OPEN ACCESS JOURNAL AT INIST-CNRS Gene...

20
Gene Section Review Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 821 Atlas of Genetics and Cytogenetics in Oncology and Haematology OPEN ACCESS JOURNAL AT INIST-CNRS FGFR1 (Fibroblast Growth Factor Receptor 1) Jean-Loup Huret Genetics, Dept Medical Information, University of Poitiers, CHU Poitiers Hospital, F-86021 Poitiers, France (JLH) Published in Atlas Database: December 2008 Online updated version : http://AtlasGeneticsOncology.org/Genes/FGFR1113.html DOI: 10.4267/2042/44609 This article is an update of : Pébusque MJ. FGFR1 (Fibroblast Growth Factor Receptor 1). Atlas Genet Cytogenet Oncol Haematol 2001;5(1):9-10 HuretJL. FGFR1 (Fibroblast Growth Factor Receptor 1). Atlas Genet Cytogenet Oncol Haematol 1998;2(2):35 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2009 Atlas of Genetics and Cytogenetics in Oncology and Haematology Identity Other names: BFGFR (basic fibroblast growth factor receptor); FLT2 (FMS-like tyrosine kinase 2); FGFBR (FGFb receptor); FLG (FMS-like gene); CEK (chicken embryo kinase); CD331 (CD331 antigen); H2 (heparin- binding growth factor receptor); KAL2 (Kallmann); N- SAM (NCC-IT-cell derived stomach cancer amplified gene) HGNC (Hugo): FGFR1 Location: 8p12 DNA/RNA FGFR1 (8p12) - Courtesy Mariano Rocchi, Resources for Molecular Cytogenetics. Fig 1. FGFR1 (8p12) gene and protein. FGFR1 comprises an extracellular domain, a transmembrane domain, and an intracellular domain. The extracellular domain contains a signal peptide (SP), two (IgII, IgIII) or three (IgI, IgII, and IgIII) Ig-like domains; it is followed by the transmembrane domain (TM), and an intracellular domain comprising a juxtamembrane domain (JM), two tyrosine kinase subdomains (TK1,TK2) interrupted by a short kinase insert (KI), and a C-terminal tail (CT). The acidic box (AB) indicated in the extracellular domain is a specific feature of FGF receptors; a CHD (cell adhesion molecule (CAM) homogogy domain) and a HB domain (binding domain for heparin or heparin sulfate proteoglycan HSPG) are present in the extracellular part. A NB (nuclear binding domain), and the PLC site (interaction with PLC gamma) are found in the cytosolic part of FGFR1.

Transcript of OPEN ACCESS JOURNAL AT INIST-CNRS Gene...

Page 1: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

Gene Section Review

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 821

Atlas of Genetics and Cytogenetics in Oncology and Haematology

OPEN ACCESS JOURNAL AT INIST-CNRS

FGFR1 (Fibroblast Growth Factor Receptor 1) Jean-Loup Huret

Genetics, Dept Medical Information, University of Poitiers, CHU Poitiers Hospital, F-86021 Poitiers, France (JLH)

Published in Atlas Database: December 2008

Online updated version : http://AtlasGeneticsOncology.org/Genes/FGFR1113.html DOI: 10.4267/2042/44609

This article is an update of : Pébusque MJ. FGFR1 (Fibroblast Growth Factor Receptor 1). Atlas Genet Cytogenet Oncol Haematol 2001;5(1):9-10 HuretJL. FGFR1 (Fibroblast Growth Factor Receptor 1). Atlas Genet Cytogenet Oncol Haematol 1998;2(2):35 This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 2.0 France Licence. © 2009 Atlas of Genetics and Cytogenetics in Oncology and Haematology

Identity Other names: BFGFR (basic fibroblast growth factor receptor); FLT2 (FMS-like tyrosine kinase 2); FGFBR (FGFb receptor); FLG (FMS-like gene); CEK (chicken embryo kinase); CD331 (CD331 antigen); H2 (heparin-binding growth factor receptor); KAL2 (Kallmann); N-SAM (NCC-IT-cell derived stomach cancer amplified gene)

HGNC (Hugo): FGFR1

Location: 8p12

DNA/RNA

FGFR1 (8p12) - Courtesy Mariano Rocchi, Resources for

Molecular Cytogenetics.

Fig 1. FGFR1 (8p12) gene and protein. FGFR1 comprises an extracellular domain, a transmembrane domain, and an intracellular domain. The extracellular domain contains a signal peptide (SP), two (IgII, IgIII) or three (IgI, IgII, and IgIII) Ig-like domains; it is followed by the transmembrane domain (TM), and an intracellular domain comprising a juxtamembrane domain (JM), two tyrosine kinase subdomains (TK1,TK2) interrupted by a short kinase insert (KI), and a C-terminal tail (CT). The acidic box (AB) indicated in the extracellular domain is a specific feature of FGF receptors; a CHD (cell adhesion molecule (CAM) homogogy domain) and a HB domain (binding domain for heparin or heparin sulfate proteoglycan HSPG) are present in the extracellular part. A NB (nuclear binding domain), and the PLC site (interaction with PLC gamma) are found in the cytosolic part of FGFR1.

Page 2: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 822

Fig 2. FGFR1 splice forms. Showing soluble forms (A, B) and membrane-attached forms (C, D, E); forms IIIa (B), IIIb (C), and IIIc (D, E), according to the 3' part of the third Ig-like domain; forms alpha (C, D) with 3 Ig-like domains, and beta (E), with only 2. Exons are shown on the top right (forms a, b, c, d, and e). Adapted from Johnson and Williams 1993, and SpliceCenter data.

Description The DNA has been cloned in 1989 (Lee et al., 1989).

Transcription The gene spans 57,697 bases on reverse strand; there are numerous splicing forms, with mRNAs from about 3 kb to 5.688 kb for the canonical form, made of 18 exons. 11 transcripts for Ensembl, 18 transcripts for Swiss-Prot. Different transcripts are expressed in different cells and tissues. Some isoforms are truncated in the extracellular domain (IgI, in particular, is often missing), others in the intracellular domain (TK2 missing) or both. Secreted forms lack the transmembrane domain; TK truncated forms are kinase-deficient. Exon numbering is variable.

Protein Description The canonical form comprises 822 amino acids; 100-135 kDa glycoprotein from a 90-115 kDa protein core. FGFR1 is composed of an extracellular ligand-binding domain, a unique transmembrane domain, and a catalytic (tyrosine kinase) cytosolic domain. The extracellular region (with the N-terminus) contains a signal peptide (amino acids (aa) 1-21), 3 immunoglobulin-like loops (aa: 25-119, 158-246, and 255-357 according to Swiss-Prot), with an acidic box (seven glutamic acids in 127-133) between IgI and IgII, a CHD (Cell adhesion molecule (CAM) homogogy domain) in 151-170, and an heparin-binding domain in

the begining of the IgII (aa 166-177); this extracellular region is followed by a transmembrane domain (aa 377-397) and an intracellular domain composed of a juxtamembrane domain, which serves as a binding site for phosphotyrosine binding (PTB) domains of proteins such as FRS2 (see below), and a tyrosine kinase domain (aa 478-767) (two kinase domains interrupted by a short kinase insert of 14 amino acids), and a C-terminal tail. A part of the C-terminal tail of FGFR1 containing 28 amino acids binds the SH2 domain of PLCG (PLC gamma). Phosphorylation of Tyrosine 766 is the interactive item with PLC gamma (Itoh et al., 1990; Johnson and Williams, 1993) (Fig 1). A triad of residues N546, E562, and K638 would form a network of hydrogen bounds acting as a molecular brake keeping the kinase in an autoinhibited state (Chen et al., 2007). Details on crystal structure can be found in Eswarakumar et al., 2005. The five tyrosine autophosphorylation sites in the catalytic core of FGFR1 are phosphorylated by a precisely controlled and ordered reaction. The rate of catalysis of two substrates is enhanced by 50- and 500-fold after autophosphorylation of Y653 and Y654, respectively, in the activation loop of FGFR1 (Furdui et al., 2006).

FGFR1 splice variants - isoforms Soluble forms/forms without TK activity: Splice variants generate isoforms with different ligand-binding specificities, and differently expressed in different cells and tissues. Some isoforms are truncated in the extracellular domain, lacking Ig-like domains, other in the intracellular domain, lacking the kinase

Page 3: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 823

domain, or both. They are soluble forms if the transmembrane domain is missing. However, if soluble forms can result from variant splicing, they can also result from proteolytic cleavage (Groth and Lardelli, 2002). A short transcript ending 32 amino acids after the first Ig-like domain (See Fig 2A) has been found. It is reminiscent of similar forms of N-CAM (Johnson and Williams, 1993). Variants lacking parts of the TK domain(s) may act as down regulators of the signal, since they can heterodimerize with active forms, leading to non-functionnal dimer receptors. FGFR1 IIIa/ FGFR1 IIIb/ FGFR1 IIIc: Three possible variants of Ig-like domain III are known for FGFR1, FGFR2, and FGFR3 (Johnson et al., 1991). FGFR1 IIIa (Fig 2B) ends downstream of IgIII-like domain. It is a secreted receptor devoid of signaling capacity. It is expressed in adult tissues and in cell lines. It may be essential during mesoderm induction. FGFR1 IIIa binds FGF2 with a higher affinity than FGF1, contrarity to FGFR IIIb. FGFR IIIc binds both with equal affinity. FGFR1 IIIb (Fig 2C) is expressed in epithelial lineages, FGFR1 IIIc (Fig 2D and E; compare the exons with form IIIb) in mesenchymal lineages. FGFR1 IIIc is also preferentially expressed in cell lines. Whereas FGFR1 IIIc was found expressed in nearly all tissues examined, FGFR1 IIIa was found expresssed in brain, muscle and skin, and FGFR IIIb in skin, brain, kidney, muscle and placenta (Johnson and Williams, 1993). Changes in FGFR isoform expression seem to regulate tumorigenesis and malignant transformation (Liu et al., 2007). Targeted disruption of FGFR1 IIIc provoked mouse embryo lethality due to a defect in cell migration through the primitive streack, whereas FGFR1 IIIb deficient mouse embryo showed no obvious defect (reviewed in Eswarakumar et al., 2005). FGFR1alpha/FGFR1beta: FGFR1 IIIb and FGFR1 IIIc can have 3 Ig-like domains (called FGFR1alpha), or, in case IgI (encoded by exon 3, also called "the alpha-exon") is deleted by splicing, only 2 Ig like domains (FGFR1beta) (ex Fig 2D versus 2E). Two intronic silencing sequence (ISS) elements flank the alternatively spliced alpha-exon. These ISS appear to regulate the alpha-exon exclusion. The splicing is at least partly regulated by SFPQ (1p34, splicing factor proline/glutamine-rich). Note on SFPQ: see the glioblastoma section herein below. Almost all tissues contain both forms of FGFR1. The first Ig domain may be deleted without ligand binding consequence. However, it's splicing is tissue-dependant: the FGFR1 with 3 Ig like domains (FGFR1alpha) is predominant during mouse embryogenesis, while the form with only 2 Ig like domains (FGFR1beta) is equally expressed after birth in most tissues. FGFR1beta exhibits a 10-fold higher affinity for FGF1 and FGF2 than FGFR1alpha. Only FGFR1alpha has been found in the nucleus.

Aberrant splicing of the alpha-exon has been associated with pancreatic cancer, breast cancer, and glioblastoma (Bruno et al., 2004).

Receptor specificity FGFs (fibroblast growth factors): The various FGFR1 isoforms have different affinities for FGFs (Table 1); however, the only FGFs that FGFR1 binds with high affinity are FGF1 (alias aFGF: acidic FGF) and FGF2 (alias bFGF: basic FGF). Heparan sulfate proteoglycans (HSPG) may play an important role in the modulation of the different FGFs binding affinities. HSPG are polysaccharides with various additions of sulfate groups which may be critical for FGF-FGFRs affinities (Fig 3). In a given cell at a given state of proliferation/differentiation, a given HSPG would favour a given FGF-FGFR binding and signaling. TABLE I: FGFs and targets FGFRs (from Zang et al., 2006)

FGF subfamilies FGF FGFR

FGF1 (secreted or intracellular)

FGF1

FGF2

all FGFRs

FGFR 1c, 3c > 2c, 1b, 4∆

FGF4 (secreted) FGF4, FGF5, FGF6

FGFR 1c, 2c > 3c, 4∆

FGF7 (secreted)

FGF3, FGF7, FGF10, FGF22

FGFR 2b > 1b

FGF8 (secreted) FGF8, FGF17, FGF18

FGFR 3c > 4∆, > 2c > 1c > 3b

FGF9 (secreted) FGF9, FGF16, FGF20

FGFR 3c > 2c > 1c, 3b > 4∆

FGF19 (secreted) FGF19, FGF21, FGF23

FGFR 1c, 2c, 3c, 4∆ (weak)

FGF11 (intracellular)

FGF11, FGF12, FGF13, FGF14

Not known

Cell adhesion molecules: FGFR1 possesses a CHD (Cell adhesion molecule (CAM) homology domain). Cell adhesion molecules L1CAM (Xq28; L1 cell adhesion molecule), NCAM1 and NCAM2 (11q23 and 21q21; neural cell adhesion molecules) and the members of the vast cadherin family are transmembrane receptors that maintain adhesion between epithelial cells. FGF1 and FGF2 induce the internalization of surface FGFR1 and surface CDH1 (16q22; alias E-cadherin) in an endosome before

Page 4: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 824

nuclear import into the nucleus (Bryant et al., 2005). CDH11 (16q21; cadherin 11) and FGFR1 can interact directly through their extracellular domains. The neuronal cell adhesion molecule L1CAM also interacts directly with FGFR1. L1CAM promotes axonal outgrowth through an interaction with FGFR1. The extracellular domain of L1CAM binds to the combined second and third Ig-like domains of FGFR1. Activation of FGFR1 is both necessary and sufficient to account for the ability of CAMs to stimulate axonal growth. PLC gamma (see below) is the downstream effector of this response (Doherty and Walsh, 1996; Saffell et al., 1997; Boscher and Mege, 2008; Kulahin et al., 2008).

Fig 3. FGF and FGFR1 signaling from the membrane: short version ... FGF, FGFR and heparan sulfate proteoglycans (HSPG) interact with each other, form a complex. Activation of the FGF-FGFR complex recruits FRS2, PTPN11, GRB2, SOS and GAB1, and also PLCG. The signal further implicates the RAS/RAF/MAPK pathway, the PI3K/AKT/mTOR pathway, the PLC gamma pathway, the JAK/STAT pathway, and the IKK/NF-KB pathway.

Expression FGFR1, 2 and 3 have distinct expresssion patterns but with overlaps in some tissues (Zhang et al., 2006). FGFR1 and FGFR2 exhibit broad expression in the embryo and latter in life, in contrast with other FGFRs. FGFR1 is predominantly expressed in the brain and in mesenchymal tissues in the embryo, in brain, bone, kidney, skin, lung, heart and muscle in the adulte, but

not in liver (Johnson and Williams, 1993) Note: expression of the various isoforms of FGFR1 is shortly described herein above.

Localisation Plasma membrane, indeed, but also cytosol and nucleus (see figure next page).

Function Shortly: Fibroblast growth factors receptors are a family of four receptor tyrosine kinases, named FGFR1 to 4. Each recognizes a unique subset of the fibroblast growth factors (FGF) family of ligands (Table 1). There are 22 FGF, from FGF1 to FGF23, with no FGF number 15 (see Ornitz and Itoh, 2001). The first family of FGFs consists of FGF1 and FGF2; they can either be secreted or remain intracellular (they possess a nuclear localization signal). The second and third families comprise FGF3-10 and 16-23 and are secreted; the last family (FGF11-14) remain intracellular, they do not bind FGFRs; they are called FHFs (FGF homologous factors). FGFs bind FGFRs to regulate cell growth, migration and differentiation during embryogenesis, and homeostasis later in life. They act both in mesenchymal and epithelial cells. The ligand-receptor complex, in association with heparin or heparan sulfate proteoglycan (FGF and FGFR both have heparan-binding sites), induces receptor dimerization. The FGFR1 signaling is achieved by receptor conformational changes upon ligand binding, leading to dimerization and subsequent activation by autophosphorylation of TK intracellular domains. It then activates the MAP (mitogen-activated protein) kinase pathway, necessary for cell cycle progression. However, FGFR1 dimerization leads to the activation of a number of other signaling molecules including the PI3K/AKT/mTOR pathway, the phospholipase Cgamma (PLCgamma) pathway, the JAK-STAT pathway, and, more indirectly, the IKK/NF-KB pathway (Fig 3). All these pathways interact with other, as is 'summarized' Figure 4. These functions depend on: 1- the expression of one or multiple FGF and FGF receptors, 2- the various and numerous splicings of the FGFR mRNA, 3- the cell and tissue involved (review in Naski and Ornitz, 1998; Ornitz and Itoh, 2001). FGF-FGFR binding Surface cell heparin or heparan sulfate proteoglycans (HSPG) interact with FGF to induce growth factor polymerization, binding to FGFR and subsequent dimerization of FGFRs. It is essential for the dimerization and activation of the FGF-FGFR complex.

Page 5: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 825

Fig 4. FGF and FGFR1 signaling from the membrane AKT (v-akt murine thymoma viral oncogene homologs 1, 2, 3) (alias: PKB): AKT1 (14q32); AKT2 (19q13); AKT3 (1q44) (Altomare and Testa, 2007); - BAD (11q13) (BCL2-antagonist of cell death); - CASP9 (1p36) (caspase 9, apoptosis-related cysteine peptidase) (Bartolomeo and Cecconi, 2006); - CBL (11q23) (Cas-Br-M (murine) ecotropic retroviral transforming sequence) (Dikic and Schmidt, 2007); - CCND (cyclin D): CCND1 (11q13); CCND2 (12p13); CCND3 (6p21); - CDKN1A (6p21) (cyclin-dependent kinase inhibitor 1A) (Javelaud and Besancon, 2001); - CREBBP (16p13) (CREB binding protein (Rubinstein-Taybi syndrome)) (alias: CBP) (Kalkhoven, 2004); - ELK1 (Xp11) (ELK1, member of ETS oncogene family); - FOS (v-fos FBJ murine osteosarcoma viral oncogene homolog) and FOS-like antigen): FOS (14q24); FOSB (19q13); FOSL1 (11q13); FOSL2 (2p23) (Zenz, 2008); - FOXO (forkhead box O): FOXO1 (13q14); FOXO3 (6q21); FOXO4 (Xq13); FOXO6 (1p34) (Salih and Brunet, 2008); - FRAP1 (1p36) (alias mTOR) (Altomare and Testa, 2008); - FRS2 (12q15) (fibroblast growth factor receptor substrate 2) (Gotoh, 2008); - GAB1 (4q31) (GRB2-associated binding protein 1); - GRB2 (17q25) (growth factor receptor-bound protein 2) (Athauda and Bottaro, 2007); - INSR (19p13) (insulin receptor); - JAK (janus kinase (a protein tyrosine kinase)): JAK1 (1p31); JAK2 (9p24); JAK3 (19p13); TYK2 (19p13) (tyrosine kinase 2) (Murray, 2007; Shi and Amin, 2007; Steelman et al., 2008); - MAPK (mitogen-activated protein kinase ): MAPK1 (22q11) (alias: ERK2); MAPK3 (16p11) (alias: ERK1) (Seger and Krebs, 1995; Chang and Karin, 2001; Pearson et al., 2001); - MAP2K (mitogen-activated protein kinase kinase 1): MAP2K1 (15q22) (alias: MAPKK1, MEK1); MAP2K2 (19p13) (alias: MAPKK2, MEK2) (Seger and Krebs, 1995; Chang and Karin, 2001; Pearson et al., 2001); - MDM2 (12q15) (transformed mouse 3T3 cell double minute 2, p53 binding protein) (Duan and Villalona-Calero, 2006); - MYC (8q24) (v-myc myelocytomatosis viral oncogene homolog (avian)); - PDPK1 (16p13) (3-phosphoinositide dependent protein kinase-1) (alias: PDK1) (Dempsey et al., 2000); - PI3K (phosphoinositide-3-kinase, catalytic, alpha, beta, delta, gamma polypeptide): PIK3CA (3q26); PIK3CB (3q22); PIK3CD (1p36); PIK3CG (7q22); - PKD1 (14q11) (protein kinase D) (alias: PRKCM (mu)) (Wang et al., 2006); - PLCG: (phospholipase C, gamma 1, 2): PLCG1 (20q12), PLCG2 (16q24); - PRKC: protein kinase C family (alias PKC): PRKCA (alpha) (17q24) (alias: PKCA); PRKCB (beta) (16p12) (alias: PKCB); PRKCD (delta) (3p21), PRKCE (epsilon) (2p21); PRKCG (gamma) (19q13) (alias: PKCC, PKCG); PRKCH (eta) (14q23) (alias: PKCL, PRKCI); (iota) (3q26) (alias: PKCI); PRKCQ (theta) (10p15); PRKCZ (zeta) (1p36) (alias: PKC2) (Dempsey et al., 2000; Poole et al., 2004; Leitges, 2007; Malavez et al., 2008; Nelson et al., 2008); - PTEN (10q23) (phosphatase and tensin homolog deleted on chromosome ten) (Yin and Shen, 2008); - PTPN11 (12q24) (protein tyrosine phosphatase, non-receptor type, 11) (alias: SHP2) (Gadina et al., 1999; Tartaglia and Gelb, 2005); - RAF (v-raf murine sarcoma viral oncogene homolog): ARAF (Xp11); BRAF (7q34); RAF1 (3p25) (Domingo and Schwartz, 2004); - RAS (RAS viral oncogene homolog): HRAS (11p15) (Harvey); KRAS (12p12) (Kirsten); NRAS (1p13) (neuroblastoma) (Watzinger and Lion, 1999); - RHEB (7q36) (ras homolog enriched in brain) (Nobukini and Thomas, 2004); - RIN1 (11q13) (ras and rab interactor 1); - RPS6KA1 (1p36) (ribosomal protein S6 kinase, 90kDa, polypeptide 1) (Roux, 2008); - SHC1 (1q21) (SHC (src homology 2 domain containing) transforming protein 1); - SOS1 (2p21) (son of sevenless homolog 1); - SPRY (sprouty homolog (Drosophila)): SPRY1 (4q28); SPRY2 (13q31); SPRY4 (5q31) (Guy et al., 2003; Mason et al., 2006); - STAT (signal transducer and activator of transcription): STAT1 (2q32); STAT2 (12q13); STAT3 (17q21); STAT4 (2q32); STAT5a (17q11); STAT5b (17q11); STAT6 (12q13) (Schindler et al., 2007); - TSC1 (9q34) (tuberous Sclerosis 1) (Nobukini and Thomas, 2004); - TSC2 (16p13) (tuberous Sclerosis 2) (Nobukini and Thomas, 2004); - YWHAQ (2p25) (alias: 14-3-3); Note: IP3 (inositol 1,4,5-trisphosphate), DAG (diacylglycerol), PIP3 (Phosphatidylinositol (3,4,5)-trisphosphate (PtdIns(3,4,5)P3), are not genes/proteins.

Page 6: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 826

Recent studies showed that KAL1 (Xp22; Kallmann syndrome 1 sequence) acts as an FGFR1-specific modulator and coligand that physically interacts with the FGFR1-FGF-heparin sulfate proteoglycan complex and amplifies the resulting downstream signaling responses (Gonzalez-Martinez et al., 2004). Note: KAL1, like FGFR1, is involved in Kallmann syndrome (see below). Proteins which contain either a Src homology (SH2) domain, or a phosphotyrosine binding (PTB) domain can be phosphorylated/activated by the dimerized/activated receptor (herein FGFR1). FRS2, GRB2 and partners FRS2 (fibroblast growth factor receptor substrate 2) contains a PTB domain. Activation of the FGF-FGFR complex allows FRS2 to be phosphorylated, and then bind to GRB2 (growth factor receptor-bound protein 2) and PTPN11 (protein tyrosine phosphatase, non-receptor type, 11, alias SHP2). FGFRs (as well as other receptor tyrosine kinases), and also SHC1 (SHC (src homology 2 domain containing) transforming protein 1), PTPN11 and GAB1 (GRB2-associated binding protein 1) can bind GRB2 (Athauda and Bottaro, 2007). Phosphorylation of SHC1 and GAB1 induces binding to GRB2 and SOS1 (son of sevenless homolog 1) (Nelson et al., 2008) resulting in a multi-protein complex (Fig 3). GRB2 is constituvely associated with SOS. RAS/RAF/MAPK pathway GRB2-SOS stimulates the exchange of GTP to GDP on RAS (RAS viral oncogene homolog). RAS induces a phosphorylation cascade towards the nucleus, involving RAF (v-raf murine sarcoma viral oncogene homolog), MAP2K1 (mitogen-activated protein kinase kinase 1, alias MAPKK or MEK), MAPK (mitogen-activated protein kinase, alias ERK), ELK1 (ELK1, member of ETS oncogene family) and RPS6KA1 (ribosomal protein S6 kinase, 90kDa, polypeptide 1), towards cell cycle processes, differentiation, and homeostasis. PTPN11 positively regulates the RAS/RAF/MAPK pathway (Athauda and Bottaro, 2007). PRKC, a member of the PLC gamma pathway, phosphorylates a number of substrates, including MAP2K and YWHAQ (alias: 14-3-3) (Nelson et al., 2008). PKD1, another PLC gamma pathway member, upregulates the RAS/RAF/MAPK pathway by phosphorylating RIN1 (ras and rab interactor 1) and blocking its interaction with RAS. PI3K/AKT/mTOR pathway The complex FRS2-GRB2-GAB1 enables tyrosine phosphorylation of GAB1. GAB1, then, activates the PI3K/AKT/mTOR pathway, involving PI3K (phosphoinositide-3-kinase, catalytic, alpha, beta, delta, gamma polypeptide), PIP3 (Phosphatidylinositol (3,4,5)-trisphosphate (PtdIns(3,4,5)P3)), and AKT (v-akt murine thymoma viral oncogene homologs 1, 2, 3, alias: PKB). AKT has a great number of targets (Fig 4),

FRAP1 (alias mTOR) in particular. It has a role in glucose homeostasis, ribosomes and proteines syntheses, angiogenesis, stem cell maintenance, differentiation, survival, apoptosis, and cell cycle (Altomare and Testa, 2007). PLC gamma pathway PLCG induces the pathway involving DAG (diacylglycerol) and IP3 (inositol 1,4,5-trisphosphate) by lipid hydrolysis of PtdIns(4,5)P2, a rise of intracellular Ca++ concentration, PRKC (protein kinase C), and PKD1 (protein kinase D) through phosphorylation towards ion channels regulations, and various processes such as cell growth and differentiation, apoptosis and survival, cell motility and immune response. To be noted that PDPK1, regulated by PI3K from the PI3K/AKT/mTOR pathway, phosphorylates PRKC (Dempsey et al., 2000; Wang, 2006; Kheifets and Mochly-Rosen, 2007). JAK/STAT pathway PTPN11, once activated in the FRS2/PTPN11/GRB2/GAB1/SOS complex, provokes STAT (signal transducer and activator of transcription) dephosphorylation (review on JAK-STAT in Schindler et al., 2007). PRKC (member of the PLC gamma pathway) regulates STATs (Malavez et al., 2008). The JAK/STAT pathway regulates transcription, cell growth and differentiation, inflammation and immune response. IKK/NF-KB pathway PKD1 activates the NF-KB pathway. AKT, a member of the PI3K/AKT/mTOR pathway, activates CHUK (10q24, alias IKKA) (Altomare and Testa, 2007). RPS6KA1, a member of the RAS/RAF/MAPK pathway, inactivates NFKBIA (14q13) (Roux, 2008). PRKC regulates NFKB (Malavez et al., 2008). The IKK/NFKB pathway regulates survival processes. SPRY and CBL inhibition SPRY (sprouty homolog (Drosophila)) competes with PTPN11 and FRS2 for binding to the GRB2-SOS complex, and inhibits the RAS/RAF/MAPK pathway. On the other hand, SPRY prevents CBL-mediated ubiquitylation, endocytosis and degradation of FGFR (Guy et al., 2003; Mason et al., 2006; Dikic and Schmidt, 2007). Note: SPRY, like FGFR1, is involved in nonsyndromic cleft lip and palate (see below). Nuclear FGFR1 FGFs may: 1- be secreted (FGF 3-10, 16-23) out of the cell, bind on surface receptors (FGFRs) of other cells and activate signaling cascades as above described; 2- remain intra cellular (FGF 11-14); or 3- both (FGF1, FGF2). Intracytoplasmic FGFs can translocate to the nucleus and act as nuclear signaling molecules. All the same, FGFRs can be found inserted in the cytoplasmic membrane, but also in the cytosol, and in the nuclear compartment (Fig 5).

Page 7: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 827

Fig 5. Intracellular FGF and FGFR. Adapted from Bryant and Stow, 2005 and Stachowiak et al., 2007. dotted lines: migration; solid lines activations. ARF6 (14q21) (ADP-ribosylation factor 6); DNM2 (19p13) (dynamin 2); KPNB1 (17q21) (karyopherin beta 1, alias importin beta); PRKCD (3p21) (protein kinase C, alias PKCD); RAB5 (a, b, c) (3p14, 12q13, 17q21) (RAB5, member RAS). FGFR1 (and also FGFR2 and 3 but not FGFR4) contains an atypical transmembrane domain (TM), with a probable beta-sheet conformation, instead of the more membrane-stable alpha-helical conformation of other single TM tyrosine kinase receptors (Myers et al., 2003). ARF6 (14q21; ADP-ribosylation factor 6) and DNM2 (19p13; dynamin 2) facilitate surface FGFR1 internalization, RAB5 (RAB5; member RAS) facilitates the trafic into the endosome. RPS6KA1 would also favor FGFR1 release from the membrane to the cytosol and also prior to nuclear import. KPNB1 (17q21; importin beta) would facilitate FGFR1 nuclear import. VHL (3p25; von Hippel-Lindau tumor suppressor) is recruited to FGFR1-containing endosomal vesicles and exhibits a functional relationship with RAB5A and DNM2 in FGFR1 internalization. In cooperation with CREBBP, Nuclear FGFR1 (nFGFR1) up-regulates gene transcription of FGF2, CCND1 (11q13; cyclin D1), JUN (1p32; jun oncogene), NEFL (8p21; neurofilament, light polypeptide), TH (11p15; tyrosine hydroxylase) (Groth and Lardelli, 2002; Bryant and Stow, 2005; Hsu et al., 2006; Stachowiak et al., 2007).

Homology With other FGFR, namely: FGFR2, FGFR3, and FGFR4. FGFRL1 (4p16; fibroblast growth factor receptor-like 1) is a related protein that lacks the intracytoplasmic tyrosine kinase domain (Kim et al., 2001). FGFRs are highly conserved through evolution.

Implicated in Epithelial-to-mesenchymal transition Note FGFR1 is expressed during the mesoderm induction in the embryo. FGFR1 drives the epithelial-to-mesenchymal transition (EMT) of primitive epiblast cells into mesoderm cells (Ciruna et al., 1997; Ciruna and Rossant, 2001). In an EMT, epithelial cells lose their polarity, increase their motility, and begin to express mesenchymal markers, such as VIM (10p13; vimentin), becoming mesenchymal-like. This process has also been linked to cancer progression, in which epithelial cells lose differentiation markers, such as CDH1 (E-cadherin), and acquire increased migratory capacity, leading to stromal invasion and metastasis (Thiery, 2002; Acevedo et al., 2007).

Page 8: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 828

Angiogenesis Note FGF2 is an angiogenic factor, through interaction with FGFR1. Prostaglandin E2 stimulates FGF2/FGFR1 signaling (Finetti et al., 2008). FGF1 activation of FGFR1 signaling was found restricted to ligand activation of the FGFR1beta isoform. Transfection of endothelial cells with human cDNA encoding FGFR1beta induced cellular proliferation and formation of neovascular structures (Zhang et al., 2006).

Stem-cell myeloproliferative disorder Disease Stem-cell myeloproliferative disorder involving lymphoid (T- and B-cell) and myeloid lineages, called the 8p11 myeloproliferative syndrome (EMS) with variable presentations, e.g. a polycythaemia vera evolving towards an atypical myeloproliferative disorder, or a T-cell lymphoblastic lymphoma relapsing as an acute myeloblastic leukemia (AML) (Chaffanet et al., 1998; Reiter et al., 1998; Smedley et al., 1998; Xiao et al., 1998; Popovici et al., 1999; Guasch et al., 2000; Mugneret et al., 2000; Demiroglu et al., 2001; Fioretos et al., 2001; Sohal et al., 2001; Roy et al., 2002; Guasch et al., 2003; Grand et al., 2004; Vizmanos et al., 2004; Belloni et al., 2005; Walz et al., 2005; De Melo and Reid, 2006; Etienne et al., 2007; Hidalgo-Curtis et al., 2008; Mozziconacci et al., 2008; Park et al., 2008; Richebourg et al., 2008). The myeloproliferative syndrome transforms rapidly into a myelomonocytic leukemia. It has usually a poor outcome, since it is refractory to current chemotherapies, including imatinib. It involves FGFR1 on 8p11-12, and a variable partner (Fig 6 and 7).

Prognosis Very poor (median survival: 12 mths).

Cytogenetics The translocations are: t(6;8)(q27; p12) involving FGFR1OP (FGFR1 Oncogene Partner) t(7;8)(q34;p12) involving TRIM24 (transcriptional intermediary factor 1) t(8;9)(p12;q33) involving CEP110 (Centrosome protein 110) (review in Mozziconacci et al., 2008) t(8;11)(p12;p15) ins(12;8)(p11;p12p21) involving FGFR1OP2 (FGFR1 oncogene partner 2) t(8;12)(p12;q15) involving CPSF6 (cleavage and polyadenylation specific factor 6, 68kDa)

t(8;13)(p12;q12 ) involving ZMYM2 (zinc finger, MYM-type 2, alias ZNF198) t(8;17)(p12;q11) involving MYO18A (myosin XVIIIA) t(8;17)(p12;q25) t(8;19)(p12;q13.3) involving ERVK6 (endogenous retroviral sequence K, 6) t(8;22)(p12;q11) involving BCR (breakpoint cluster region) The translocation is the sole anomaly in half of the cases; additional anomalies are: duplication of one of the derivative chromosomes: 15% (strangely the der(partner) and the der(8) are equally found); 7/del(7q): 10%; +21: 10%; +8: 5%.

Hybrid/Mutated gene 5' partner - 3' FGFR1. All the hybrid genes involve fusion of the 5' partner to FGFR1 exon 9.

Abnormal protein Fusion transcripts encode large proteins containing the N-term of the translocation partner, and the tyrosine kinase domain of FGFR1 in the C-term.

Oncogenesis Constitutive activation of FGFR1, due to the presence of dimerization domains in the partner. However, some of the above mentioned partners do not seem to carry dimerization domains. Such hybrids partner-FGFR1, like CPSF6-FGFR1, may be functional in the cytosol or in the nucleus.

Anaplastic large cell lymphoma Oncogenesis FGFR1 copy number gain was observed in 3 of 15 anaplastic large cell lymphoma (ALCL) cases (Mao et al., 2003).

Breast cancer Note FGFs and FGFRs are expressed during ductal morphogenesis and their expression decreases throughout pregnancy and lactation. FGFR1 is mainly expressed during ductal morphogenesis. Additionally, expression of a dominant negative FGFR2 in the mammary gland during pregnancy has been shown to inhibit lobuloalveolar development. Inducible FGFR1 activation results in increased lateral budding of the mammary ductal epithelium, and induces alveolar hyperplasia and invasive lesions in the mouse (Welm et al., 2002). Inducible FGFR1 activation disrupt cell polarity, induce proliferation, and promotes cell survival. Inducible FGFR1 activation leads to dissociation of CDH1 from the cell membrane and expression of mesenchymal markers (Xian et al., 2005).

Page 9: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 829

Fig 6. FGFR1 fusion protein.

Oncogenesis Amplification of 8p11.2-p12 is reported to be found in 10 to 15% of all breast cancers (Ugolini et al., 1999). Amplification of 8p11.2-p12 seems to comprise at least 4 different and independant amplicons. One of those contains WHSC1L1 (Wolf-Hirschhorn syndrome candidate 1-like 1), DDHD2 (DDHD domain containing 2), BLP1 (BBP-like protein 1; alias TM2D2), PPAPDC1B (phosphatidic acid phosphatase type 2 domain containing 1B), LSM1 (LSM1 homolog, U6 small nuclear RNA associated (S. cerevisiae)), and FGFR1. This 8p11-12 amplification has a pejorative effect on survival in breast cancer (Gelsi-Boyer et al., 2005). The FGFR1 region was found amplified in 9.4% of 319 breast tumors (Letessier et al., 2006). In an analysis of 33 primary breast tumors, FGFR1 was not included in the minimal region of amplification. Instead, the genes FLJ14299 (zinc finger protein 703; alias ZNF703), C8orf2 (ER lipid raft associated 2; alias ERLIN2), BRF2 (BRF2, subunit of RNA polymerase III transcription initiation factor, BRF1-like) and RAB11FIP1, mapped within the 8p11-12 minimal amplicon, and they showed a strong correlation between amplification and overexpression (Garcia et al., 2005). In 6 of 13 cases of classic lobular carcinomas, FGFR1's region was amplified and FGFR1 was tought to be the amplicon driver, since all cases with FGFR1 copy number gains also showed protein overexpression. Furthermore, siRNA directed against FGFR1 reduced cell viability of a breast cancer cell line (Reis-Filho et al., 2006). In a cohort of 880 breast tumours with various histology, FGFR1 amplification was observed in 8.7% of the tumours and was significantly more prevalent in patients over 50 years of age, and in tumours that lacked ERBB2/HER2 expression. FGFR1 amplification was confirmed as an independent prognostic factor for overall survival in patients with oestrogen-receptor-positive tumours, where FGFR1 amplification was the strongest independent predictor of a poor outcome (Elbauomy Elsheikh et al., 2007). FGFR1beta was found preponderant in breast cancer, and FGFR1alpha in normal breast cells (Luqmani et al., 1995). Finally, a S125L mutation was found in one of 25 cases of breast cancer (Stephens et al., 2005).

Prostate cancer Note The prostate is composed of stromal cells and epithelial cells. Stromal cells secrete paracrine factors for the maintenance and growth of the epithelium, some of which are under the control of androgens. FGF2, 7 and

9 are the main FGFs that the stromal cells secrete. Prostate epithelial cells express multiple FGF receptors. FGFR1 and FGFR2 are expressed in the basal epithelial cells of the prostate but not the luminal cells. FGFR3 IIIb and FGFR4 are also expressed in normal epithelium. FGFR1 is present exclusively as the IIIc isoform, while FGFR2 is present exclusively as the IIIb (FGF7 specific) isoform in the epithelium. FGFR3 is also present in prostatic epithelium, predominantly in the IIIb isoform. FGFR4 is also expressed in prostatic epithelium in the luminal epithelial cells (review in Kwabi-Addo et al., 2004).

Oncogenesis FGFR1 is over-expressed in benign prostatic hyperplasia whereas FGFR2-IIIc and FGFR3 are not (Boget et al., 2001). Transcripts for FGFR1 are found in prostate cancer cells (Shain et al., 2004), while FGFR2 is down regulated (Naimi et al., 2002). Both FGFR1 and FGFR4 have been found overexpressed in a study of 138 malignant prostates (Sahadevan et al., 2007). Chronic activation of FGFR1 in mouse prostate epithelial cells induces progressive prostate intraepithelial neoplasia (Wang et al., 2004). The FGFR1-IIIb isoform was expressed in all cases of prostate cancer, while FGFR1-IIIc mRNA was not. FGFR1-IIIb transcripts were detected in four out of six cases of benign prostatic hyperplasia (Leung et al., 1997). Although FGFR1 was found overexpressed in prostate cancer, it was without any significant correlation to clinical parameters including tumour grade, stage, and outcome, according to some studies (Leung et al., 1997; Giri et al., 1999). Conversely, Devilard et al., 2006 found that FGFR1, TACC1 (8p11; transforming, acidic coiled-coil containing protein 1) and WT1 (11p13; Wilms tumor 1) were expressed at higher level in prostate carcinoma samples than in benign prostate tissue, at both mRNA and protein levels, especially so in pT3 and N1/M1 samples. Transfection and expression of FGFR1 in premalignant cells accelerated progression to the malignant phenotype; restauration of FGFRIIIb in cells expressing FGFR1 restored epithelial cell differenciation (Feng et al., 1997). FGF2 was found in cells surrounding the cancer cells (fibroblasts and endothelium), and FGFR1 and FGFR2 expression were found increased in poorly differenciated prostate cancers, which would enhence the response of cancer cells to FGF2 (Giri et al., 1999). Activation of inducible FGFR1 led to epithelial-to-mesenchymal transition (like with breast cells) and progression to adenocarcinoma in the mouse. Mice not only developed well-differentiated adenocarcinoma,

Page 10: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 830

but also exhibited several distinct malignant phenotypes: prostatic intraepithelial neoplasia, adenocarcinoma, transitional sarcomatoid-carcinoma, and frank sarcoma. Mice developed a greater incidence of a transitional sarcomatoid carcinoma with increasing age, consistent with the appearance of an epithelial-mesenchymal transition. Experimental up-regulation of FGFR1 provoked SOX9 increase. SOX9 (17q23; SRY (sex determining region Y)-box 9) is known to act with SNAI1 (20q13; snail homolog 1 (Drosophila)) and SNAI2 (8q12; snail homolog 2 (Drosophila)) to reduce CDH1, leading to a loss of cell-cell contact and increased migration (Acevedo et al., 2007). Enhanced mesenchymal expression of FGF10 leeds to the formation of cancers from murine prostate cells. Inhibition of FGFR1 signaling by dominant-negative FGFR1 reverts FGF10-induced adenocarcinoma (Memarzadeh et al., 2007). Amplification of FGFR1 and many other loci were found associated with the development of hormone resistance of the cancer cells (Edwards et al., 2003). SPRY1 (4q28; Sprouty1) and SPRY2 (13q31; Sprouty2) mRNAs, antagonists of FGF signaling (see above), are decreased in human prostate cancer (Kwabi-Addo, Wang et al., 2004; Fritzsche et al., 2006). Inducible FGFR1 provokes angiogenesis in the prostate of mice; ANGPT1 and ANGPT2 (angiopoietins 1 and 2, 8q23 and 8p23 respectively) were regulated by FGFR1 signaling and differentially expressed (Winter et al., 2007).

Bladder cancer Note FGFR1 is normally expressed in normal urothelium.

Oncogenesis Amplicons in 3p25 and 8p12 are frequent in bladder cancers. FGFR1 and RAF1 (3p25; v-raf-1 murine leukemia viral oncogene homolog 1) were found amplified in some bladder cancers and deleted in other (Simon et al., 2001).

Rhabdomyosarcoma (RMS) Note Muscle: FGFR1 blocks myogenesis by forcing the myoblasts to re-enter S-phase, instead of escaping in G1 to progress towards differenciation. Myoblast proliferation can be induced by a chimeric FGFR1 (Whitney et al., 2001). Myoblasts expressing a chimeric Fgfr1 were injected into infarcted hearts of mice. The treatment induced an increase in graft size, showing that selective proliferation can be induced (Stevens et al., 2007).

Oncogenesis FGFR1 over-expression was detected in 2 out of 2 embryonal rhabdomyosarcomas, 6 out of 6 alveolar rhabdomyosarcomas, and in the one pleomorphic adult variant tested, as well as in the RMS cell lines tested. A hypomethylation of a CpG island upstream to FGFR1

exon 1 was identified in the primary RMS tumors (Goldstein et al., 2007).

Glioblastoma Note FGFR1 is poorly expressed in normal glia. It is expressed as a form FGFR1alpha (containing the 3 Ig-like domains). Nuclear FGFR1 induces neuronal differentiation. FGFR1 is particularly expressed in the forebrain and in the midbrain-hindbrain boundary. FGFR1 may be the receptor for FGF8 mediated signaling in the midbrain-hindbrain boundary.

Oncogenesis In a study of 22 malignant astrocytomas, an alternatively spliced form FGFR1beta containing two Ig-like domains (and lacking Ig I, corresponding to exon 3, also called the alpha-exon) was preferentially expressed. Conversely, FGFR2 expression was abundant in normal white matter and in all low-grade astrocytomas but was not seen in malignant astrocytomas (Yamaguchi et al., 1994). FGFR1beta exhibits a tenfold higher affinity for FGF1 and FGF2 than FGFR1alpha. FGFR1 gene with alpha-exon inclusion targeted to glioblastoma cells had no discernable effect on cell growth in culture, but was associated with an increase in unstimulated CASP3 (4q35; caspase 3, apoptosis-related cysteine peptidase) and CASP7 (10q25; caspase 7, apoptosis-related cysteine peptidase) activity (Bruno et al., 2004). SFPQ (1p34; splicing factor proline/glutamine-rich, alias PTBP, polypyrimidine tract-binding protein) was identified as a regulator of FGFR1 splicing. SFPQ expression was found strongly increased in malignant glioblastoma multiforme tumors, but not in a low-grade astrocytoma case (Jin et al., 2000). Two mutations in FGFR1 were found (N546K and R576W), and no amplification, in a study of 19 glioblastomas (Rand et al., 2005).

Uveal melanoma Oncogenesis FGF1, FGF2 and FGFR1 are overexpressed in primary uveal melanomas. FGF2/FGFR1 mediate the RAS/RAF/MAPK over-activation which controls the proliferation of uveal melanoma cells (Lefevre et al., 2008).

Skin melanoma Oncogenesis Advanced stages of human melanoma have a high degree of vascularity. They express high levels of FGF2/FGFR1. Inhibiting FGF2 and FGFR1 suffices to inhibit tumor growth due to massive induction of melanoma cell apoptosis (Valesky et al., 2002).

Thyroid carcinoma Note FGFR2 is the only FGFR expressed in normal thyroid

Page 11: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 831

tissue, and this expression is diminuished in thyroid cancer. FGFR1 is nearly not expressed in normal thyroid, but significantly expressed in multinodular goiter.

Oncogenesis Enhanced expression of FGF1, FGF2, and FGFR1 accompany thyroid hyperplasia and are not exclusively associated with the neoplastic state. These factors may be involved in the pathogenesis of uncontrolled thyroid growth observed in these conditions (Thompson et al., 1998). FGF2 immunoreactivity was detected in a small percentage and FGFR1 in a high percentage of adenomatous goiters (Shingu et al., 1998). FGFR1 is expressed in hyperplastic goiters, benign adenomas, and carcinomas (St Bernard et al., 2005). FGFR1 and FGFR3 are expressed in well-differentiated tumours, while FGFR4 is expressed in aggressive tumours. FGF2 and FGFR1 were both detected in a high percentage of papillary carcinomas, 10 follicular carcinomas, 3 anaplastic carcinomas (Shingu et al., 1998). FGFR1 is expressed in thyroid carcinoma cell lines through propagation of the MAPK activation and promotion of tumor progression. In contrast, FGFR2 is down-regulated. FGFR2-IIIb and FGFR1 compete with each other for FRS2 activation (Kondo et al., 2007).

Oral squamous cell carcinoma (OSCC) Oncogenesis An amplicon on 8p12 was found in a number of tumours. High FGFR1 expression was found in 12% of 178 tumours; there was a significant higher prevalence of T1/T2 versus T3/T4, stages I/II vs III/IV, but not between N0 and N1-3, nor in overall survival. Prevalence of low stages and superficial tumours may suggest a role of FGFR1 in the early steps of transformation of the oral epithelium (Freier et al., 2007).

Salivary gland tumors Note The expression of FGFR1 is highest early during gland development, inducing branching morphogenesis.

Oncogenesis In 10 patients with pleomorphic adenoma of the salivary gland, the expression of FGF1, FGF2, and FGFR1 appeared to be related to the proliferative activity of tumor cells in the tubular and solid areas, whereas loss of FGFR1 expression may be associated with the differentiation of tumor cells into myxoid and chondroid tissue types (Myoken et al., 1997). A subgroup of pleomorphic salivary gland adenomas showed a ring chromosome 8: r(8)(p12q12) with amplification of a hybrid gene 5'-FGFR1 3'-PLAG1, and also multiple copies of an intact PLAG1 (8q12; pleiomorphic adenoma gene 1) (Persson et al., 2008).

Esophageal squamous cell carcinoma Oncogenesis An amplicon on 8p12, including FGFR1, was found in 2 of 32 cases of esophagus cancer (Ishizuk et al., 2002). Co-expression of FGF1 and FGFR1 is predictive of a poor prognosis in patients with esophageal squamous cell carcinoma (Sugiura et al., 2007).

Hepatocellular carcinoma Note FGFR1 is exclusively expressed in nonparenchymal cells and FGFR4 exclusively expressed in the hepatocytes.

Oncogenesis Ectopic expression of FGFR1 in hepatocytes increased the number of proliferating hepatocytes but failed to induce preneoplasia or neoplasia. Ectopic FGFR1 was incapable of initiation of hepatomas; however, it was a strong promoter of carcinogenesis both as a proliferative enhancer and as a promoter of neoangiogenesis mediated by VEGF (6p12; vascular endothelial growth factor A) (Huang et al., 2006).

Colorectal carcinoma Oncogenesis FGFR1 is overexpressed, and FGFR3 is frequently downregulated in colorectal carcinoma. FGFR3 may promote cell differentiation (Jang 2005).

Lung cancer Oncogenesis Patients with high FGFR1 expression had significantly shorter survival than patients with weak or moderate expression. However, FGFR1 expression is not an independent prognostic factor in the presence of the stage (Volm et al., 1997). A single somatic mutation in FGFR1 (P252T) was observed in a bronchoalveolar cancer from a study of 26 primary lung neoplasms (Davies et al., 2005).

Pancreatic carcinoma Note At early stages of pancreas development, FGFR1-IIIb is expressed by pancreatic epithelial cells, while at later stages of development, FGFR1-IIIb expression decreases, concomitant with the expected decrease in the number of progenitor cells (Cras-Meneur and Scharfmann, 2002). FGF1alpha is expressed in normal pancreatic tissue.

Oncogenesis A significant correlation was found between FGFR1 expression level and advanced stages in pancreatic carcinoma. Low FGFR1 expression was correlated with longer survival (Ohta et al., 1995).

Page 12: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 832

Patients with tumors that showed high expression of VEGF and FGF2 had significantly shorter survival (Kuwahara et al., 2003). Pancreatic adenocarcinoma overexpresses fibroblast growth factor ligands (FGF-1 and FGF-2) and FGFR1beta in about 90% of the time. Overexpression of FGFR1alpha inhibits pancreatic adenocarcinoma cells (Vickers et al., 2002). FGFR1-IIIc enhances and FGFR1-IIIb inhibits pancreatic cancer cell growth. Expression of FGFR1-IIIb inhibited the transformed phenotype of human pancreatic cancer cells. This was associated with a reduced p44/42 MAPK phosphorylation and an enhanced activity of JNK (10q11; JUN N-terminal kinase, alias MAPK8) and p38. The antiproliferative effects of FGFR1-IIIb were confirmed in a xenograft model. IIIc enhanced and IIIb inhibited basal cell proliferation in pancreatic cancer and non-cancer cells. Expression of FGFR1-IIIc in nonmalignant pancreatic ductal cells resulted in cellular transformation and in vivo tumor formation, whereas inhibition of FGFR1-IIIc resulted in a reversion of the malignant phenotype in pancreatic cancer cells. FGFR1-IIIc promotes pancreatic ductal adenocarcinomas cell growth through phosphorylation of FRS2, PLCG, and activation of the RAS/RAF/MAPK pathway (Kornmann et al., 2002; Liu et al., 2007). FGFR1-IIIb and FGFR1-IIIc are coexpressed in pancreatic cancer and these isoforms are differentially regulated by growth factors FGFs, IGF1R (15q26; insulin-like growth factor 1 receptor) and EGF (4q25; epidermal growth factor), and by CCND1 (11q13; cyclin D1) (Chen et al., 2008).

Pfeiffer syndrome Note Signaling in endochondral bone formation in the embryo: Null mice embryos lacking Fgfr1 die shortly after gastrulation. Prehypertrophic chondrocytes, hyperthrophic chondrocytes and osteoblasts express FGFR1. In mature bone, Fgfr1 and Fgfr2 continue to be expressed in osteoblasts (Ornitz, 2005; White et al., 2005). FGFR1 may function as a negative regulator of long bone development rather than increasing skull bone growth. FGFR1 expression is positively regulated by thyroid hormone T3 in osteoblasts. FGFR1 is important for early limb bud development and distal skeletal patterning. It is expressed in limb bud mesenchyme that gives rise to mesenchymal condensations and eventually to the chondrogenic and osteogenic lineage. Inactivation of FGFR1 in osteo-chondro-progenitor cells delayed osteoblast differentiation; conversely, inactivation of FGFR1 in differentiated osteoblasts accelerated differentiation. It appears that FGFR1 expression in osteoblasts is necessary to maintain the balance between bone formation and remodeling (Jacob et al., 2006). Pfeiffer syndrome is due to an activating mutation of FGFR1.

Disease Pfeiffer syndrome is an autosomal dominant

craniosynostosis syndrome. It is defined by the association of a coronal synostosis, with or without sagittal synostosis, turricephaly, hydrocephalus, hypertelorism, low nasal bridge, radiohumeral synostosis, broad thumbs and first toes, partial syndactyly, and usually no mental deficiency. Type I to III have been defined according to the severity of the symptoms (Webster and Donoghue, 1997; for review on craniosynostosis, see Cunningham et al., 2007). Pfeiffer syndrome is due to mutations in FGFR1 or in FGFR2. A unique mutation in FGFR1 is known to provoke Pfeiffer syndrome: P252R, where a proline is replaced by an arginine in the IgII-III linker region (see figure I) (Muenke et al., 1994; Schell et al., 1995; Rossi et al., 2003; Ibrahimi et al., 2004). Pfeiffer syndrome with a mutation in FGFR2 exhibits the heavier form, those with a mutation in FGFR1 exhibits the milder form (type I) of the disease. It is an activating mutation, which enhance the affinity of FGF9, causing inappropriate dimerization of FGFR1 and signal transduction (Ibrahimi et al., 2004). Note: Jackson-Weiss syndrome is caused by mutations in codons 342, 344, or 359, or deletion at codon 319 in FGFR2 gene. No case of Jackson-Weiss syndrome is due to a mutation in FGFR1, altough this is sometimes found written in databases.

Osteoglophonic/osteoglyphic dysplasia Note Activating mutations of FGFR1.

Disease Osteoglophonic dysplasia (or, better called, osteoglyphic dysplasia (Greenberg and Lewis, 1990)) is an autosomal dominant condition, but most cases are de novo. It is a skeletal dysplasia syndrome that shares characteristics with craniosynostosis syndromes and rhizomelic dwarfing syndromes. Features also include prominent supraorbital ridge, depressed nasal root, multiple unerupted teeth, diaphyseal fibrous lesions, and speech delay (Sklower Brooks et al., 1996). Mutations within FGFR1 are within the IgIII (also called D3) domain, the linker region and the initial transmembrane domain: N330I, Y374C, C381R (White et al., 2005; Farrow et al., 2006). They appear to be activating mutations, causing inappropriate dimerization of FGFR1.

Nonsyndromic cleft lip and palate (NS CLP) Note Loss-of-function mutations of FGFR1.

Disease Nonsyndromic cleft lip and palate (NS CLP) is a complex birth defect resulting from a combination of genetic and environmental factors, and the genetics of the disease are still poorly known. Mutations were found so far in MSX1 (4p16; msh homeobox 1), FOXE1 (9q22; forkhead box E1 (thyroid transcription

Page 13: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 833

factor 2)), GLI2 (2q14; GLI-Kruppel family member GLI2), MSX2 (5q35; msh homeobox 2), SKI (1p36; v-ski sarcoma viral oncogene homolog (avian)), SPRY2 and, recently in FGF family members, namely: FGFR1, FGFR2, FGFR3, and FGF8. The mutations found in FGFR1 were I300T, M369I, E467K, and R609X. These loss-of-function mutations map to the IgIII-TM linker, the TM-TK linker, and to the TK domain. Penetrance appears to be incomplete (Kress et al., 2000; Riley et al., 2007).

Kallmann Syndrome (isolated idiopathic hypogonadotropic hypogonadism, isolated anosmia) Note Loss-of-function mutations of FGFR1.

Disease Kallmann syndrome (KS) is characterized by the association of anosmia and idiopathic or isolated hypogonadotropic hypogonadism (IHH) (pubertal development failure due to an impaired secretion of LH and FSH, in the absence of any hypothalamic-pituitary cause). Neurologic defects, including hearing loss, cleft palate, dental agenesis and renal aplasia may occur. Hypogonadism is due to gonadotropin releasing hormone (GnRH) deficiency and anosmia is related to hypoplasia of the olfactory bulbs; GnRH neurons originate in the nasal placode like olfactory neurons; they both migrate. GnRH neuroblasts formation/migration deficiency during embryogenesis may be responsible for Kallman syndrome (Gonzalez-Martinez et al., 2004). Incomplete penetrance of hypogonadism and/or anosmia is frequently described in Kallmann syndrome; cases may be inherited or sporadic. Kallmann syndrome is found in 10/100 000 males and 2/100 000 females. There is X-linked cases, autosomal dominant cases, and autosomal recessive transmissions. Kallmann syndrome is genetically heterogeneous, and may be due to mutations in KAL1 (Xp22.3; also called anosmin-1,causing the X-linked form of KS), FGFR1 (also called KAL2 for Kallmann 2, often associated with cleft palate and dental agenesis), PROK2 (3p13; prokineticin 2), and PROKR2 (20p12; prokineticin receptor 2). However, only 25% of Kallmann syndrome cases are due to mutations in these four genes. To be noted is that KAL1 has recently be found to acts as an FGFR1-specific modulator and coligand that physically interacts with the FGFR1-FGF-heparin sulfate proteoglycan complex (specifically through FGFR1 IIIc), and amplifies the FGF/FGFR1 downstream signaling responses in olfactory neuroblasts (Gonzalez-Martinez et al., 2004). Mutations or interstitial deletions in FGFR1 are found in 10% of individuals with Kallmann syndrome. Mutations within FGFR1 are variable: G48S, N77K, R78C, G97D, Y99C, 303-304insCC, C101F, V102I, S107X, D129A, A167S, C178S, D224H, G237S,

G237D, L245P, R250W (2 cases), R254Q, G270D, V273M (2 cases), E274G, C277Y, P283R, 936G->A, E324X, S332C, Y339C, A343V, S346C, c.1081G>C (splice site), R365fsX41, P366 liter, S439fs, A520T, I538V, V607M, Y613fsX42, K618fsX654, H621R, R622X, R622G, R622Q, 1970-1971delCA, c.1977+1G>A (splice site), R661X, W666R, Q680X, G687R, IV15+1G->A, G703S, G703R, M719R, P722S (2 cases), S685F, I693F, P722H and N724K double mutation, Y730X, P745S, P772S, V795I, R822C; However, the N77K and R822C changes were also found in healthy control individuals. The above noted mutations are dispersed in variable sites of FGFR1 (Ig domains, Ig-Ig linkers, IgIII-TM linker, TK domains, C-term); they appear to be dominant loss-of-function mutations. FGFR1 mutations also account for some of the mixed pedigrees, extending to hypogonadotropic hypogonadism and normal olfaction or, conversely, to isolated anosmia, or even asymptomatic carriers at times (Dode et al., 2003; Sato et al., 2004; Sato et al., 2005; Albuisson et al., 2005; Karges and de Roux, 2005; Pitteloud, Acierno et al., 2006; Pitteloud, Meysing, et al., 2006; Zenaty et al., 2006; Trarbach et al., 2008; Dode et al., 2007).

Depression, bipolar disorder, schizophrenia Disease Raised FGFR1 mRNA was found in the hippocampus and the gyrus in case of major depression and in cases with shizophrenia as well (Gaughan et al., 2006), and in the cortex of patients with major depression - but not in shizophrenia cases - (Tochigi et al., 2008). Conversely, decreased FGFR1 mRNA was found in the hippocampus in rats with social defeat experience (Turner, Calvo et al., 2008). Reduced density of dopamine neurons was found in transgenic mice expressing a dominant negative FGFR1 mutant; the mice presented similar structural and biochemical changes in dopamine neurons as what is found in shizophrenia patients (Klejbor et al., 2006). Caution must be taken when comparing the results, since, methodology, histology and anatomic regions of the cells under study, and diagnoses of the population studied may not be comparable from one study to another. Rats with an increased locomotor response to a novel environment exhibited increased FGFR1 mRNA in the hippocampus compared to other rats. Cocaine injections decreased FGFR1 mRNA in the hippocampus and increased FGFR1 mRNA in the prefrontal cortex. In rats with an increased locomotor response, cocaine decreased gene expression in the hippocampus and did not affect it in the prefrontal cortex, while, in other rats, cocaine did not affect gene expression in the hippocampus and increased gene expression in the prefrontal cortex (Turner, Flagel et al., 2008).

Page 14: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 834

Breakpoints

References Lee PL, Johnson DE, Cousens LS, Fried VA, Williams LT. Purification and complementary DNA cloning of a receptor for basic fibroblast growth factor. Science. 1989 Jul 7;245(4913):57-60

Greenberg F, Lewis RA. Osteoglophonic dysplasia. J Med Genet. 1990 Mar;27(3):213

Itoh N, Terachi T, Ohta M, Seo MK. The complete amino acid sequence of the shorter form of human basic fibroblast growth factor receptor deduced from its cDNA. Biochem Biophys Res Commun. 1990 Jun 15;169(2):680-5

Wennström S, Sandström C, Claesson-Welsh L. cDNA cloning and expression of a human FGF receptor which binds acidic and basic FGF. Growth Factors. 1991;4(3):197-208

Johnson DE, Williams LT. Structural and functional diversity in the FGF receptor multigene family. Adv Cancer Res. 1993;60:1-41

Muenke M, Schell U, Hehr A, Robin NH, Losken HW, Schinzel A, Pulleyn LJ, Rutland P, Reardon W, Malcolm S. A common mutation in the fibroblast growth factor receptor 1 gene in Pfeiffer syndrome. Nat Genet. 1994 Nov;8(3):269-74

Yamaguchi F, Saya H, Bruner JM, Morrison RS. Differential expression of two fibroblast growth factor-receptor genes is associated with malignant progression in human astrocytomas. Proc Natl Acad Sci U S A. 1994 Jan 18;91(2):484-8

Luqmani YA, Mortimer C, Yiangou C, Johnston CL, Bansal GS, Sinnett D, Law M, Coombes RC. Expression of 2 variant forms

of fibroblast growth factor receptor 1 in human breast. Int J Cancer. 1995 Aug 22;64(4):274-9

Ohta T, Yamamoto M, Numata M, Iseki S, Tsukioka Y, Miyashita T, Kayahara M, Nagakawa T, Miyazaki I, Nishikawa K, Yoshitake Y. Expression of basic fibroblast growth factor and its receptor in human pancreatic carcinomas. Br J Cancer. 1995 Oct;72(4):824-31

Schell U, Hehr A, Feldman GJ, Robin NH, Zackai EH, de Die-Smulders C, Viskochil DH, Stewart JM, Wolff G, Ohashi H. Mutations in FGFR1 and FGFR2 cause familial and sporadic Pfeiffer syndrome. Hum Mol Genet. 1995 Mar;4(3):323-8

Seger R, Krebs EG. The MAPK signaling cascade. FASEB J. 1995 Jun;9(9):726-35

Doherty P, Walsh FS. CAM-FGF Receptor Interactions: A Model for Axonal Growth Mol Cell Neurosci. 1996 Aug;8(2/3):99-111

Sklower Brooks S, Kassner G, Qazi Q, Keogh MJ, Gorlin RJ. Osteoglophonic dysplasia: review and further delineation of the syndrome. Am J Med Genet. 1996 Dec 11;66(2):154-62

Ciruna BG, Schwartz L, Harpal K, Yamaguchi TP, Rossant J. Chimeric analysis of fibroblast growth factor receptor-1 (Fgfr1) function: a role for FGFR1 in morphogenetic movement through the primitive streak. Development. 1997 Jul;124(14):2829-41

Feng S, Wang F, Matsubara A, Kan M, McKeehan WL. Fibroblast growth factor receptor 2 limits and receptor 1 accelerates tumorigenicity of prostate epithelial cells. Cancer Res. 1997 Dec 1;57(23):5369-78

Page 15: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 835

Leung HY, Mehta P, Gray LB, Collins AT, Robson CN, Neal DE. Keratinocyte growth factor expression in hormone insensitive prostate cancer. Oncogene. 1997 Aug 28;15(9):1115-20

Myoken Y, Myoken Y, Okamoto T, Sato JD, Kan M, McKeehan WL, Fujihara M, Takada K. Immunohistochemical localization of fibroblast growth factor-1 (FGF-1), FGF-2 and fibroblast growth factor receptor-1 (FGFR-1) in pleomorphic adenoma of the salivary glands. J Oral Pathol Med. 1997 Jan;26(1):17-22

Saffell JL, Williams EJ, Mason IJ, Walsh FS, Doherty P. Expression of a dominant negative FGF receptor inhibits axonal growth and FGF receptor phosphorylation stimulated by CAMs. Neuron. 1997 Feb;18(2):231-42

Volm M, Koomägi R, Mattern J, Stammler G. Prognostic value of basic fibroblast growth factor and its receptor (FGFR-1) in patients with non-small cell lung carcinomas. Eur J Cancer. 1997 Apr;33(4):691-3

Webster MK, Donoghue DJ. FGFR activation in skeletal disorders: too much of a good thing. Trends Genet. 1997 May;13(5):178-82

Chaffanet M, Popovici C, Leroux D, Jacrot M, Adélaïde J, Dastugue N, Grégoire MJ, Hagemeijer A, Lafage-Pochitaloff M, Birnbaum D, Pébusque MJ. t(6;8), t(8;9) and t(8;13) translocations associated with stem cell myeloproliferative disorders have close or identical breakpoints in chromosome region 8p11-12. Oncogene. 1998 Feb 19;16(7):945-9

Naski MC, Ornitz DM. FGF signaling in skeletal development. Front Biosci. 1998 Aug 1;3:d781-94

Reiter A, Sohal J, Kulkarni S, Chase A, Macdonald DH, Aguiar RC, Gonçalves C, Hernandez JM, Jennings BA, Goldman JM, Cross NC. Consistent fusion of ZNF198 to the fibroblast growth factor receptor-1 in the t(8;13)(p11;q12) myeloproliferative syndrome. Blood. 1998 Sep 1;92(5):1735-42

Shingu K, Fujimori M, Ito K, Hama Y, Kasuga Y, Kobayashi S, Itoh N, Amano J. Expression of fibroblast growth factor-2 and fibroblast growth factor receptor-1 in thyroid diseases: difference between neoplasms and hyperplastic lesions. Endocr J. 1998 Feb;45(1):35-43

Smedley D, Hamoudi R, Clark J, Warren W, Abdul-Rauf M, Somers G, Venter D, Fagan K, Cooper C, Shipley J. The t(8;13)(p11;q11-12) rearrangement associated with an atypical myeloproliferative disorder fuses the fibroblast growth factor receptor 1 gene to a novel gene RAMP. Hum Mol Genet. 1998 Apr;7(4):637-42

Thompson SD, Franklyn JA, Watkinson JC, Verhaeg JM, Sheppard MC, Eggo MC. Fibroblast growth factors 1 and 2 and fibroblast growth factor receptor 1 are elevated in thyroid hyperplasia. J Clin Endocrinol Metab. 1998 Apr;83(4):1336-41

Xiao S, Nalabolu SR, Aster JC, Ma J, Abruzzo L, Jaffe ES, Stone R, Weissman SM, Hudson TJ, Fletcher JA. FGFR1 is fused with a novel zinc-finger gene, ZNF198, in the t(8;13) leukaemia/lymphoma syndrome. Nat Genet. 1998 Jan;18(1):84-7

Gadina M, Sudarshan C, O'Shea JJ. IL-2, but not IL-4 and other cytokines, induces phosphorylation of a 98-kDa protein associated with SHP-2, phosphatidylinositol 3'-kinase, and Grb2. J Immunol. 1999 Feb 15;162(4):2081-6

Giri D, Ropiquet F, Ittmann M. Alterations in expression of basic fibroblast growth factor (FGF) 2 and its receptor FGFR-1 in human prostate cancer. Clin Cancer Res. 1999 May;5(5):1063-71

Popovici C, Zhang B, Grégoire MJ, Jonveaux P, Lafage-Pochitaloff M, Birnbaum D, Pébusque MJ. The t(6;8)(q27;p11) translocation in a stem cell myeloproliferative disorder fuses a novel gene, FOP, to fibroblast growth factor receptor 1. Blood. 1999 Feb 15;93(4):1381-9

Ugolini F, Adélaïde J, Charafe-Jauffret E, Nguyen C, Jacquemier J, Jordan B, Birnbaum D, Pébusque MJ. Differential expression assay of chromosome arm 8p genes identifies Frizzled-related (FRP1/FRZB) and Fibroblast Growth Factor Receptor 1 (FGFR1) as candidate breast cancer genes. Oncogene. 1999 Mar 11;18(10):1903-10

Watzinger F, Lion T.. RAS family. Atlas Genet Cytogenet Oncol Haematol. March 1999. URL : http://AtlasGeneticsOncology.org/Deep/Ras.html

Dempsey EC, Newton AC, Mochly-Rosen D, Fields AP, Reyland ME, Insel PA, Messing RO. Protein kinase C isozymes and the regulation of diverse cell responses. Am J Physiol Lung Cell Mol Physiol. 2000 Sep;279(3):L429-38

Goodman RH, Smolik S. CBP/p300 in cell growth, transformation, and development. Genes Dev. 2000 Jul 1;14(13):1553-77

Guasch G, Mack GJ, Popovici C, Dastugue N, Birnbaum D, Rattner JB, Pébusque MJ. FGFR1 is fused to the centrosome-associated protein CEP110 in the 8p12 stem cell myeloproliferative disorder with t(8;9)(p12;q33). Blood. 2000 Mar 1;95(5):1788-96

Jin W, McCutcheon IE, Fuller GN, Huang ES, Cote GJ. Fibroblast growth factor receptor-1 alpha-exon exclusion and polypyrimidine tract-binding protein in glioblastoma multiforme tumors. Cancer Res. 2000 Mar 1;60(5):1221-4

Kress W, Petersen B, Collmann H, Grimm T. An unusual FGFR1 mutation (fibroblast growth factor receptor 1 mutation) in a girl with non-syndromic trigonocephaly. Cytogenet Cell Genet. 2000;91(1-4):138-40

Mugneret F, Chaffanet M, Maynadié M, Guasch G, Favre B, Casasnovas O, Birnbaum D, Pébusque MJ. The 8p12 myeloproliferative disorder. t(8;19)(p12;q13.3): a novel translocation involving the FGFR1 gene. Br J Haematol. 2000 Nov;111(2):647-9

Boget S, Cereser C, Parvaz P, Leriche A, Revol A. Fibroblast growth factor receptor 1 (FGFR1) is over-expressed in benign prostatic hyperplasia whereas FGFR2-IIIc and FGFR3 are not. Eur J Endocrinol. 2001 Sep;145(3):303-10

Chang L, Karin M. Mammalian MAP kinase signalling cascades. Nature. 2001 Mar 1;410(6824):37-40

Ciruna B, Rossant J. FGF signaling regulates mesoderm cell fate specification and morphogenetic movement at the primitive streak. Dev Cell. 2001 Jul;1(1):37-49

Demiroglu A, Steer EJ, Heath C, Taylor K, Bentley M, Allen SL, Koduru P, Brody JP, Hawson G, Rodwell R, Doody ML, Carnicero F, Reiter A, Goldman JM, Melo JV, Cross NC. The t(8;22) in chronic myeloid leukemia fuses BCR to FGFR1: transforming activity and specific inhibition of FGFR1 fusion proteins. Blood. 2001 Dec 15;98(13):3778-83

Fioretos T, Panagopoulos I, Lassen C, Swedin A, Billström R, Isaksson M, Strömbeck B, Olofsson T, Mitelman F, Johansson B. Fusion of the BCR and the fibroblast growth factor receptor-1 (FGFR1) genes as a result of t(8;22)(p11;q11) in a myeloproliferative disorder: the first fusion gene involving BCR but not ABL. Genes Chromosomes Cancer. 2001 Dec;32(4):302-10

Page 16: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 836

Javelaud D, Besancon F.. CDKN1A (cyclin-dependent kinase inhibitor 1A). Atlas Genet Cytogenet Oncol Haematol. April 2001. URL : http://AtlasGeneticsOncology.org/Genes/CDKN1AID139.html

Kim I, Moon S, Yu K, Kim U, Koh GY. A novel fibroblast growth factor receptor-5 preferentially expressed in the pancreas(1). Biochim Biophys Acta. 2001 Mar 19;1518(1-2):152-6

Ornitz DM, Itoh N. Fibroblast growth factors. Genome Biol. 2001;2(3):REVIEWS3005

Pearson G, Robinson F, Beers Gibson T, Xu BE, Karandikar M, Berman K, Cobb MH. Mitogen-activated protein (MAP) kinase pathways: regulation and physiological functions. Endocr Rev. 2001 Apr;22(2):153-83

Simon R, Richter J, Wagner U, Fijan A, Bruderer J, Schmid U, Ackermann D, Maurer R, Alund G, Knönagel H, Rist M, Wilber K, Anabitarte M, Hering F, Hardmeier T, Schönenberger A, Flury R, Jäger P, Fehr JL, Schraml P, Moch H, Mihatsch MJ, Gasser T, Sauter G. High-throughput tissue microarray analysis of 3p25 (RAF1) and 8p12 (FGFR1) copy number alterations in urinary bladder cancer. Cancer Res. 2001 Jun 1;61(11):4514-9

Sohal J, Chase A, Mould S, Corcoran M, Oscier D, Iqbal S, Parker S, Welborn J, Harris RI, Martinelli G, Montefusco V, Sinclair P, Wilkins BS, van den Berg H, Vanstraelen D, Goldman JM, Cross NC. Identification of four new translocations involving FGFR1 in myeloid disorders. Genes Chromosomes Cancer. 2001 Oct;32(2):155-63

Whitney ML, Otto KG, Blau CA, Reinecke H, Murry CE. Control of myoblast proliferation with a synthetic ligand. J Biol Chem. 2001 Nov 2;276(44):41191-6

Cras-Méneur C, Scharfmann R. FGFR1-IIIb is a putative marker of pancreatic progenitor cells. Mech Dev. 2002 Aug;116(1-2):205-8

Groth C, Lardelli M. The structure and function of vertebrate fibroblast growth factor receptor 1. Int J Dev Biol. 2002;46(4):393-400

Ishizuka T, Tanabe C, Sakamoto H, Aoyagi K, Maekawa M, Matsukura N, Tokunaga A, Tajiri T, Yoshida T, Terada M, Sasaki H. Gene amplification profiling of esophageal squamous cell carcinomas by DNA array CGH. Biochem Biophys Res Commun. 2002 Aug 9;296(1):152-5

Kornmann M, Ishiwata T, Matsuda K, Lopez ME, Fukahi K, Asano G, Beger HG, Korc M. IIIc isoform of fibroblast growth factor receptor 1 is overexpressed in human pancreatic cancer and enhances tumorigenicity of hamster ductal cells. Gastroenterology. 2002 Jul;123(1):301-13

Naimi B, Latil A, Fournier G, Mangin P, Cussenot O, Berthon P. Down-regulation of (IIIb) and (IIIc) isoforms of fibroblast growth factor receptor 2 (FGFR2) is associated with malignant progression in human prostate. Prostate. 2002 Aug 1;52(3):245-52

Roy S, Szer J, Campbell LJ, Juneja S. Sequential transformation of t(8;13)-related disease: a case report. Acta Haematol. 2002;107(2):95-7

Thiery JP. Epithelial-mesenchymal transitions in tumour progression. Nat Rev Cancer. 2002 Jun;2(6):442-54

Valesky M, Spang AJ, Fisher GW, Farkas DL, Becker D. Noninvasive dynamic fluorescence imaging of human melanomas reveals that targeted inhibition of bFGF or FGFR-1 in melanoma cells blocks tumor growth by apoptosis. Mol Med. 2002 Feb;8(2):103-12

Vickers SM, Huang ZQ, MacMillan-Crow L, Greendorfer JS, Thompson JA. Ligand activation of alternatively spliced

fibroblast growth factor receptor-1 modulates pancreatic adenocarcinoma cell malignancy. J Gastrointest Surg. 2002 Jul-Aug;6(4):546-53

Welm BE, Freeman KW, Chen M, Contreras A, Spencer DM, Rosen JM. Inducible dimerization of FGFR1: development of a mouse model to analyze progressive transformation of the mammary gland. J Cell Biol. 2002 May 13;157(4):703-14

Dodé C, Levilliers J, Dupont JM, De Paepe A, Le Dû N, Soussi-Yanicostas N, Coimbra RS, Delmaghani S, Compain-Nouaille S, Baverel F, Pêcheux C, Le Tessier D, Cruaud C, Delpech M, Speleman F, Vermeulen S, Amalfitano A, Bachelot Y, Bouchard P, Cabrol S, Carel JC, Delemarre-van de Waal H, Goulet-Salmon B, Kottler ML, Richard O, Sanchez-Franco F, Saura R, Young J, Petit C, Hardelin JP. Loss-of-function mutations in FGFR1 cause autosomal dominant Kallmann syndrome. Nat Genet. 2003 Apr;33(4):463-5

Edwards J, Krishna NS, Witton CJ, Bartlett JM. Gene amplifications associated with the development of hormone-resistant prostate cancer. Clin Cancer Res. 2003 Nov 1;9(14):5271-81

Guasch G, Popovici C, Mugneret F, Chaffanet M, Pontarotti P, Birnbaum D, Pébusque MJ. Endogenous retroviral sequence is fused to FGFR1 kinase in the 8p12 stem-cell myeloproliferative disorder with t(8;19)(p12;q13.3). Blood. 2003 Jan 1;101(1):286-8

Guy GR, Wong ES, Yusoff P, Chandramouli S, Lo TL, Lim J, Fong CW. Sprouty: how does the branch manager work? J Cell Sci. 2003 Aug 1;116(Pt 15):3061-8

Kuwahara K, Sasaki T, Kuwada Y, Murakami M, Yamasaki S, Chayama K. Expressions of angiogenic factors in pancreatic ductal carcinoma: a correlative study with clinicopathologic parameters and patient survival. Pancreas. 2003 May;26(4):344-9

Mao X, Orchard G, Lillington DM, Russell-Jones R, Young BD, Whittaker S. Genetic alterations in primary cutaneous CD30+ anaplastic large cell lymphoma. Genes Chromosomes Cancer. 2003 Jun;37(2):176-85

Myers JM, Martins GG, Ostrowski J, Stachowiak MK. Nuclear trafficking of FGFR1: a role for the transmembrane domain. J Cell Biochem. 2003 Apr 15;88(6):1273-91

Rossi M, Jones RL, Norbury G, Bloch-Zupan A, Winter RM. The appearance of the feet in Pfeiffer syndrome caused by FGFR1 P252R mutation. Clin Dysmorphol. 2003 Oct;12(4):269-74

Bruno IG, Jin W, Cote GJ. Correction of aberrant FGFR1 alternative RNA splicing through targeting of intronic regulatory elements. Hum Mol Genet. 2004 Oct 15;13(20):2409-20

Domingo E, Schwartz S Jr.. BRAF. Atlas Genet Cytogenet Oncol Haematol. September 2004. URL : http://AtlasGeneticsOncology.org/Genes/BRAFID828.html

González-Martínez D, Kim SH, Hu Y, Guimond S, Schofield J, Winyard P, Vannelli GB, Turnbull J, Bouloux PM. Anosmin-1 modulates fibroblast growth factor receptor 1 signaling in human gonadotropin-releasing hormone olfactory neuroblasts through a heparan sulfate-dependent mechanism. J Neurosci. 2004 Nov 17;24(46):10384-92

Grand EK, Grand FH, Chase AJ, Ross FM, Corcoran MM, Oscier DG, Cross NC. Identification of a novel gene, FGFR1OP2, fused to FGFR1 in 8p11 myeloproliferative syndrome. Genes Chromosomes Cancer. 2004 May;40(1):78-83

Ibrahimi OA, Zhang F, Eliseenkova AV, Linhardt RJ, Mohammadi M. Proline to arginine mutations in FGF receptors 1 and 3 result in Pfeiffer and Muenke craniosynostosis

Page 17: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 837

syndromes through enhancement of FGF binding affinity. Hum Mol Genet. 2004 Jan 1;13(1):69-78

Kalkhoven E. CBP and p300: HATs for different occasions. Biochem Pharmacol. 2004 Sep 15;68(6):1145-55

Kwabi-Addo B, Ozen M, Ittmann M. The role of fibroblast growth factors and their receptors in prostate cancer. Endocr Relat Cancer. 2004 Dec;11(4):709-24

Kwabi-Addo B, Wang J, Erdem H, Vaid A, Castro P, Ayala G, Ittmann M. The expression of Sprouty1, an inhibitor of fibroblast growth factor signal transduction, is decreased in human prostate cancer. Cancer Res. 2004 Jul 15;64(14):4728-35

Nobukini T, Thomas G. The mTOR/S6K signalling pathway: the role of the TSC1/2 tumour suppressor complex and the proto-oncogene Rheb. Novartis Found Symp. 2004;262:148-54; discussion 154-9, 265-8

Poole AW, Pula G, Hers I, Crosby D, Jones ML. PKC-interacting proteins: from function to pharmacology. Trends Pharmacol Sci. 2004 Oct;25(10):528-35

Sato N, Katsumata N, Kagami M, Hasegawa T, Hori N, Kawakita S, Minowada S, Shimotsuka A, Shishiba Y, Yokozawa M, Yasuda T, Nagasaki K, Hasegawa D, Hasegawa Y, Tachibana K, Naiki Y, Horikawa R, Tanaka T, Ogata T. Clinical assessment and mutation analysis of Kallmann syndrome 1 (KAL1) and fibroblast growth factor receptor 1 (FGFR1, or KAL2) in five families and 18 sporadic patients. J Clin Endocrinol Metab. 2004 Mar;89(3):1079-88

Shain SA. Exogenous fibroblast growth factors maintain viability, promote proliferation, and suppress GADD45alpha and GAS6 transcript content of prostate cancer cells genetically modified to lack endogenous FGF-2. Mol Cancer Res. 2004 Nov;2(11):653-61

Vizmanos JL, Hernández R, Vidal MJ, Larráyoz MJ, Odero MD, Marín J, Ardanaz MT, Calasanz MJ, Cross NC. Clinical variability of patients with the t(6;8)(q27;p12) and FGFR1OP-FGFR1 fusion: two further cases. Hematol J. 2004;5(6):534-7

Wang F, McKeehan K, Yu C, Ittmann M, McKeehan WL. Chronic activity of ectopic type 1 fibroblast growth factor receptor tyrosine kinase in prostate epithelium results in hyperplasia accompanied by intraepithelial neoplasia. Prostate. 2004 Jan 1;58(1):1-12

Albuisson J, Pêcheux C, Carel JC, Lacombe D, Leheup B, Lapuzina P, Bouchard P, Legius E, Matthijs G, Wasniewska M, Delpech M, Young J, Hardelin JP, Dodé C. Kallmann syndrome: 14 novel mutations in KAL1 and FGFR1 (KAL2). Hum Mutat. 2005 Jan;25(1):98-9

Belloni E, Trubia M, Gasparini P, Micucci C, Tapinassi C, Confalonieri S, Nuciforo P, Martino B, Lo-Coco F, Pelicci PG, Di Fiore PP. 8p11 myeloproliferative syndrome with a novel t(7;8) translocation leading to fusion of the FGFR1 and TIF1 genes. Genes Chromosomes Cancer. 2005 Mar;42(3):320-5

Bryant DM, Stow JL. Nuclear translocation of cell-surface receptors: lessons from fibroblast growth factor. Traffic. 2005 Oct;6(10):947-54

Bryant DM, Wylie FG, Stow JL. Regulation of endocytosis, nuclear translocation, and signaling of fibroblast growth factor receptor 1 by E-cadherin. Mol Biol Cell. 2005 Jan;16(1):14-23

Davies H, Hunter C, Smith R, Stephens P, Greenman C, Bignell G, Teague J, Butler A, Edkins S, Stevens C, Parker A, O'Meara S, Avis T, Barthorpe S, Brackenbury L, Buck G, Clements J, Cole J, Dicks E, Edwards K, Forbes S, Gorton M, Gray K, Halliday K, Harrison R, Hills K, Hinton J, Jones D, Kosmidou V, Laman R, Lugg R, Menzies A, Perry J, Petty R,

Raine K, Shepherd R, Small A, Solomon H, Stephens Y, Tofts C, Varian J, Webb A, West S, Widaa S, Yates A, Brasseur F, Cooper CS, Flanagan AM, Green A, Knowles M, Leung SY, Looijenga LH, Malkowicz B, Pierotti MA, Teh BT, Yuen ST, Lakhani SR, Easton DF, Weber BL, Goldstraw P, Nicholson AG, Wooster R, Stratton MR, Futreal PA. Somatic mutations of the protein kinase gene family in human lung cancer. Cancer Res. 2005 Sep 1;65(17):7591-5

Eswarakumar VP, Lax I, Schlessinger J. Cellular signaling by fibroblast growth factor receptors. Cytokine Growth Factor Rev. 2005 Apr;16(2):139-49

Garcia MJ, Pole JC, Chin SF, Teschendorff A, Naderi A, Ozdag H, Vias M, Kranjac T, Subkhankulova T, Paish C, Ellis I, Brenton JD, Edwards PA, Caldas C. A 1 Mb minimal amplicon at 8p11-12 in breast cancer identifies new candidate oncogenes. Oncogene. 2005 Aug 4;24(33):5235-45

Gelsi-Boyer V, Orsetti B, Cervera N, Finetti P, Sircoulomb F, Rougé C, Lasorsa L, Letessier A, Ginestier C, Monville F, Esteyriès S, Adélaïde J, Esterni B, Henry C, Ethier SP, Bibeau F, Mozziconacci MJ, Charafe-Jauffret E, Jacquemier J, Bertucci F, Birnbaum D, Theillet C, Chaffanet M. Comprehensive profiling of 8p11-12 amplification in breast cancer. Mol Cancer Res. 2005 Dec;3(12):655-67

Jang JH. Reciprocal relationship in gene expression between FGFR1 and FGFR3: implication for tumorigenesis. Oncogene. 2005 Jan 27;24(5):945-8

Karges B, de Roux N. Molecular genetics of isolated hypogonadotropic hypogonadism and Kallmann syndrome. Endocr Dev. 2005;8:67-80

Ornitz DM. FGF signaling in the developing endochondral skeleton. Cytokine Growth Factor Rev. 2005 Apr;16(2):205-13

Rand V, Huang J, Stockwell T, Ferriera S, Buzko O, Levy S, Busam D, Li K, Edwards JB, Eberhart C, Murphy KM, Tsiamouri A, Beeson K, Simpson AJ, Venter JC, Riggins GJ, Strausberg RL. Sequence survey of receptor tyrosine kinases reveals mutations in glioblastomas. Proc Natl Acad Sci U S A. 2005 Oct 4;102(40):14344-9

Sato N, Hasegawa T, Hori N, Fukami M, Yoshimura Y, Ogata T. Gonadotrophin therapy in Kallmann syndrome caused by heterozygous mutations of the gene for fibroblast growth factor receptor 1: report of three families: case report. Hum Reprod. 2005 Aug;20(8):2173-8

St Bernard R, Zheng L, Liu W, Winer D, Asa SL, Ezzat S. Fibroblast growth factor receptors as molecular targets in thyroid carcinoma. Endocrinology. 2005 Mar;146(3):1145-53

Stephens P, Edkins S, Davies H, Greenman C, Cox C, Hunter C, Bignell G, Teague J, Smith R, Stevens C, O'Meara S, Parker A, Tarpey P, Avis T, Barthorpe A, Brackenbury L, Buck G, Butler A, Clements J, Cole J, Dicks E, Edwards K, Forbes S, Gorton M, Gray K, Halliday K, Harrison R, Hills K, Hinton J, Jones D, Kosmidou V, Laman R, Lugg R, Menzies A, Perry J, Petty R, Raine K, Shepherd R, Small A, Solomon H, Stephens Y, Tofts C, Varian J, Webb A, West S, Widaa S, Yates A, Brasseur F, Cooper CS, Flanagan AM, Green A, Knowles M, Leung SY, Looijenga LH, Malkowicz B, Pierotti MA, Teh B, Yuen ST, Nicholson AG, Lakhani S, Easton DF, Weber BL, Stratton MR, Futreal PA, Wooster R. A screen of the complete protein kinase gene family identifies diverse patterns of somatic mutations in human breast cancer. Nat Genet. 2005 Jun;37(6):590-2

Tartaglia M, Gelb BD.. PTPN11 (Protein tyrosine phosphatase, non-receptor type, 11). Atlas Genet Cytogenet Oncol Haematol. February 2005. URL : http://AtlasGeneticsOncology.org/Genes/PTPN11ID41910ch12q24.html

Page 18: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 838

Walz C, Chase A, Schoch C, Weisser A, Schlegel F, Hochhaus A, Fuchs R, Schmitt-Gräff A, Hehlmann R, Cross NC, Reiter A. The t(8;17)(p11;q23) in the 8p11 myeloproliferative syndrome fuses MYO18A to FGFR1. Leukemia. 2005 Jun;19(6):1005-9

White KE, Cabral JM, Davis SI, Fishburn T, Evans WE, Ichikawa S, Fields J, Yu X, Shaw NJ, McLellan NJ, McKeown C, Fitzpatrick D, Yu K, Ornitz DM, Econs MJ. Mutations that cause osteoglophonic dysplasia define novel roles for FGFR1 in bone elongation. Am J Hum Genet. 2005 Feb;76(2):361-7

Xian W, Schwertfeger KL, Vargo-Gogola T, Rosen JM. Pleiotropic effects of FGFR1 on cell proliferation, survival, and migration in a 3D mammary epithelial cell model. J Cell Biol. 2005 Nov 21;171(4):663-73

De Melo VAS, Reid AG.. t(8;13)(p12;q12) in an atypical chronic myeloid leukaemia case. Atlas Genet Cytogenet Oncol Haematol. December 2006. URL : http://AtlasGeneticsOncology.org/Reports/0813ReidID100018.html

Devilard E, Bladou F, Ramuz O, Karsenty G, Dalès JP, Gravis G, Nguyen C, Bertucci F, Xerri L, Birnbaum D. FGFR1 and WT1 are markers of human prostate cancer progression. BMC Cancer. 2006 Nov 30;6:272

Di Bartolomeo S, Cecconi F.. CASP-9 (caspase 9, apoptosis-related cysteine peptidase). Atlas Genet Cytogenet Oncol Haematol. December 2006. URL : http://AtlasGeneticsOncology.org/Genes/CASP9ID423ch1p36.html

Duan W, Villalona-Calero MA.. MDM2 (transformed mouse 3T3 cell double minute 2, p53 binding protein). Atlas Genet Cytogenet Oncol Haematol. December 2006. URL : http://AtlasGeneticsOncology.org/Genes/MDM2ID115ch12q15.html

Farrow EG, Davis SI, Mooney SD, Beighton P, Mascarenhas L, Gutierrez YR, Pitukcheewanont P, White KE. Extended mutational analyses of FGFR1 in osteoglophonic dysplasia. Am J Med Genet A. 2006 Mar 1;140(5):537-9

Fritzsche S, Kenzelmann M, Hoffmann MJ, Müller M, Engers R, Gröne HJ, Schulz WA. Concomitant down-regulation of SPRY1 and SPRY2 in prostate carcinoma. Endocr Relat Cancer. 2006 Sep;13(3):839-49

Furdui CM, Lew ED, Schlessinger J, Anderson KS. Autophosphorylation of FGFR1 kinase is mediated by a sequential and precisely ordered reaction. Mol Cell. 2006 Mar 3;21(5):711-7

Gaughran F, Payne J, Sedgwick PM, Cotter D, Berry M. Hippocampal FGF-2 and FGFR1 mRNA expression in major depression, schizophrenia and bipolar disorder. Brain Res Bull. 2006 Jul 31;70(3):221-7

Hsu T, Adereth Y, Kose N, Dammai V. Endocytic function of von Hippel-Lindau tumor suppressor protein regulates surface localization of fibroblast growth factor receptor 1 and cell motility. J Biol Chem. 2006 Apr 28;281(17):12069-80

Huang X, Yu C, Jin C, Kobayashi M, Bowles CA, Wang F, McKeehan WL. Ectopic activity of fibroblast growth factor receptor 1 in hepatocytes accelerates hepatocarcinogenesis by driving proliferation and vascular endothelial growth factor-induced angiogenesis. Cancer Res. 2006 Feb 1;66(3):1481-90

Jacob AL, Smith C, Partanen J, Ornitz DM. Fibroblast growth factor receptor 1 signaling in the osteo-chondrogenic cell lineage regulates sequential steps of osteoblast maturation. Dev Biol. 2006 Aug 15;296(2):315-28

Klejbor I, Myers JM, Hausknecht K, Corso TD, Gambino AS, Morys J, Maher PA, Hard R, Richards J, Stachowiak EK, Stachowiak MK. Fibroblast growth factor receptor signaling

affects development and function of dopamine neurons - inhibition results in a schizophrenia-like syndrome in transgenic mice. J Neurochem. 2006 Jun;97(5):1243-58

Letessier A, Sircoulomb F, Ginestier C, Cervera N, Monville F, Gelsi-Boyer V, Esterni B, Geneix J, Finetti P, Zemmour C, Viens P, Charafe-Jauffret E, Jacquemier J, Birnbaum D, Chaffanet M. Frequency, prognostic impact, and subtype association of 8p12, 8q24, 11q13, 12p13, 17q12, and 20q13 amplifications in breast cancers. BMC Cancer. 2006 Oct 13;6:245

Mason JM, Morrison DJ, Basson MA, Licht JD. Sprouty proteins: multifaceted negative-feedback regulators of receptor tyrosine kinase signaling. Trends Cell Biol. 2006 Jan;16(1):45-54

Pitteloud N, Acierno JS Jr, Meysing A, Eliseenkova AV, Ma J, Ibrahimi OA, Metzger DL, Hayes FJ, Dwyer AA, Hughes VA, Yialamas M, Hall JE, Grant E, Mohammadi M, Crowley WF Jr. Mutations in fibroblast growth factor receptor 1 cause both Kallmann syndrome and normosmic idiopathic hypogonadotropic hypogonadism. Proc Natl Acad Sci U S A. 2006 Apr 18;103(16):6281-6

Pitteloud N, Meysing A, Quinton R, Acierno JS Jr, Dwyer AA, Plummer L, Fliers E, Boepple P, Hayes F, Seminara S, Hughes VA, Ma J, Bouloux P, Mohammadi M, Crowley WF Jr. Mutations in fibroblast growth factor receptor 1 cause Kallmann syndrome with a wide spectrum of reproductive phenotypes. Mol Cell Endocrinol. 2006 Jul 25;254-255:60-9

Reis-Filho JS, Simpson PT, Turner NC, Lambros MB, Jones C, Mackay A, Grigoriadis A, Sarrio D, Savage K, Dexter T, Iravani M, Fenwick K, Weber B, Hardisson D, Schmitt FC, Palacios J, Lakhani SR, Ashworth A. FGFR1 emerges as a potential therapeutic target for lobular breast carcinomas. Clin Cancer Res. 2006 Nov 15;12(22):6652-62

Trarbach EB, Costa EM, Versiani B, de Castro M, Baptista MT, Garmes HM, de Mendonca BB, Latronico AC. Novel fibroblast growth factor receptor 1 mutations in patients with congenital hypogonadotropic hypogonadism with and without anosmia. J Clin Endocrinol Metab. 2006 Oct;91(10):4006-12

Wang QJ. PKD at the crossroads of DAG and PKC signaling. Trends Pharmacol Sci. 2006 Jun;27(6):317-23

Zenaty D, Bretones P, Lambe C, Guemas I, David M, Léger J, de Roux N. Paediatric phenotype of Kallmann syndrome due to mutations of fibroblast growth factor receptor 1 (FGFR1). Mol Cell Endocrinol. 2006 Jul 25;254-255:78-83

Zhang P, Greendorfer JS, Jiao J, Kelpke SC, Thompson JA. Alternatively spliced FGFR-1 isoforms differentially modulate endothelial cell activation of c-YES. Arch Biochem Biophys. 2006 Jun 1;450(1):50-62

Zhang X, Ibrahimi OA, Olsen SK, Umemori H, Mohammadi M, Ornitz DM. Receptor specificity of the fibroblast growth factor family. The complete mammalian FGF family. J Biol Chem. 2006 Jun 9;281(23):15694-700

Acevedo VD, Gangula RD, Freeman KW, Li R, Zhang Y, Wang F, Ayala GE, Peterson LE, Ittmann M, Spencer DM. Inducible FGFR-1 activation leads to irreversible prostate adenocarcinoma and an epithelial-to-mesenchymal transition. Cancer Cell. 2007 Dec;12(6):559-71

Altomare DA, Testa JR.. AKT2 (v-akt murine thymoma viral oncogene homolog 2). Atlas Genet Cytogenet Oncol Haematol. July 2007. URL : http://AtlasGeneticsOncology.org/Genes/AKT2ID517ch19q13.html

Athauda G, Bottaro DP.. GRB2 (Growth factor receptor-bound protein 2). Atlas Genet Cytogenet Oncol Haematol. May 2007. URL :

Page 19: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 839

http://AtlasGeneticsOncology.org/Genes/GRB2ID386ch17q25.html

Chen H, Ma J, Li W, Eliseenkova AV, Xu C, Neubert TA, Miller WT, Mohammadi M. A molecular brake in the kinase hinge region regulates the activity of receptor tyrosine kinases. Mol Cell. 2007 Sep 7;27(5):717-30

Cunningham ML, Seto ML, Ratisoontorn C, Heike CL, Hing AV. Syndromic craniosynostosis: from history to hydrogen bonds. Orthod Craniofac Res. 2007 May;10(2):67-81

Dikic I, Schmidt MH. Malfunctions within the Cbl interactome uncouple receptor tyrosine kinases from destructive transport. Eur J Cell Biol. 2007 Sep;86(9):505-12

Dodé C, Fouveaut C, Mortier G, Janssens S, Bertherat J, Mahoudeau J, Kottler ML, Chabrolle C, Gancel A, François I, Devriendt K, Wolczynski S, Pugeat M, Pineiro-Garcia A, Murat A, Bouchard P, Young J, Delpech M, Hardelin JP. Novel FGFR1 sequence variants in Kallmann syndrome, and genetic evidence that the FGFR1c isoform is required in olfactory bulb and palate morphogenesis. Hum Mutat. 2007 Jan;28(1):97-8

Elbauomy Elsheikh S, Green AR, Lambros MB, Turner NC, Grainge MJ, Powe D, Ellis IO, Reis-Filho JS. FGFR1 amplification in breast carcinomas: a chromogenic in situ hybridisation analysis. Breast Cancer Res. 2007;9(2):R23

Etienne A, Gelsi-Boyer V, Carbuccia N, Adélaïde J, Barba G, La Starza R, Murati A, Eclache V, Birg F, Birnbaum D, Mozziconacci MJ, Mecucci C, Chaffanet M. Combined translocation with ZNF198-FGFR1 gene fusion and deletion of potential tumor suppressors in a myeloproliferative disorder. Cancer Genet Cytogenet. 2007 Mar;173(2):154-8

Freier K, Schwaenen C, Sticht C, Flechtenmacher C, Mühling J, Hofele C, Radlwimmer B, Lichter P, Joos S. Recurrent FGFR1 amplification and high FGFR1 protein expression in oral squamous cell carcinoma (OSCC). Oral Oncol. 2007 Jan;43(1):60-6

Goldstein M, Meller I, Orr-Urtreger A. FGFR1 over-expression in primary rhabdomyosarcoma tumors is associated with hypomethylation of a 5' CpG island and abnormal expression of the AKT1, NOG, and BMP4 genes. Genes Chromosomes Cancer. 2007 Nov;46(11):1028-38

Kheifets V, Mochly-Rosen D. Insight into intra- and inter-molecular interactions of PKC: design of specific modulators of kinase function. Pharmacol Res. 2007 Jun;55(6):467-76

Kondo T, Zheng L, Liu W, Kurebayashi J, Asa SL, Ezzat S. Epigenetically controlled fibroblast growth factor receptor 2 signaling imposes on the RAS/BRAF/mitogen-activated protein kinase pathway to modulate thyroid cancer progression. Cancer Res. 2007 Jun 1;67(11):5461-70

Leitges M. Functional PKC in vivo analysis using deficient mouse models. Biochem Soc Trans. 2007 Nov;35(Pt 5):1018-20

Liu Z, Neiss N, Zhou S, Henne-Bruns D, Korc M, Bachem M, Kornmann M. Identification of a fibroblast growth factor receptor 1 splice variant that inhibits pancreatic cancer cell growth. Cancer Res. 2007 Mar 15;67(6):2712-9

Memarzadeh S, Xin L, Mulholland DJ, Mansukhani A, Wu H, Teitell MA, Witte ON. Enhanced paracrine FGF10 expression promotes formation of multifocal prostate adenocarcinoma and an increase in epithelial androgen receptor. Cancer Cell. 2007 Dec;12(6):572-85

Murray PJ. The JAK-STAT signaling pathway: input and output integration. J Immunol. 2007 Mar 1;178(5):2623-9

Riley BM, Mansilla MA, Ma J, Daack-Hirsch S, Maher BS, Raffensperger LM, Russo ET, Vieira AR, Dodé C, Mohammadi M, Marazita ML, Murray JC. Impaired FGF signaling

contributes to cleft lip and palate. Proc Natl Acad Sci U S A. 2007 Mar 13;104(11):4512-7

Sahadevan K, Darby S, Leung HY, Mathers ME, Robson CN, Gnanapragasam VJ. Selective over-expression of fibroblast growth factor receptors 1 and 4 in clinical prostate cancer. J Pathol. 2007 Sep;213(1):82-90

Schindler C, Levy DE, Decker T. JAK-STAT signaling: from interferons to cytokines. J Biol Chem. 2007 Jul 13;282(28):20059-63

Shi P, Amin HM.. JAK3 (Janus Kinase 3 or Just Another Kinase 3). Atlas Genet Cytogenet Oncol Haematol. July 2007. URL : http://AtlasGeneticsOncology.org/Genes/JAK3ID41032ch19p13.html

Stachowiak MK, Maher PA, Stachowiak EK. Integrative nuclear signaling in cell development--a role for FGF receptor-1. DNA Cell Biol. 2007 Dec;26(12):811-26

Stevens KR, Rolle MW, Minami E, Ueno S, Nourse MB, Virag JI, Reinecke H, Murry CE. Chemical dimerization of fibroblast growth factor receptor-1 induces myoblast proliferation, increases intracardiac graft size, and reduces ventricular dilation in infarcted hearts. Hum Gene Ther. 2007 May;18(5):401-12

Sugiura K, Ozawa S, Kitagawa Y, Ueda M, Kitajima M. Co-expression of aFGF and FGFR-1 is predictive of a poor prognosis in patients with esophageal squamous cell carcinoma. Oncol Rep. 2007 Mar;17(3):557-64

Winter SF, Acevedo VD, Gangula RD, Freeman KW, Spencer DM, Greenberg NM. Conditional activation of FGFR1 in the prostate epithelium induces angiogenesis with concomitant differential regulation of Ang-1 and Ang-2. Oncogene. 2007 Jul 26;26(34):4897-907

Altomare DA, Testa JR .. FRAP1 (FK506 binding protein 12-rapamycin associated protein 1). Atlas Genet Cytogenet Oncol Haematol. June 2008. URL : http://AtlasGeneticsOncology.org/Genes/FRAP1ID40639ch1p36.html

Boscher C, Mège RM. Cadherin-11 interacts with the FGF receptor and induces neurite outgrowth through associated downstream signalling. Cell Signal. 2008 Jun;20(6):1061-72

Chen G, Wang J, Liu Z, Kornmann M. Exon III splicing of fibroblast growth factor receptor 1 is modulated by growth factors and cyclin D1. Pancreas. 2008 Aug;37(2):159-64

Finetti F, Solito R, Morbidelli L, Giachetti A, Ziche M, Donnini S. Prostaglandin E2 regulates angiogenesis via activation of fibroblast growth factor receptor-1. J Biol Chem. 2008 Jan 25;283(4):2139-46

Gotoh N. Regulation of growth factor signaling by FRS2 family docking/scaffold adaptor proteins. Cancer Sci. 2008 Jul;99(7):1319-25

Hidalgo-Curtis C, Chase A, Drachenberg M, Roberts MW, Finkelstein JZ, Mould S, Oscier D, Cross NC, Grand FH. The t(1;9)(p34;q34) and t(8;12)(p11;q15) fuse pre-mRNA processing proteins SFPQ (PSF) and CPSF6 to ABL and FGFR1. Genes Chromosomes Cancer. 2008 May;47(5):379-85

Kulahin N, Li S, Hinsby A, Kiselyov V, Berezin V, Bock E. Fibronectin type III (FN3) modules of the neuronal cell adhesion molecule L1 interact directly with the fibroblast growth factor (FGF) receptor. Mol Cell Neurosci. 2008 Mar;37(3):528-36

Malavez Y, Gonzalez-Mejia ME, Doseff AI.. PRKCD (protein kinase C, delta). Atlas Genet Cytogenet Oncol Haematol. March 2008. URL :

Page 20: OPEN ACCESS JOURNAL AT INIST-CNRS Gene Sectiondocuments.irevues.inist.fr/.../44609/12-2008-FGFR1113.pdf · 2019. 12. 6. · second and third Ig-like domains of FGFR1. Activation of

FGFR1 (Fibroblast Growth Factor Receptor 1) Huret JL

Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11) 840

http://AtlasGeneticsOncology.org/Genes/PRKCDID42901ch3p21.html

Mozziconacci MJ, Carbuccia N, Prebet T, Charbonnier A, Murati A, Vey N, Chaffanet M, Birnbaum D. Common features of myeloproliferative disorders with t(8;9)(p12;q33) and CEP110-FGFR1 fusion: report of a new case and review of the literature. Leuk Res. 2008 Aug;32(8):1304-8

Nelson TJ, Sun MK, Hongpaisan J, Alkon DL. Insulin, PKC signaling pathways and synaptic remodeling during memory storage and neuronal repair. Eur J Pharmacol. 2008 May 6;585(1):76-87

Park TS, Song J, Kim JS, Yang WI, Song S, Kim SJ, Suh B, Choi JR. 8p11 myeloproliferative syndrome preceded by t(8;9)(p11;q33), CEP110/FGFR1 fusion transcript: morphologic, molecular, and cytogenetic characterization of myeloid neoplasms associated with eosinophilia and FGFR1 abnormality. Cancer Genet Cytogenet. 2008 Mar;181(2):93-9

Richebourg S, Theisen O, Plantier I, Parry A, Soenen-Cornu V, Lepelley P, Preudhomme C, Renneville A, Laï JL, Roche-Lestienne C. Chronic myeloproliferative disorder with t(8;22)(p11;q11) can mime clonal cytogenetic evolution of authentic chronic myelogeneous leukemia. Genes Chromosomes Cancer. 2008 Oct;47(10):915-8

Roux PP.. RPS6KA1 (ribosomal protein S6 kinase, 90kDa, polypeptide 1) Atlas Genet Cytogenet Oncol Haematol. March 2008. URL :

http://AtlasGeneticsOncology.org/Genes/RPS6KA1ID43477ch1p36.html

Salih DA, Brunet A. FoxO transcription factors in the maintenance of cellular homeostasis during aging. Curr Opin Cell Biol. 2008 Apr;20(2):126-36

Steelman LS, Abrams SL, Whelan J, Bertrand FE, Ludwig DE, Bäsecke J, Libra M, Stivala F, Milella M, Tafuri A, Lunghi P, Bonati A, Martelli AM, McCubrey JA. Contributions of the Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT pathways to leukemia. Leukemia. 2008 Apr;22(4):686-707

Tochigi M, Iwamoto K, Bundo M, Sasaki T, Kato N, Kato T. Gene expression profiling of major depression and suicide in the prefrontal cortex of postmortem brains. Neurosci Res. 2008 Feb;60(2):184-91

Turner CA, Calvo N, Frost DO, Akil H, Watson SJ. The fibroblast growth factor system is downregulated following social defeat. Neurosci Lett. 2008 Jan 10;430(2):147-50

Turner CA, Flagel SB, Clinton SM, Akil H, Watson SJ. Cocaine interacts with the novelty-seeking trait to modulate FGFR1 gene expression in the rat. Neurosci Lett. 2008 Dec 3;446(2-3):105-7

Yin Y, Shen WH. PTEN: a new guardian of the genome. Oncogene. 2008 Sep 18;27(41):5443-53

Zenz R, Eferl R, Scheinecker C, Redlich K, Smolen J, Schonthaler HB, Kenner L, Tschachler E, Wagner EF. Activator protein 1 (Fos/Jun) functions in inflammatory bone and skin disease. Arthritis Res Ther. 2008;10(1):201

Lefèvre G, Babchia N, Calipel A, Mouriaux F, Faussat AM, Mrzyk S, Mascarelli F. Activation of the FGF2/FGFR1 autocrine loop for cell proliferation and survival in uveal melanoma cells. Invest Ophthalmol Vis Sci. 2009 Mar;50(3):1047-57

This article should be referenced as such:

Huret JL. FGFR1 (Fibroblast Growth Factor Receptor 1). Atlas Genet Cytogenet Oncol Haematol. 2009; 13(11):821-840.