoz, E. Demir, and C. Sander, “ChiBE: interactive visualization and manipulation of BioPAX pathway models,” Bioinformatics , vol. 26, no. 3, pp. 429–431, 2010. View at Scopus A. Dilek, M. E. Belviranli, and U. Dogrusoz, “VISIBIOweb: visualization and layout services for BioPAX pathway models,” Nucleic Acids Research , vol. 38, no. 2, Article ID gkq352, pp. W150–W154, 2010. View at Publisher · View at Google Scholar · View at Scopus M. P. van Iersel, T. Kelder, A. R. Pico et al., “Presenting and exploring biological pathways with PathVisio,” BMC Bioinformatics , vol. 9, article 399, 2008. View at Publisher · View at Google Scholar · View at Scopus T. S. K. Prasad, R. Goel, K. Kandasamy et al., “Human protein reference database—2009 update,” Nucleic Acids Research , vol. 37, no. 1, pp. D767–D772, 2009. View at Publisher · View at Google Scholar · View at Scopus var _gaq = _gaq || []; _gaq.push(['_setAccount', 'UA-8578054-2']); _gaq.push(['_trackPageview']); (function () { var ga = document.createElement('script'); ga.type = 'text/javascript'; ga.async = true; ga.src = ('https:' == document.location.protocol ? 'https://ssl' : 'http://www') + '.google-analytics.com/ga.js'; var s = document.getElementsByTagName('script')[0]; s.parentNode.insertBefore(ga, s); })();

Journal

Journal of Signal TransductionHindawi Publishing Corporation

Published: Apr 16, 2014

There are no references for this article.