Signaling by FGFR1 (Homo sapiens)

From WikiPathways

Revision as of 13:53, 16 August 2017 by ReactomeTeam (Talk | contribs)
Jump to: navigation, search
36, 42, 74, 109, 130...4952, 76, 79, 82, 991, 8, 75, 12520, 10597105, 137, 152119, 16294, 97122, 15729, 39, 51, 100, 112...63, 70, 105, 15252, 76, 79, 82, 9932, 92, 131, 15666, 117, 14912, 18, 22, 35, 37...5, 78, 118, 1294958, 119, 17088, 111, 146, 15433, 114, 17036, 7027, 15811577, 89, 104, 14415863, 1521, 8, 55, 83, 12512224, 84, 1343, 11, 19, 21, 26...25, 30, 47, 68, 95...11, 26, 69, 104, 106...44, 93, 15733, 114, 162, 17032, 92, 131, 15636, 7088, 111, 14641, 126288, 108, 146, 15470, 88, 105, 128, 13733, 114, 119, 159, 170210, 4433, 114, 162, 17036, 7054, 103997, 1211, 8, 43, 55, 12596, 98, 10316, 25, 30, 47, 68...58, 119, 17013233, 687, 23, 38, 40, 50...20, 62, 151, 15353, 62, 15252, 76, 79, 82, 9943, 15863, 116, 13470, 116, 146, 15281, 1337, 23, 38, 40, 140...52, 76, 79, 82, 9910, 44, 9852, 76, 79, 99, 161...6, 46, 104, 114, 144...119, 17024, 61, 84, 166101, 13958, 119, 170cytosolActivated FGFR1:FLRTGRB2-1:SOS1UBB(153-228) FGFR1OP2-p-2Y-FGFR1 fusion ActivatedFGFR1:p-FRS2:p-PTPN11:GRB2:GAB1:PI3KBAG4(1-126):FGFR1(208-822) fusion GalNAc-T178-FGF23(25-251) GalNAc-T178-FGF23(25-251) FGF9 FGF20 FGF8-1 FGF23(25-251) p-8Y-FGFR1b FGF2(10-155) FGF10 KL-2 HS GRB2-1 CNTRL-FGFR1 fusion PPP2R1A FGF3 FGF4 FGF3 SOS1 UBC(77-152) MYO18A-p-2Y-FGFR1 fusion ZMYM2-FGFR1 fusion CPSF6-p-2Y-FGFR1 fusion FGF20 KL-1 FGF8-1 ActivatedFGFR1:p-FRS3SPRED1 FGFR1b FGF6 p-8Y-FGFR1c p-6Y-FRS2 KL-2 p-Y55,Y227-SPRY2 ActivatedFGFR1:p-FRS2:GRB2:GAB1:PIK3R1FGFRL1 FGF23(25-251) p-8Y-FGFR1c KL-1 p-8Y-FGFR1b p-8Y- FGFR1 R576W FGF22 Dovitinib Dovitinib Klotho bound toFGF23KL-2 FGFR1OP-p-FGFR1 fusion FGF23(25-251) CBL p-STAT5A, p-STAT5BFGF17-1 FGF5-1 FGFR1OP-FGFR1 fusion FGFR1 P252S FGFR1c homodimerFGFR1c-binding FGFsFGFR1OP-FGFR1 fusion FGF20 STAT3 FGF10 cytosolic p-FGFR1fusion mutantdimersFGF1 FGF4 FGF20 HS NRAS FGF22 FGF23(25-251) ADPFGFR1OP2-p-2Y-FGFR1 fusion FGF3 FGF20 CPSF6-FGFR1 fusion GalNAc-T178-FGF23(25-251) HS p-FGFR1 fusionmutantdimers:PIK3R1UBC(153-228) UBC(1-76) TRIM24-p-2Y-FGFR1 fusion AZD4547 FGF1 ADPADPFGF6 UBA52(1-76) PI(3,4,5)P3RPS27A(1-76) FGFR1 N546K p-8Y-FGFR1c HS UBB(153-228) HS KL-2 FGFR1b-binding FGFsFRS3 FLRT3 GalNAc-T178-FGF23(25-251) KL-1 UBC(609-684) Activated FGFR1chomodimerFGF20 PP2A(A:C):S112/S121-pSPRY2TRIM24-FGFR1 fusion p-8Y-FGFR1b KL-2 p-8Y-FGFR1b p-8Y-FGFR1b FGF1 FGF6 GalNAc-T178-FGF23(25-251) FGF22 FGF6 FGF17-1 FGFR1OP2-p-2Y-FGFR1 fusion GAB1 KL-2 MYO18A-p-2Y-FGFR1 fusion FGF4 FGF6 pY177-BCR-p-FGFR1 fusion p-Y-GAB2 CBL FGF2(10-155) UBC(457-532) ADPp-6Y-FRS2 FGF4 LRRFIP1-p-2Y-FGFR1 fusion FGF17-1 FGF8-1 p-8Y-FGFR1c GalNAc-T178-FGF23(25-251) FGFR1c P252R FGFR1bUBC(77-152) ATPPPP2CB FGF6 FGF22 FGF20 KL-1 FGF10 FGF8-1 HS UBC(381-456) Activated FGFR1FGF23(25-251) p-8Y-FGFR1b ATPFGF1 GAB1 UBA52(1-76) FGF8-1 GDPFGF17-1 FGF5-1 FGFR1(22-763):TACC1(571-805) fusion FGF10 SOS1 FGF17-1 p-8Y-FGFR1b HSFGF2(10-155) BRAFHS FGF10 FGF1 FGF10 FGF20 FGF8-1 FGF1 FGF4 FGF6 p-5Y-FRS3 p-Y55,Y227-SPRY2 ZMYM2-FGFR1 fusion FGF1 Activated FGFR1cbound toFGF23:KlothoUBB(1-76) FGF5-1 UBB(1-76) p-T202,Y204-MAPK3 KL-1 GRB2-1 FGF6 FGF1 ActivatedFGFR1:p-8T-FRS2FGF17-1 FGF5-1 FGF3 GalNAc-T178-FGF23(25-251) cytosolic FGFR1fusion mutantsKL-1 FGF20 BGJ398 GalNAc-T178-FGF23(25-251) GTPGRB2:GAB1HS pY177-BCR-pY-FGFR1mutant:GRB2:p-GAB2FGF6 GRB2-1 GalNAc-T178-FGF23(25-251) UBC(533-608) KL-2 ATPFGFR1 R576W FGFRL1 UBC(305-380) FGF23(25-251) FGF22 FGFR1c UBC(305-380) FGF8-1 FGFR1(22-763):TACC1(571-805) fusion SHC1-2 GAB2 FGF23(25-251) SHC1-2 PPP2CB GalNAc-T178-FGF23(25-251) FGF20 FGFR1 P252S FGF8-1 BCR-p-FGFR1 fusion FGFR1 P252T S111/S120p-SPRY2:B-RAFPPP2CB FGF1 p-5Y-FRS3 FGF8-1 Activated FGFR1bhomodimerBAG4(1-126):FGFR1(208-822) fusion FGF20 FGF10 FGFR1OP-p-FGFR1fusion mutant dimerFGF23(25-251) FGF8-1 UBC(609-684) FGF17-1 FGF1 KL-1 p-FGFR1 mutantfusions:PI3KKL-2 ERLIN2(1-185):p-8Y FGFR1(c.-88-822) fusion HSp-Y194,Y195,Y272-SHC1-3 p-8Y-FGFR1c FGFR1 R576W CNTRL-FGFR1 fusion FGF17-1 STAT5A,STAT5BFGF20 KL-2 GRB2-1 ActivatedFGFR1:p-SHC1:GRB2:SOS1p-6Y FGFR1(22-763):TACC1(571-805) fusion FGFR1OP2-p-2Y-FGFR1 fusion FGF8-1 GRB2:GAB2UBB(153-228) FGF23(25-251) PPP2R1A FGF4 Ub:Y55/Y227-pSPRY2:CBLFGF10 PD173074 UBC(77-152) FGF5-1 ZMYM2-p-2Y-FGFR1 fusion Brivanib FGF2(10-155) FLRT1 GalNAc-T178-FGF23(25-251) GalNAc-T178-FGF23(25-251) FGF3 FLRT2 FGF1 p-Y239,Y240,Y317-SHC1-2 FGF2(10-155) pY-STAT1,3BRAF KL-1 FGF18 pY177-BCR-p-FGFR1 fusion ActivatedFGFR1:p-FRS:p-PTPN11FGFR1OP-p-FGFR1 fusion FGF6 FGF20 SU5402 Plasma membraneFGFR1 fusion dimersFGF1 FGF9 FGF8-1 FGF6 FGF5-1 UBB(153-228) PIK3R1 FGF8-1 STAT1,3KL-1 GalNAc-T178-FGF23(25-251) FGF17-1 FGF1 HS Dovitinib p-8Y-FGFR1c PPP2R1A GDP UBC(229-304) SPRY2 FGF20 p-8Y-FGFR1b FGF23(25-251) HS FGFR1 P252X mutantsE3810 p-S111,S120-SPRY2 FGF6 FGF4 UBB(153-228) E3810 FGF3 p-Y546,Y584-PTPN11 GRB2-1 ADPKL-1 FGFRL1-bindingFGFs:FGFRL1 dimerFGFR1 mutants withenhanced kinaseactivitycytosolic FGFR1fusion mutantdimersPIK3R1 FGF5-1 FGF2(10-155) FGF22 FGF8-1 p-6Y-FRS2 ADPp-8Y-FGFR1c CUX1-FGFR1 fusion UBB(77-152) SPRY2 PPP2R1A FGF8-1 cytosolic p-FGFR1fusion mutantdimersHS GalNAc-T178-FGF23(25-251) TRIM24-FGFR1 fusion ATPUbFGF5-1 FGF3 FGF5-1 UBC(381-456) PIK3CA GalNAc-T178-FGF23(25-251) FGF6 FGF17-1 FGF17-1 ADPFGF2(10-155) FGF4 FGF23(25-251) FGF8-1 p-8Y-FGFR1b FGF22 PPP2CA FGF22 FGF6 ActivatedFGFR1:p-FRS2:p-PTPN11FGF5-1 CPSF6-p-2Y-FGFR1 fusion UBC(609-684) GalNAc-T178-FGF23(25-251) p-8Y-FGFR1c PIP3 activates AKTsignalingFGF23(25-251) p-8Y-FGFR1c ZMYM2-p-2Y-FGFR1 fusion FRS2 PPP2CA UBC(229-304) GalNAc-T178-FGF23(25-251) PP2A (A:C)FGF9 pY177-BCR-p-FGFR1 fusion PPP2CA UBC(609-684) UBC(153-228) FGF4 Y55/Y227-pSPRY2:CBLFGFR1OP2-FGFR1 fusion FGF1 MYO18A-FGFR1 fusion FGF22 MYO18A-p-2Y-FGFR1 fusion FGF5-1 GalNAc-T178-FGF23(25-251) FGF20 TRIM24-FGFR1 fusion GRB2-1 KL-2 GalNAc-T178-FGF23(25-251) SU5402 GAB1 HS FGF5-1 FGF6 GalNAc-T178-FGF23(25-251) FGF17-1 PPP2R1A FGF17-1 PP2A(A:C):Y55/Y227-pSPRY2FGF22 FGFR1cATPFGF23(25-251) FGF2(10-155) MYO18A-FGFR1 fusion FGF4 FGF4 FGFR1OP2-FGFR1 fusion CBLGalNAc-T178-FGF23(25-251) GalNAc-T178-FGF23(25-251) FGF2(10-155) activatedFGFR1:p-4Y-PLCG1FGF17-1 HS UBC(305-380) FGF4 FGF20 KL-2 FGF3 FGFRL1dimer:SPRED1/2dimerp-8Y-FGFR1c FGF3 UBB(77-152) UBB(77-152) FGFR1c KL-2 FGF3 PIK3R1 MYO18A-FGFR1 fusion FGF1 FGFR1OP-p-FGFR1 fusion FGF10 FGF4 FGF4 KL-2 FGF22 RPS27A(1-76) FGF9 ATPp-Y701-STAT1 p-Y-GAB2 FGF17-1 BCR-FGFR1 fusion FGF10 FGF17-1 p-8Y-FGFR1b KL-1 FGF23(25-251) FGF10 FGF22 UBC(77-152) PPP2CA RPS27A(1-76) ADPSPRED2 FGF22 FGF5-1 p-8Y-FGFR1b RPS27A(1-76) ATPGAB1 p-6Y-FRS2 FGF23(25-251) FGF2(10-155) p-8Y-FGFR1c FGF20 p-Y177-BCR-pY-FGFR1mutant:GRB2:p-GAB2:PIK3R1FGF23(25-251) FGF5-1 ZMYM2-p-2Y-FGFR1 fusion PPP2CB FGF20 ATPFGFR1OP-p-FGFR1 fusion p-Y699-STAT5B FGF4 p-S111,S120-SPRY2FGF2(10-155) KAL1 AZD4547 FGF8-1 UBB(1-76) TRIM24-p-2Y-FGFR1 fusion p-8Y-FGFR1c BCR-p-FGFR1 fusion HS UBA52(1-76) ActivatedFGFR:p-FRS2:p-PTPN11:GRB2:GAB1:PIK3R1GalNAc-T178-FGF23(25-251) FGF9 FGF17-1 FGF3 HS GAB1 p-8Y-FGFR1c p-8Y-FGFR1c FGF22 FGF4 FGF17-1 PIK3R1 p-8Y-FGFR1b ATPPI(4,5)P2UBB(1-76) p-6Y-FRS2 FGF10 FGF6 RPS27A(1-76) GAB2 p-Y546,Y584-PTPN11 BCR-p-FGFR1 fusion FGF9 FGF2(10-155) FLRT1 FGF9 FGF3 FGF2(10-155) BCR-FGFR1 fusion p-5Y-FRS3 plasma membraneFGFR1 fusionsPI(3,4,5)P3FGF8-1 RAF/MAP kinasecascadeFGF22 ActivatedFGFR1:p-FRSp-8Y-FGFR1c FGF3 FGF1 FGF20 FGF10 FGF5-1 CNTRL-p-2Y-FGFR1 fusion FGF8-1 HSFGFRL1 pY177-BCR-p-FGFR1fusion mutant dimerFGF3 GRB2-1 p-8Y-FGFR1c FGF22 FGF3 KL-2 PD173074 FGF5-1 UBC(305-380) PP2A(A:C):SPRY2FGF2(10-155) FGF4 UBC(609-684) PIK3R1FGF1 FGF23(25-251) UBB(77-152) UBC(1-76) FGF6 UBC(381-456) FGF2(10-155) FGFR1c FGF10 PD173074 FGF4 STAT5A ATPPPA2A (A:C):Y55/Y227p-SPRY2:GRB2FGF10 ATPFGF20 UBC(533-608) LRRFIP1-FGFR1 fusion FGF20 BCR-FGFR1 fusion FGF2(10-155) FGF2(10-155) ATPCUX1-p-2Y-FGFR1 fusion FGF9 FGF2(10-155) GRB2-1 KRAS FGF9 BAG4(1-126):p-8Y FGFR1(208-822) fusion UBC(229-304) FGF3 FGF9 FGF5-1 KL-2 PPP2CB FGF23(25-251) p-5Y-FRS3 FGF22 p-8Y-FGFR1b FGF5-1 FGF17-1 Overexpressed FGFR1homodimersHS FGF10 FGFR1 P252T p-4Y-PLCG1 Ub-Activated FGFR1complex:Ub-p-FRS2KL-1 FGFRL1-binding FGFsPPP2R1A FGF20 PIK3R1 PPA2A(A:C):S112/S115p-SPRY2UBC(153-228) KL-2 SPRY2 ZMYM2-p-2Y-FGFR1 fusion KL-1 SPRED1 PPP2R1A FGF4 p-Y546,Y584-PTPN11 FGF20 FGF4 FGF8-1 KRAS UBC(229-304) HS OverexpressedFGFR1:TKIsHSFGFR1c P252R KL-1 SU5402 FGF5-1 Activated FGFR1:SHC1p-6Y-FRS2 FGF4 HS ADPFGF9 FGF10 FGF6 UBC(457-532) PTPN11 FGF3 FGF4 CNTRL-p-2Y-FGFR1 fusion HS FGF2(10-155) FGF20 FGF6 FGFR1b KL-2 GRB2-1 FGF2(10-155) FGF10 KL-2 KL-1 FGF8-1 FGF1 LRRFIP1-FGFR1 fusion GalNAc-T178-FGF23(25-251) Midostaurin STAT5B HS FGF23(25-251) FGF5-1 p-8T-FRS2 KL-1 ADPFGF1 FLRT3 p-8Y-FGFR1c FGF23(25-251) GalNAc-T178-FGF23(25-251) FGFR1b UBB(77-152) UBC(457-532) FGF9 pY177-BCR-p-FGFR1 fusion p-8Y-FGFR1c FGF9 FGF5-1 FGF6 p-Y371-CBL HS FGF9 CUX1-p-2Y-FGFR1 fusion FGF2(10-155) BGJ398 Brivanib alaninate Tyrosine kinaseinhibitors ofoverexpressed FGFR1FGF5-1 UBC(1-76) BCR-p-FGFR1 fusion p-6Y-FRS2 HS FGF23 bound toKlotho and FGFR1cFGF5-1 FGF9 Brivanib alaninate FGF17-1 PLCG1 GalNAc-T178-FGF23(25-251) Tyrosine kinaseinhibitors of FGFR1fusion mutantsActivatedFGFR1:p-SHC1KL-2 CNTRL-p-2Y-FGFR1 fusion FGF10 p-8Y-FGFR1b GRB2-1 UBC(533-608) FGF8-1 KL-2 p-8Y-FGFR1c PPP2CA FGF1 GRB2-1 FGF1 FGF9 p-8Y-FGFR1b ActivatedFGFR1:p-FRS2:GRB2:GAB1:PI3Kp-8Y-FGFR1 K656E PIK3R1 FGF17-1 FGF9 FGF4 FGF9 KL-1 FGF3 p-Y55,Y227-SPRY2 p-8Y-FGFR1 P252T HRAS HS FGF10 GalNAc-T178-FGF23(25-251) FGF1 PIK3R1 FGF8-1 KL-2 FGFR1 P252X mutantdimers bound toFGFsSHC1 p46,p52p-8Y-FGFR1c HS FGFR1c ADPFGF5-1 FGFR1c:KAL1UBC(381-456) Activated FGFR1mutant dimers withenhanced kinaseactivityKL-1 KL-2 FGF9 FGF9 CPSF6-p-2Y-FGFR1 fusion ATPERLIN2(1-185):FGFR1(c.-88-822) fusion FGF22 p-8Y-FGFR1b KAL1 FGF17-1 p-Y-GAB2 TRIM24-p-2Y-FGFR1 fusion LRRFIP1-FGFR1 fusion FGF10 p-Y239,Y240,Y317-SHC1-2 FGFR1c KL-2 FGF2(10-155) p21 RAS:GDPPIK3CA FGF22 FGF2(10-155) FGF4 HS FGF9 HS FGF23(25-251) DAG and IP3signalingFGFR1c homodimerbound to FGFFGF3 FGF2(10-155) FGFR1 K656E p-8Y-FGFR1c P252R p-8Y-FGFR1b p-6Y-FRS2 FGFR1 N546K CUX1-p-2Y-FGFR1 fusion ATPFGF10 UBC(77-152) Brivanib PPP2CA GalNAc-T178-FGF23(25-251) FGF22 FGF22 FGF3 p-8Y-FGFR1c ERLIN2(1-185):FGFR1(c.-88-822) fusion ATPFGF10 PPA2A(A:C):SPRY2p-8Y-FGFR1 N546K p-Y194,Y195,Y272-SHC1-3 Activated FGFR1:FRS2FGF18 FGFR1OP-FGFR1 fusion FGF5-1 GRB2-1 FGF3 LRRFIP1-p-2Y-FGFR1 fusion FGF2(10-155) UBB(1-76) ActivatedFGFR1:p-FRS:PTPN11FGF9 FGFR1OP2-FGFR1 fusion HS UBC(1-76) pY177-BCR-pY-FGFR1mutant:GRB2:p-GAB1:PI3KFGF22 FGF4 FGF4 FGF20 UBC(153-228) PIK3CAHS FGF1 p-6Y-FRS2 ActivatedFGFR1:p-FRS2:GRB2:SOS1ZMYM2-FGFR1 fusion FGF5-1 pY177-BCR1-p-FGFR1mutant:GRB2:GAB2UBC(457-532) FGF1 GRB2-1 p-8Y-FGFR1b p-Y371-CBL:GRB2FGF23(25-251) KL-2 LRRFIP1-p-2Y-FGFR1 fusion KL-1 FLRT2 FGF10 p-8Y-FGFR1b FGF23(25-251) FGF3 p-8Y-FGFR1 P252S HS FGF22 SOS1 FGF4 FGF2(10-155) GalNAc-T178-FGF23(25-251) FGF20 ATPGRB2:GAB1:PIK3R1ADPp-8Y-FGFR1c p-Y371-CBL HS FGF2(10-155) FGF6 UBC(457-532) FGF9 p-Y371-CBL FRS3FGF8-1 FGF17-1 FGF2(10-155) CNTRL-FGFR1 fusion p-8Y-FGFR1b FGF6 GRB2-1 FGFR1 mutant dimerswith enhancedkinase activityFGF17-1 FGF9 p-6Y-FRS2 BCR-p-FGFR1 fusion FGF17-1 Midostaurin FGF2(10-155) FGF8-1 KL-1 FGF4 PPP2CB FGF2(10-155) FGF10 PIK3CA PPP2CA KAL1FGF3 FGF17-1 Ub-(Y55/Y227)p-SPRY2FGF1 CPSF6-FGFR1 fusion activatedFGFR1:PLCG1p-8Y-FGFR1c KL-1 p-8Y-FGFR1b ADPSU5402 ADPFGF3 p-T250,T255,T385,S437-MKNK1p-6Y-FRS2 p-4Y-PLCG1p-S112,S121-SPRY2 FGF5-1 FGF1 HS SPRED1/2 dimerFGF8-1 KL-2 FGF2(10-155) NRAS Activated FGFR1:FRS3KL-2 p-Y705-STAT3 UBC(229-304) FGF20 Plasma membrane p-YFGFR1 fusion dimersp-Y546,Y584-PTPN11 p-Y546,Y584-PTPN11 FGF22 FGF22 FGF22 STAT1 FGF8-1 p-T185,Y187-MAPK1 p-Y55,Y227-SPRY2 PTPN11ATPDovitinib ADPFGF17-1 FGF6 p-8Y-FGFR1b FLRT1,2,3p-Y55,Y227-SPRY2 SRC-1GRB2-1 AZ 2171 ADPFGF1 ADPFGF6 PPP2CB FGFR1c FGFR1b ActivatedoverexpressedFGFR1c homodimerSHC1-3 FGF4 FGFR1 K656E CUX1-FGFR1 fusion FGF4 FGF9 FGF17-1 PD173074 BRAF KAL1:HSFGF8-1 FGF2(10-155) p-5Y-FRS3 FGF5-1 FGF23(25-251) p-6Y-FRS2 ActivatedFGFR1:p-FRS2:p-PTPN11:p-CBL:GRB2GRB2-1 FGF10 FGF6 KL-1 FGF17-1 UBC(533-608) p-8Y-FGFR1c HRAS UBC(153-228) p-Y546,Y584-PTPN11 FGF1 FGF6 BCR-p-FGFR1 fusionmutant dimerp-S112,S115-SPRY2 PLCG1ATPKL-1 ATPSHC1-3 FGF6 ADPFGF3 FGF1 p-Y694-STAT5A FGF1 UBA52(1-76) FGF9 FGFRL1 dimerPIK3R1 ActivatedFGFR1:p-FRS2FGF10 PiFGF22 Activated FGFR1P252X mutantsKL-1 GTP FGFR1b homodimerbound to FGFFGF8-1 SPRY2:B-RAFFGF10 ADPUBC(533-608) GRB2-1 p21 RAS:GTPHS UBC(1-76) FGF6 p-8Y-FGFR1b PI(3,4,5)P3 FGF3 FGF8-1 PIK3CA ADPFGF2(10-155) ActivatedoverexpressedFGFR1b homodimerp-8Y-FGFR1c AZ 2171 FGF9 UBC(381-456) HS FGF22 ATPGRB2-1 UbATPp-8Y-FGFR1b CPSF6-FGFR1 fusion GAB1 FGF5-1 FGFR1 fusion mutantdimers:TKIsKL-1 GalNAc-T178-FGF23(25-251) GRB2-1UBA52(1-76) FGFR1b homodimerFGF20 CUX1-FGFR1 fusion ADPFGF17-1 CUX1-p-2Y-FGFR1 fusion FGF1 KL-1 p-T,Y MAPK dimersFGF5-1 FGF23(25-251) SPRED2 FGF9 FGF23(25-251) FGF3 p-8Y-FGFR1c FRS2UBC(305-380) PIK3R1 p-8Y-FGFR1b pY177-BCR-p-FGFR1 fusion FGF3 17010714, 59, 148, 150854817013, 102115721, 26, 1559221, 26, 15517013, 10221, 26, 15517035117, 5613185928210792855717, 564831069213, 1024831, 60, 65, 71, 76...8249047107118217097114, 120, 144486, 46, 104, 114, 170114, 120, 1445114, 59, 148, 15011122571701449729, 132928076, 7947518514, 59, 148, 15051, 112, 13230, 681547, 9521, 26, 1551199711930, 688213134, 67, 72, 87, 110...921441444897112, 163112, 1631702028, 90107905785574831061117021, 26, 15547, 951121, 26, 15521, 26, 15547, 951068115817010729, 132112, 1635731110738013, 10210714417017010610614414428, 9014417076, 7985170314, 59, 148, 15048106131


Description

The 22 members of the fibroblast growth factor (FGF) family of growth factors mediate their cellular responses by binding to and activating the different isoforms encoded by the four receptor tyrosine kinases (RTKs) designated FGFR1, FGFR2, FGFR3 and FGFR4. These receptors are key regulators of several developmental processes in which cell fate and differentiation to various tissue lineages are determined. Unlike other growth factors, FGFs act in concert with heparin or heparan sulfate proteoglycan (HSPG) to activate FGFRs and to induce the pleiotropic responses that lead to the variety of cellular responses induced by this large family of growth factors. An alternative, FGF-independent, source of FGFR activation originates from the interaction with cell adhesion molecules, typically in the context of interactions on neural cell membranes and is crucial for neuronal survival and development.

Upon ligand binding, receptor dimers are formed and their intrinsic tyrosine kinase is activated causing phosphorylation of multiple tyrosine residues on the receptors. These then serve as docking sites for the recruitment of SH2 (src homology-2) or PTB (phosphotyrosine binding) domains of adaptors, docking proteins or signaling enzymes. Signaling complexes are assembled and recruited to the active receptors resulting in a cascade of phosphorylation events.

This leads to stimulation of intracellular signaling pathways that control cell proliferation, cell differentiation, cell migration, cell survival and cell shape, depending on the cell type or stage of maturation.
View original pathway at:Reactome.

Comments

Reactome-Converter 
Pathway is converted from Reactome ID: 5654736
Reactome-version 
Reactome version: 61
Reactome Author 
Reactome Author: de Bono, Bernard

Try the New WikiPathways

View approved pathways at the new wikipathways.org.

Quality Tags

Ontology Terms

 

Bibliography

View all...
  1. Smedley D, Demiroglu A, Abdul-Rauf M, Heath C, Cooper C, Shipley J, Cross NC.; ''ZNF198-FGFR1 transforms Ba/F3 cells to growth factor independence and results in high level tyrosine phosphorylation of STATS 1 and 5.''; PubMed Europe PMC Scholia
  2. Soussi-Yanicostas N, Faivre-Sarrailh C, Hardelin JP, Levilliers J, Rougon G, Petit C.; ''Anosmin-1 underlying the X chromosome-linked Kallmann syndrome is an adhesion molecule that can modulate neurite growth in a cell-type specific manner.''; PubMed Europe PMC Scholia
  3. Greulich H, Pollock PM.; ''Targeting mutant fibroblast growth factor receptors in cancer.''; PubMed Europe PMC Scholia
  4. Chase A, Grand FH, Cross NC.; ''Activity of TKI258 against primary cells and cell lines with FGFR1 fusion genes associated with the 8p11 myeloproliferative syndrome.''; PubMed Europe PMC Scholia
  5. Hall AB, Jura N, DaSilva J, Jang YJ, Gong D, Bar-Sagi D.; ''hSpry2 is targeted to the ubiquitin-dependent proteasome pathway by c-Cbl.''; PubMed Europe PMC Scholia
  6. Ong SH, Guy GR, Hadari YR, Laks S, Gotoh N, Schlessinger J, Lax I.; ''FRS2 proteins recruit intracellular signaling pathways by binding to diverse targets on fibroblast growth factor and nerve growth factor receptors.''; PubMed Europe PMC Scholia
  7. Trueb B, Amann R, Gerber SD.; ''Role of FGFRL1 and other FGF signaling proteins in early kidney development.''; PubMed Europe PMC Scholia
  8. Petiot A, Ferretti P, Copp AJ, Chan CT.; ''Induction of chondrogenesis in neural crest cells by mutant fibroblast growth factor receptors.''; PubMed Europe PMC Scholia
  9. Dutt A, Salvesen HB, Chen TH, Ramos AH, Onofrio RC, Hatton C, Nicoletti R, Winckler W, Grewal R, Hanna M, Wyhs N, Ziaugra L, Richter DJ, Trovik J, Engelsen IB, Stefansson IM, Fennell T, Cibulskis K, Zody MC, Akslen LA, Gabriel S, Wong KK, Sellers WR, Meyerson M, Greulich H.; ''Drug-sensitive FGFR2 mutations in endometrial carcinoma.''; PubMed Europe PMC Scholia
  10. Gartside MG, Chen H, Ibrahimi OA, Byron SA, Curtis AV, Wellens CL, Bengston A, Yudt LM, Eliseenkova AV, Ma J, Curtin JA, Hyder P, Harper UL, Riedesel E, Mann GJ, Trent JM, Bastian BC, Meltzer PS, Mohammadi M, Pollock PM.; ''Loss-of-function fibroblast growth factor receptor-2 mutations in melanoma.''; PubMed Europe PMC Scholia
  11. Yu K, Herr AB, Waksman G, Ornitz DM.; ''Loss of fibroblast growth factor receptor 2 ligand-binding specificity in Apert syndrome.''; PubMed Europe PMC Scholia
  12. 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.''; PubMed Europe PMC Scholia
  13. Cargnello M, Roux PP.; ''Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases.''; PubMed Europe PMC Scholia
  14. Gerber SD, Steinberg F, Beyeler M, Villiger PM, Trueb B.; ''The murine Fgfrl1 receptor is essential for the development of the metanephric kidney.''; PubMed Europe PMC Scholia
  15. 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.''; PubMed Europe PMC Scholia
  16. Dodé C, Hardelin JP.; ''Kallmann syndrome.''; PubMed Europe PMC Scholia
  17. Zhen Y, Sørensen V, Jin Y, Suo Z, Wiedłocha A.; ''Indirubin-3'-monoxime inhibits autophosphorylation of FGFR1 and stimulates ERK1/2 activity via p38 MAPK.''; PubMed Europe PMC Scholia
  18. Ollendorff V, Guasch G, Isnardon D, Galindo R, Birnbaum D, Pébusque MJ.; ''Characterization of FIM-FGFR1, the fusion product of the myeloproliferative disorder-associated t(8;13) translocation.''; PubMed Europe PMC Scholia
  19. Hu Y, Bouloux PM.; ''Novel insights in FGFR1 regulation: lessons from Kallmann syndrome.''; PubMed Europe PMC Scholia
  20. Williams EJ, Furness J, Walsh FS, Doherty P.; ''Activation of the FGF receptor underlies neurite outgrowth stimulated by L1, N-CAM, and N-cadherin.''; PubMed Europe PMC Scholia
  21. Popovici C, Adélaïde J, Ollendorff V, Chaffanet M, Guasch G, Jacrot M, Leroux D, Birnbaum D, Pébusque MJ.; ''Fibroblast growth factor receptor 1 is fused to FIM in stem-cell myeloproliferative disorder with t(8;13).''; PubMed Europe PMC Scholia
  22. Brown MD, Sacks DB.; ''Protein scaffolds in MAP kinase signalling.''; PubMed Europe PMC Scholia
  23. Lim J, Yusoff P, Wong ES, Chandramouli S, Lao DH, Fong CW, Guy GR.; ''The cysteine-rich sprouty translocation domain targets mitogen-activated protein kinase inhibitory proteins to phosphatidylinositol 4,5-bisphosphate in plasma membranes.''; PubMed Europe PMC Scholia
  24. Robertson A, MacColl GS, Nash JA, Boehm MK, Perkins SJ, Bouloux PM.; ''Molecular modelling and experimental studies of mutation and cell-adhesion sites in the fibronectin type III and whey acidic protein domains of human anosmin-1.''; PubMed Europe PMC Scholia
  25. Roskoski R.; ''MEK1/2 dual-specificity protein kinases: structure and regulation.''; PubMed Europe PMC Scholia
  26. 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.''; PubMed Europe PMC Scholia
  27. Cross MJ, Hodgkin MN, Roberts S, Landgren E, Wakelam MJ, Claesson-Welsh L.; ''Tyrosine 766 in the fibroblast growth factor receptor-1 is required for FGF-stimulation of phospholipase C, phospholipase D, phospholipase A(2), phosphoinositide 3-kinase and cytoskeletal reorganisation in porcine aortic endothelial cells.''; PubMed Europe PMC Scholia
  28. Tavormina PL, Bellus GA, Webster MK, Bamshad MJ, Fraley AE, McIntosh I, Szabo J, Jiang W, Jiang W, Jabs EW, Wilcox WR, Wasmuth JJ, Donoghue DJ, Thompson LM, Francomano CA.; ''A novel skeletal dysplasia with developmental delay and acanthosis nigricans is caused by a Lys650Met mutation in the fibroblast growth factor receptor 3 gene.''; PubMed Europe PMC Scholia
  29. Urakawa I, Yamazaki Y, Shimada T, Iijima K, Hasegawa H, Okawa K, Fujita T, Fukumoto S, Yamashita T.; ''Klotho converts canonical FGF receptor into a specific receptor for FGF23.''; PubMed Europe PMC Scholia
  30. Rubin C, Litvak V, Medvedovsky H, Zwang Y, Lev S, Yarden Y.; ''Sprouty fine-tunes EGF signaling through interlinked positive and negative feedback loops.''; PubMed Europe PMC Scholia
  31. 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).''; PubMed Europe PMC Scholia
  32. Roskoski R.; ''ERK1/2 MAP kinases: structure, function, and regulation.''; PubMed Europe PMC Scholia
  33. 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.''; PubMed Europe PMC Scholia
  34. Wu Y, Chen Z, Ullrich A.; ''EGFR and FGFR signaling through FRS2 is subject to negative feedback control by ERK1/2.''; PubMed Europe PMC Scholia
  35. Gotoh N.; ''Regulation of growth factor signaling by FRS2 family docking/scaffold adaptor proteins.''; PubMed Europe PMC Scholia
  36. Chen J, Deangelo DJ, Kutok JL, Williams IR, Lee BH, Wadleigh M, Duclos N, Cohen S, Adelsperger J, Okabe R, Coburn A, Galinsky I, Huntly B, Cohen PS, Meyer T, Fabbro D, Roesel J, Banerji L, Griffin JD, Xiao S, Fletcher JA, Stone RM, Gilliland DG.; ''PKC412 inhibits the zinc finger 198-fibroblast growth factor receptor 1 fusion tyrosine kinase and is active in treatment of stem cell myeloproliferative disorder.''; PubMed Europe PMC Scholia
  37. Wang R, Wang L, Li Y, Hu H, Shen L, Shen X, Pan Y, Ye T, Zhang Y, Luo X, Zhang Y, Pan B, Li B, Li H, Zhang J, Pao W, Ji H, Sun Y, Chen H.; ''FGFR1/3 tyrosine kinase fusions define a unique molecular subtype of non-small cell lung cancer.''; PubMed Europe PMC Scholia
  38. Soler G, Nusbaum S, Varet B, Macintyre EA, Vekemans M, Romana SP, Radford-Weiss I.; ''LRRFIP1, a new FGFR1 partner gene associated with 8p11 myeloproliferative syndrome.''; PubMed Europe PMC Scholia
  39. 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.''; PubMed Europe PMC Scholia
  40. Harkiolaki M, Tsirka T, Lewitzky M, Simister PC, Joshi D, Bird LE, Jones EY, O'Reilly N, Feller SM.; ''Distinct binding modes of two epitopes in Gab2 that interact with the SH3C domain of Grb2.''; PubMed Europe PMC Scholia
  41. Eswarakumar VP, Lax I, Schlessinger J.; ''Cellular signaling by fibroblast growth factor receptors.''; PubMed Europe PMC Scholia
  42. 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.''; PubMed Europe PMC Scholia
  43. Lao DH, Yusoff P, Chandramouli S, Philp RJ, Fong CW, Jackson RA, Saw TY, Yu CY, Guy GR.; ''Direct binding of PP2A to Sprouty2 and phosphorylation changes are a prerequisite for ERK inhibition downstream of fibroblast growth factor receptor stimulation.''; PubMed Europe PMC Scholia
  44. Gotoh N, Laks S, Nakashima M, Lax I, Schlessinger J.; ''FRS2 family docking proteins with overlapping roles in activation of MAP kinase have distinct spatial-temporal patterns of expression of their transcripts.''; PubMed Europe PMC Scholia
  45. Jacquemier J, Adelaide J, Parc P, Penault-Llorca F, Planche J, deLapeyriere O, Birnbaum D.; ''Expression of the FGFR1 gene in human breast-carcinoma cells.''; PubMed Europe PMC Scholia
  46. Mohammadi M, McMahon G, Sun L, Tang C, Hirth P, Yeh BK, Hubbard SR, Schlessinger J.; ''Structures of the tyrosine kinase domain of fibroblast growth factor receptor in complex with inhibitors.''; PubMed Europe PMC Scholia
  47. Guasch G, Ollendorff V, Borg JP, Birnbaum D, Pébusque MJ.; ''8p12 stem cell myeloproliferative disorder: the FOP-fibroblast growth factor receptor 1 fusion protein of the t(6;8) translocation induces cell survival mediated by mitogen-activated protein kinase and phosphatidylinositol 3-kinase/Akt/mTOR pathways.''; PubMed Europe PMC Scholia
  48. Hart KC, Robertson SC, Donoghue DJ.; ''Identification of tyrosine residues in constitutively activated fibroblast growth factor receptor 3 involved in mitogenesis, Stat activation, and phosphatidylinositol 3-kinase activation.''; PubMed Europe PMC Scholia
  49. Ong SH, Goh KC, Lim YP, Low BC, Klint P, Claesson-Welsh L, Cao X, Tan YH, Guy GR.; ''Suc1-associated neurotrophic factor target (SNT) protein is a major FGF-stimulated tyrosine phosphorylated 90-kDa protein which binds to the SH2 domain of GRB2.''; PubMed Europe PMC Scholia
  50. Koziczak M, Holbro T, Hynes NE.; ''Blocking of FGFR signaling inhibits breast cancer cell proliferation through downregulation of D-type cyclins.''; PubMed Europe PMC Scholia
  51. Mohi MG, Neel BG.; ''The role of Shp2 (PTPN11) in cancer.''; PubMed Europe PMC Scholia
  52. Chardin P, Camonis JH, Gale NW, van Aelst L, Schlessinger J, Wigler MH, Bar-Sagi D.; ''Human Sos1: a guanine nucleotide exchange factor for Ras that binds to GRB2.''; PubMed Europe PMC Scholia
  53. Silva PN, Altamentova SM, Kilkenny DM, Rocheleau JV.; ''Fibroblast growth factor receptor like-1 (FGFRL1) interacts with SHP-1 phosphatase at insulin secretory granules and induces beta-cell ERK1/2 protein activation.''; PubMed Europe PMC Scholia
  54. Lelièvre H, Chevrier V, Tassin AM, Birnbaum D.; ''Myeloproliferative disorder FOP-FGFR1 fusion kinase recruits phosphoinositide-3 kinase and phospholipase Cgamma at the centrosome.''; PubMed Europe PMC Scholia
  55. DaSilva J, Xu L, Kim HJ, Miller WT, Bar-Sagi D.; ''Regulation of sprouty stability by Mnk1-dependent phosphorylation.''; PubMed Europe PMC Scholia
  56. Schüller AC, Ahmed Z, Levitt JA, Suen KM, Suhling K, Ladbury JE.; ''Indirect recruitment of the signalling adaptor Shc to the fibroblast growth factor receptor 2 (FGFR2).''; PubMed Europe PMC Scholia
  57. Sleeman M, Fraser J, McDonald M, Yuan S, White D, Grandison P, Kumble K, Watson JD, Murison JG.; ''Identification of a new fibroblast growth factor receptor, FGFR5.''; PubMed Europe PMC Scholia
  58. McClatchey AI, Cichowski K.; ''SPRED proteins provide a NF-ty link to Ras suppression.''; PubMed Europe PMC Scholia
  59. Hu Y, González-Martínez D, Kim SH, Bouloux PM.; ''Cross-talk of anosmin-1, the protein implicated in X-linked Kallmann's syndrome, with heparan sulphate and urokinase-type plasminogen activator.''; PubMed Europe PMC Scholia
  60. 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 syndromes through enhancement of FGF binding affinity.''; PubMed Europe PMC Scholia
  61. Haines BP, Wheldon LM, Summerbell D, Heath JK, Rigby PW.; ''Regulated expression of FLRT genes implies a functional role in the regulation of FGF signalling during mouse development.''; PubMed Europe PMC Scholia
  62. Lao DH, Chandramouli S, Yusoff P, Fong CW, Saw TY, Tai LP, Yu CY, Leong HF, Guy GR.; ''A Src homology 3-binding sequence on the C terminus of Sprouty2 is necessary for inhibition of the Ras/ERK pathway downstream of fibroblast growth factor receptor stimulation.''; PubMed Europe PMC Scholia
  63. Roberts PJ, Der CJ.; ''Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer.''; PubMed Europe PMC Scholia
  64. Xiao S, McCarthy JG, Aster JC, Fletcher JA.; ''ZNF198-FGFR1 transforming activity depends on a novel proline-rich ZNF198 oligomerization domain.''; PubMed Europe PMC Scholia
  65. Missiaglia E, Selfe J, Hamdi M, Williamson D, Schaaf G, Fang C, Koster J, Summersgill B, Messahel B, Versteeg R, Pritchard-Jones K, Kool M, Shipley J.; ''Genomic imbalances in rhabdomyosarcoma cell lines affect expression of genes frequently altered in primary tumors: an approach to identify candidate genes involved in tumor development.''; PubMed Europe PMC Scholia
  66. Cha JY, Lambert QT, Reuther GW, Der CJ.; ''Involvement of fibroblast growth factor receptor 2 isoform switching in mammary oncogenesis.''; PubMed Europe PMC Scholia
  67. McKay MM, Morrison DK.; ''Integrating signals from RTKs to ERK/MAPK.''; PubMed Europe PMC Scholia
  68. Roumiantsev S, Krause DS, Neumann CA, Dimitri CA, Asiedu F, Cross NC, Van Etten RA.; ''Distinct stem cell myeloproliferative/T lymphoma syndromes induced by ZNF198-FGFR1 and BCR-FGFR1 fusion genes from 8p11 translocations.''; PubMed Europe PMC Scholia
  69. Knights V, Cook SJ.; ''De-regulated FGF receptors as therapeutic targets in cancer.''; PubMed Europe PMC Scholia
  70. Hadari YR, Gotoh N, Kouhara H, Lax I, Schlessinger J.; ''Critical role for the docking-protein FRS2 alpha in FGF receptor-mediated signal transduction pathways.''; PubMed Europe PMC Scholia
  71. Rubin C, Zwang Y, Vaisman N, Ron D, Yarden Y.; ''Phosphorylation of carboxyl-terminal tyrosines modulates the specificity of Sprouty-2 inhibition of different signaling pathways.''; PubMed Europe PMC Scholia
  72. 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.''; PubMed Europe PMC Scholia
  73. Ahmed Z, Schüller AC, Suhling K, Tregidgo C, Ladbury JE.; ''Extracellular point mutations in FGFR2 elicit unexpected changes in intracellular signalling.''; PubMed Europe PMC Scholia
  74. Onishi-Haraikawa Y, Funaki M, Gotoh N, Shibuya M, Inukai K, Katagiri H, Fukushima Y, Anai M, Ogihara T, Sakoda H, Ono H, Kikuchi M, Oka Y, Asano T.; ''Unique phosphorylation mechanism of Gab1 using PI 3-kinase as an adaptor protein.''; PubMed Europe PMC Scholia
  75. Fukumoto T, Kubota Y, Kitanaka A, Yamaoka G, Ohara-Waki F, Imataki O, Ohnishi H, Ishida T, Tanaka T.; ''Gab1 transduces PI3K-mediated erythropoietin signals to the Erk pathway and regulates erythropoietin-dependent proliferation and survival of erythroid cells.''; PubMed Europe PMC Scholia
  76. Wang JK, Gao G, Goldfarb M.; ''Fibroblast growth factor receptors have different signaling and mitogenic potentials.''; PubMed Europe PMC Scholia
  77. Schlessinger J.; ''Common and distinct elements in cellular signaling via EGF and FGF receptors.''; PubMed Europe PMC Scholia
  78. Wong ES, Lim J, Low BC, Chen Q, Guy GR.; ''Evidence for direct interaction between Sprouty and Cbl.''; PubMed Europe PMC Scholia
  79. Dutt A, Ramos AH, Hammerman PS, Mermel C, Cho J, Sharifnia T, Chande A, Tanaka KE, Stransky N, Greulich H, Gray NS, Meyerson M.; ''Inhibitor-sensitive FGFR1 amplification in human non-small cell lung cancer.''; PubMed Europe PMC Scholia
  80. Pitteloud N, Meysing A, Quinton R, Acierno JS, Dwyer AA, Plummer L, Fliers E, Boepple P, Hayes F, Seminara S, Hughes VA, Ma J, Bouloux P, Mohammadi M, Crowley WF.; ''Mutations in fibroblast growth factor receptor 1 cause Kallmann syndrome with a wide spectrum of reproductive phenotypes.''; PubMed Europe PMC Scholia
  81. 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.''; PubMed Europe PMC Scholia
  82. Raffioni S, Zhu YZ, Bradshaw RA, Thompson LM.; ''Effect of transmembrane and kinase domain mutations on fibroblast growth factor receptor 3 chimera signaling in PC12 cells. A model for the control of receptor tyrosine kinase activation.''; PubMed Europe PMC Scholia
  83. Hart KC, Robertson SC, Kanemitsu MY, Meyer AN, Tynan JA, Donoghue DJ.; ''Transformation and Stat activation by derivatives of FGFR1, FGFR3, and FGFR4.''; PubMed Europe PMC Scholia
  84. Dailey L, Ambrosetti D, Mansukhani A, Basilico C.; ''Mechanisms underlying differential responses to FGF signaling.''; PubMed Europe PMC Scholia
  85. Wheldon LM, Haines BP, Rajappa R, Mason I, Rigby PW, Heath JK.; ''Critical role of FLRT1 phosphorylation in the interdependent regulation of FLRT1 function and FGF receptor signalling.''; PubMed Europe PMC Scholia
  86. di Martino E, L'Hôte CG, Kennedy W, Tomlinson DC, Knowles MA.; ''Mutant fibroblast growth factor receptor 3 induces intracellular signaling and cellular transformation in a cell type- and mutation-specific manner.''; PubMed Europe PMC Scholia
  87. Kan SH, Elanko N, Johnson D, Cornejo-Roldan L, Cook J, Reich EW, Tomkins S, Verloes A, Twigg SR, Rannan-Eliya S, McDonald-McGinn DM, Zackai EH, Wall SA, Muenke M, Wilkie AO.; ''Genomic screening of fibroblast growth-factor receptor 2 reveals a wide spectrum of mutations in patients with syndromic craniosynostosis.''; PubMed Europe PMC Scholia
  88. Wesche J, Haglund K, Haugsten EM.; ''Fibroblast growth factors and their receptors in cancer.''; PubMed Europe PMC Scholia
  89. Zhuang L, Villiger P, Trueb B.; ''Interaction of the receptor FGFRL1 with the negative regulator Spred1.''; PubMed Europe PMC Scholia
  90. 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.''; PubMed Europe PMC Scholia
  91. Zhou W, Feng X, Wu Y, Benge J, Zhang Z, Chen Z.; ''FGF-receptor substrate 2 functions as a molecular sensor integrating external regulatory signals into the FGF pathway.''; PubMed Europe PMC Scholia
  92. Mohammadi M, Dionne CA, Li W, Li N, Spivak T, Honegger AM, Jaye M, Schlessinger J.; ''Point mutation in FGF receptor eliminates phosphatidylinositol hydrolysis without affecting mitogenesis.''; PubMed Europe PMC Scholia
  93. 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.''; PubMed Europe PMC Scholia
  94. Beenken A, Mohammadi M.; ''The FGF family: biology, pathophysiology and therapy.''; PubMed Europe PMC Scholia
  95. 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.''; PubMed Europe PMC Scholia
  96. Cseh B, Doma E, Baccarini M.; ''"RAF" neighborhood: protein-protein interaction in the Raf/Mek/Erk pathway.''; PubMed Europe PMC Scholia
  97. Carpenter G, Ji Q.; ''Phospholipase C-gamma as a signal-transducing element.''; PubMed Europe PMC Scholia
  98. Ornitz DM, Marie PJ.; ''FGF signaling pathways in endochondral and intramembranous bone development and human genetic disease.''; PubMed Europe PMC Scholia
  99. Turner N, Grose R.; ''Fibroblast growth factor signalling: from development to cancer.''; PubMed Europe PMC Scholia
  100. Weiss J, Sos ML, Seidel D, Peifer M, Zander T, Heuckmann JM, Ullrich RT, Menon R, Maier S, Soltermann A, Moch H, Wagener P, Fischer F, Heynck S, Koker M, Schöttle J, Leenders F, Gabler F, Dabow I, Querings S, Heukamp LC, Balke-Want H, Ansén S, Rauh D, Baessmann I, Altmüller J, Wainer Z, Conron M, Wright G, Russell P, Solomon B, Brambilla E, Brambilla C, Lorimier P, Sollberg S, Brustugun OT, Engel-Riedel W, Ludwig C, Petersen I, Sänger J, Clement J, Groen H, Timens W, Sietsma H, Thunnissen E, Smit E, Heideman D, Cappuzzo F, Ligorio C, Damiani S, Hallek M, Beroukhim R, Pao W, Klebl B, Baumann M, Buettner R, Ernestus K, Stoelben E, Wolf J, Nürnberg P, Perner S, Thomas RK.; ''Frequent and focal FGFR1 amplification associates with therapeutically tractable FGFR1 dependency in squamous cell lung cancer.''; PubMed Europe PMC Scholia
  101. Roskoski R.; ''RAF protein-serine/threonine kinases: structure and regulation.''; PubMed Europe PMC Scholia
  102. Klint P, Kanda S, Claesson-Welsh L.; ''Shc and a novel 89-kDa component couple to the Grb2-Sos complex in fibroblast growth factor-2-stimulated cells.''; PubMed Europe PMC Scholia
  103. Cancer Genome Atlas Research Network.; ''Comprehensive genomic characterization defines human glioblastoma genes and core pathways.''; PubMed Europe PMC Scholia
  104. Minegishi Y, Iwanari H, Mochizuki Y, Horii T, Hoshino T, Kodama T, Hamakubo T, Gotoh N.; ''Prominent expression of FRS2beta protein in neural cells and its association with intracellular vesicles.''; PubMed Europe PMC Scholia
  105. Pollock PM, Gartside MG, Dejeza LC, Powell MA, Mallon MA, Davies H, Mohammadi M, Futreal PA, Stratton MR, Trent JM, Goodfellow PJ.; ''Frequent activating FGFR2 mutations in endometrial carcinomas parallel germline mutations associated with craniosynostosis and skeletal dysplasia syndromes.''; PubMed Europe PMC Scholia
  106. Li X, Brunton VG, Burgar HR, Wheldon LM, Heath JK.; ''FRS2-dependent SRC activation is required for fibroblast growth factor receptor-induced phosphorylation of Sprouty and suppression of ERK activity.''; PubMed Europe PMC Scholia
  107. Wong A, Lamothe B, Lee A, Schlessinger J, Lax I.; ''FRS2 alpha attenuates FGF receptor signaling by Grb2-mediated recruitment of the ubiquitin ligase Cbl.''; PubMed Europe PMC Scholia
  108. Mohammadi M, Olsen SK, Ibrahimi OA.; ''Structural basis for fibroblast growth factor receptor activation.''; PubMed Europe PMC Scholia
  109. Rieckmann T, Kotevic I, Trueb B.; ''The cell surface receptor FGFRL1 forms constitutive dimers that promote cell adhesion.''; PubMed Europe PMC Scholia
  110. Hu Y, Guimond SE, Travers P, Cadman S, Hohenester E, Turnbull JE, Kim SH, Bouloux PM.; ''Novel mechanisms of fibroblast growth factor receptor 1 regulation by extracellular matrix protein anosmin-1.''; PubMed Europe PMC Scholia
  111. Lew ED, Furdui CM, Anderson KS, Schlessinger J.; ''The precise sequence of FGF receptor autophosphorylation is kinetically driven and is disrupted by oncogenic mutations.''; PubMed Europe PMC Scholia
  112. Impagnatiello MA, Weitzer S, Gannon G, Compagni A, Cotten M, Christofori G.; ''Mammalian sprouty-1 and -2 are membrane-anchored phosphoprotein inhibitors of growth factor signaling in endothelial cells.''; PubMed Europe PMC Scholia
  113. Singh D, Chan JM, Zoppoli P, Niola F, Sullivan R, Castano A, Liu EM, Reichel J, Porrati P, Pellegatta S, Qiu K, Gao Z, Ceccarelli M, Riccardi R, Brat DJ, Guha A, Aldape K, Golfinos JG, Zagzag D, Mikkelsen T, Finocchiaro G, Lasorella A, Rabadan R, Iavarone A.; ''Transforming fusions of FGFR and TACC genes in human glioblastoma.''; PubMed Europe PMC Scholia
  114. Turner N, Pearson A, Sharpe R, Lambros M, Geyer F, Lopez-Garcia MA, Natrajan R, Marchio C, Iorns E, Mackay A, Gillett C, Grigoriadis A, Tutt A, Reis-Filho JS, Ashworth A.; ''FGFR1 amplification drives endocrine therapy resistance and is a therapeutic target in breast cancer.''; PubMed Europe PMC Scholia
  115. Steinberg F, Zhuang L, Beyeler M, Kälin RE, Mullis PE, Brändli AW, Trueb B.; ''The FGFRL1 receptor is shed from cell membranes, binds fibroblast growth factors (FGFs), and antagonizes FGF signaling in Xenopus embryos.''; PubMed Europe PMC Scholia
  116. Böttcher RT, Pollet N, Delius H, Niehrs C.; ''The transmembrane protein XFLRT3 forms a complex with FGF receptors and promotes FGF signalling.''; PubMed Europe PMC Scholia
  117. Yigzaw Y, Cartin L, Pierre S, Scholich K, Patel TB.; ''The C terminus of sprouty is important for modulation of cellular migration and proliferation.''; PubMed Europe PMC Scholia
  118. Xu H, Lee KW, Goldfarb M.; ''Novel recognition motif on fibroblast growth factor receptor mediates direct association and activation of SNT adapter proteins.''; PubMed Europe PMC Scholia
  119. Wasag B, Lierman E, Meeus P, Cools J, Vandenberghe P.; ''The kinase inhibitor TKI258 is active against the novel CUX1-FGFR1 fusion detected in a patient with T-lymphoblastic leukemia/lymphoma and t(7;8)(q22;p11).''; PubMed Europe PMC Scholia
  120. Ong SH, Lim YP, Low BC, Guy GR.; ''SHP2 associates directly with tyrosine phosphorylated p90 (SNT) protein in FGF-stimulated cells.''; PubMed Europe PMC Scholia
  121. Kyriakis JM, Avruch J.; ''Mammalian MAPK signal transduction pathways activated by stress and inflammation: a 10-year update.''; PubMed Europe PMC Scholia
  122. Kouhara H, Hadari YR, Spivak-Kroizman T, Schilling J, Bar-Sagi D, Lax I, Schlessinger J.; ''A lipid-anchored Grb2-binding protein that links FGF-receptor activation to the Ras/MAPK signaling pathway.''; PubMed Europe PMC Scholia
  123. Mohammadi M, Dikic I, Sorokin A, Burgess WH, Jaye M, Schlessinger J.; ''Identification of six novel autophosphorylation sites on fibroblast growth factor receptor 1 and elucidation of their importance in receptor activation and signal transduction.''; PubMed Europe PMC Scholia
  124. Lim J, Wong ES, Ong SH, Yusoff P, Low BC, Guy GR.; ''Sprouty proteins are targeted to membrane ruffles upon growth factor receptor tyrosine kinase activation. Identification of a novel translocation domain.''; PubMed Europe PMC Scholia
  125. Chen J, Lee BH, Williams IR, Kutok JL, Mitsiades CS, Duclos N, Cohen S, Adelsperger J, Okabe R, Coburn A, Moore S, Huntly BJ, Fabbro D, Anderson KC, Griffin JD, Gilliland DG.; ''FGFR3 as a therapeutic target of the small molecule inhibitor PKC412 in hematopoietic malignancies.''; PubMed Europe PMC Scholia
  126. 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.''; PubMed Europe PMC Scholia
  127. Gu TL, Goss VL, Reeves C, Popova L, Nardone J, Macneill J, Walters DK, Wang Y, Rush J, Comb MJ, Druker BJ, Polakiewicz RD.; ''Phosphotyrosine profiling identifies the KG-1 cell line as a model for the study of FGFR1 fusions in acute myeloid leukemia.''; PubMed Europe PMC Scholia
  128. Hanafusa H, Torii S, Yasunaga T, Matsumoto K, Nishida E.; ''Shp2, an SH2-containing protein-tyrosine phosphatase, positively regulates receptor tyrosine kinase signaling by dephosphorylating and inactivating the inhibitor Sprouty.''; PubMed Europe PMC Scholia
  129. Plotnikov A, Zehorai E, Procaccia S, Seger R.; ''The MAPK cascades: signaling components, nuclear roles and mechanisms of nuclear translocation.''; PubMed Europe PMC Scholia
  130. 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.''; PubMed Europe PMC Scholia
  131. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake H, Gusterson BA, Cooper C, Shipley J, Hargrave D, Pritchard-Jones K, Maitland N, Chenevix-Trench G, Riggins GJ, Bigner DD, Palmieri G, Cossu A, Flanagan A, Nicholson A, Ho JW, Leung SY, Yuen ST, Weber BL, Seigler HF, Darrow TL, Paterson H, Marais R, Marshall CJ, Wooster R, Stratton MR, Futreal PA.; ''Mutations of the BRAF gene in human cancer.''; PubMed Europe PMC Scholia
  132. 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).''; PubMed Europe PMC Scholia
  133. Brady SC, Coleman ML, Munro J, Feller SM, Morrice NA, Olson MF.; ''Sprouty2 association with B-Raf is regulated by phosphorylation and kinase conformation.''; PubMed Europe PMC Scholia
  134. 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.''; PubMed Europe PMC Scholia
  135. Ruhe JE, Streit S, Hart S, Wong CH, Specht K, Knyazev P, Knyazeva T, Tay LS, Loo HL, Foo P, Wong W, Pok S, Lim SJ, Ong H, Luo M, Ho HK, Peng K, Lee TC, Bezler M, Mann C, Gaertner S, Hoefler H, Iacobelli S, Peter S, Tay A, Brenner S, Venkatesh B, Ullrich A.; ''Genetic alterations in the tyrosine kinase transcriptome of human cancer cell lines.''; PubMed Europe PMC Scholia
  136. Gerber SD, Amann R, Wyder S, Trueb B.; ''Comparison of the gene expression profiles from normal and Fgfrl1 deficient mouse kidneys reveals downstream targets of Fgfrl1 signaling.''; PubMed Europe PMC Scholia
  137. Smit L, de Vries-Smits AM, Bos JL, Borst J.; ''B cell antigen receptor stimulation induces formation of a Shc-Grb2 complex containing multiple tyrosine-phosphorylated proteins.''; PubMed Europe PMC Scholia
  138. Mohammadi M, Honegger AM, Rotin D, Fischer R, Bellot F, Li W, Dionne CA, Jaye M, Rubinstein M, Schlessinger J.; ''A tyrosine-phosphorylated carboxy-terminal peptide of the fibroblast growth factor receptor (Flg) is a binding site for the SH2 domain of phospholipase C-gamma 1.''; PubMed Europe PMC Scholia
  139. Parker BC, Engels M, Annala M, Zhang W.; ''Emergence of FGFR family gene fusions as therapeutic targets in a wide spectrum of solid tumours.''; PubMed Europe PMC Scholia
  140. Wellbrock C, Karasarides M, Marais R.; ''The RAF proteins take centre stage.''; PubMed Europe PMC Scholia
  141. Jackson CC, Medeiros LJ, Miranda RN.; ''8p11 myeloproliferative syndrome: a review.''; PubMed Europe PMC Scholia
  142. Cantwell-Dorris ER, O'Leary JJ, Sheils OM.; ''BRAFV600E: implications for carcinogenesis and molecular therapy.''; PubMed Europe PMC Scholia
  143. Ong SH, Hadari YR, Gotoh N, Guy GR, Schlessinger J, Lax I.; ''Stimulation of phosphatidylinositol 3-kinase by fibroblast growth factor receptors is mediated by coordinated recruitment of multiple docking proteins.''; PubMed Europe PMC Scholia
  144. Dance M, Montagner A, Salles JP, Yart A, Raynal P.; ''The molecular functions of Shp2 in the Ras/Mitogen-activated protein kinase (ERK1/2) pathway.''; PubMed Europe PMC Scholia
  145. Turjanski AG, Vaqué JP, Gutkind JS.; ''MAP kinases and the control of nuclear events.''; PubMed Europe PMC Scholia
  146. Furdui CM, Lew ED, Schlessinger J, Anderson KS.; ''Autophosphorylation of FGFR1 kinase is mediated by a sequential and precisely ordered reaction.''; PubMed Europe PMC Scholia
  147. Curto M, Frankel P, Carrero A, Foster DA.; ''Novel recruitment of Shc, Grb2, and Sos by fibroblast growth factor receptor-1 in v-Src-transformed cells.''; PubMed Europe PMC Scholia
  148. Gorringe KL, Jacobs S, Thompson ER, Sridhar A, Qiu W, Choong DY, Campbell IG.; ''High-resolution single nucleotide polymorphism array analysis of epithelial ovarian cancer reveals numerous microdeletions and amplifications.''; PubMed Europe PMC Scholia
  149. Patterson RL, van Rossum DB, Nikolaidis N, Gill DL, Snyder SH.; ''Phospholipase C-gamma: diverse roles in receptor-mediated calcium signaling.''; PubMed Europe PMC Scholia
  150. Lemmon MA, Schlessinger J.; ''Cell signaling by receptor tyrosine kinases.''; PubMed Europe PMC Scholia
  151. Fong CW, Leong HF, Wong ES, Lim J, Yusoff P, Guy GR.; ''Tyrosine phosphorylation of Sprouty2 enhances its interaction with c-Cbl and is crucial for its function.''; PubMed Europe PMC Scholia
  152. Oliveira LM, Seminara SB, Beranova M, Hayes FJ, Valkenburgh SB, Schipani E, Costa EM, Latronico AC, Crowley WF, Vallejo M.; ''The importance of autosomal genes in Kallmann syndrome: genotype-phenotype correlations and neuroendocrine characteristics.''; PubMed Europe PMC Scholia
  153. Kanai M, Göke M, Tsunekawa S, Podolsky DK.; ''Signal transduction pathway of human fibroblast growth factor receptor 3. Identification of a novel 66-kDa phosphoprotein.''; PubMed Europe PMC Scholia
  154. Amann R, Trueb B.; ''Evidence that the novel receptor FGFRL1 signals indirectly via FGFR1.''; PubMed Europe PMC Scholia
  155. Ibrahimi OA, Zhang F, Eliseenkova AV, Itoh N, Linhardt RJ, Mohammadi M.; ''Biochemical analysis of pathogenic ligand-dependent FGFR2 mutations suggests distinct pathophysiological mechanisms for craniofacial and limb abnormalities.''; PubMed Europe PMC Scholia
  156. Lax I, Wong A, Lamothe B, Lee A, Frost A, Hawes J, Schlessinger J.; ''The docking protein FRS2alpha controls a MAP kinase-mediated negative feedback mechanism for signaling by FGF receptors.''; PubMed Europe PMC Scholia
  157. Hadari YR, Kouhara H, Lax I, Schlessinger J.; ''Binding of Shp2 tyrosine phosphatase to FRS2 is essential for fibroblast growth factor-induced PC12 cell differentiation.''; PubMed Europe PMC Scholia
  158. 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).''; PubMed Europe PMC Scholia
  159. Heath C, Cross NC.; ''Critical role of STAT5 activation in transformation mediated by ZNF198-FGFR1.''; PubMed Europe PMC Scholia
  160. Wu YM, Su F, Kalyana-Sundaram S, Khazanov N, Ateeq B, Cao X, Lonigro RJ, Vats P, Wang R, Lin SF, Cheng AJ, Kunju LP, Siddiqui J, Tomlins SA, Wyngaard P, Sadis S, Roychowdhury S, Hussain MH, Feng FY, Zalupski MM, Talpaz M, Pienta KJ, Rhodes DR, Robinson DR, Chinnaiyan AM.; ''Identification of targetable FGFR gene fusions in diverse cancers.''; PubMed Europe PMC Scholia
  161. Mason JM, Morrison DJ, Bassit B, Dimri M, Band H, Licht JD, Gross I.; ''Tyrosine phosphorylation of Sprouty proteins regulates their ability to inhibit growth factor signaling: a dual feedback loop.''; PubMed Europe PMC Scholia
  162. 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.''; PubMed Europe PMC Scholia
  163. Agazie YM, Movilla N, Ischenko I, Hayman MJ.; ''The phosphotyrosine phosphatase SHP2 is a critical mediator of transformation induced by the oncogenic fibroblast growth factor receptor 3.''; PubMed Europe PMC Scholia
  164. Sattler M, Mohi MG, Pride YB, Quinnan LR, Malouf NA, Podar K, Gesbert F, Iwasaki H, Li S, Van Etten RA, Gu H, Griffin JD, Neel BG.; ''Critical role for Gab2 in transformation by BCR/ABL.''; PubMed Europe PMC Scholia
  165. Yusoff P, Lao DH, Ong SH, Wong ES, Lim J, Lo TL, Leong HF, Fong CW, Guy GR.; ''Sprouty2 inhibits the Ras/MAP kinase pathway by inhibiting the activation of Raf.''; PubMed Europe PMC Scholia
  166. 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, Schmitt FC, Palacios J, Lakhani SR, Ashworth A.; ''FGFR1 emerges as a potential therapeutic target for lobular breast carcinomas.''; PubMed Europe PMC Scholia
  167. Hatch NE, Hudson M, Seto ML, Cunningham ML, Bothwell M.; ''Intracellular retention, degradation, and signaling of glycosylation-deficient FGFR2 and craniosynostosis syndrome-associated FGFR2C278F.''; PubMed Europe PMC Scholia

History

View all...
CompareRevisionActionTimeUserComment
114733view16:21, 25 January 2021ReactomeTeamReactome version 75
113177view11:24, 2 November 2020ReactomeTeamReactome version 74
112405view15:34, 9 October 2020ReactomeTeamReactome version 73
101309view11:20, 1 November 2018ReactomeTeamreactome version 66
100846view20:51, 31 October 2018ReactomeTeamreactome version 65
100387view19:25, 31 October 2018ReactomeTeamreactome version 64
99934view16:09, 31 October 2018ReactomeTeamreactome version 63
99489view14:42, 31 October 2018ReactomeTeamreactome version 62 (2nd attempt)
99141view12:40, 31 October 2018ReactomeTeamreactome version 62
94044view13:53, 16 August 2017ReactomeTeamreactome version 61
93669view11:30, 9 August 2017ReactomeTeamreactome version 61
87126view18:45, 18 July 2016EgonwOntology Term : 'signaling pathway' added !
86792view09:26, 11 July 2016ReactomeTeamreactome version 56
83306view10:43, 18 November 2015ReactomeTeamVersion54
81443view12:58, 21 August 2015ReactomeTeamNew pathway

External references

DataNodes

View all...
NameTypeDatabase referenceComment
ADPMetaboliteCHEBI:16761 (ChEBI)
ATPMetaboliteCHEBI:15422 (ChEBI)
AZ 2171 MetaboliteCHEBI:556867 (ChEBI) A broad specificity ATP-competitive inhibitor of FGF-, VEGF-, PDGF- and KIT receptors that is in Phase I and II clinical trials for treatment of gastric, breast and endometrial cancers.
AZD4547 MetaboliteCHEBI:63453 (ChEBI) AZD4547 (Astra Zeneca) is a pan-FGFR inhibitor in Phase I clinical trials for patients with advanced gastric cancer (NCT01457846) and for patient with advanced solid tumors with or without amplified FGFR1 or 2 (NCT00979134) and in Phase I/II trials for breast cancer patients with FGFR1 amplifications (NCT01202591).
Activated FGFR1:p-8T-FRS2ComplexR-HSA-5654260 (Reactome)
Activated FGFR1:p-FRS2:GRB2:GAB1:PI3KComplexR-HSA-5654205 (Reactome)
Activated FGFR1:p-FRS2:GRB2:GAB1:PIK3R1ComplexR-HSA-5654186 (Reactome)
Activated FGFR1:p-FRS2:GRB2:SOS1ComplexR-HSA-5654264 (Reactome)
Activated FGFR1:p-FRS2:p-PTPN11:GRB2:GAB1:PI3KComplexR-HSA-5654188 (Reactome)
Activated FGFR1:p-FRS2:p-PTPN11:p-CBL:GRB2ComplexR-HSA-5654267 (Reactome)
Activated FGFR1:p-FRS2:p-PTPN11ComplexR-HSA-5654204 (Reactome)
Activated FGFR1:p-FRS2ComplexR-HSA-5654200 (Reactome)
Activated FGFR1:p-FRS3ComplexR-HSA-5654261 (Reactome)
Activated FGFR1:p-FRS:PTPN11ComplexR-HSA-5654268 (Reactome)
Activated FGFR1:p-FRS:p-PTPN11ComplexR-HSA-5654271 (Reactome)
Activated FGFR1:p-FRSComplexR-HSA-5654262 (Reactome)
Activated FGFR1:p-SHC1:GRB2:SOS1ComplexR-HSA-5654275 (Reactome)
Activated FGFR1:p-SHC1ComplexR-HSA-5654273 (Reactome)
Activated FGFR:p-FRS2:p-PTPN11:GRB2:GAB1:PIK3R1ComplexR-HSA-5654185 (Reactome)
Activated

overexpressed

FGFR1b homodimer
ComplexR-HSA-1982038 (Reactome)
Activated

overexpressed

FGFR1c homodimer
ComplexR-HSA-1982036 (Reactome)
Activated FGFR1 P252X mutantsComplexR-HSA-2050642 (Reactome)
Activated FGFR1

mutant dimers with enhanced kinase

activity
ComplexR-HSA-2023432 (Reactome)
Activated FGFR1:FLRTComplexR-HSA-5656063 (Reactome)
Activated FGFR1:FRS2ComplexR-HSA-5654176 (Reactome)
Activated FGFR1:FRS3ComplexR-HSA-5654255 (Reactome)
Activated FGFR1:SHC1ComplexR-HSA-5654258 (Reactome)
Activated FGFR1ComplexR-HSA-5654170 (Reactome)
Activated FGFR1b homodimerComplexR-HSA-190430 (Reactome)
Activated FGFR1c

bound to

FGF23:Klotho
ComplexR-HSA-500333 (Reactome)
Activated FGFR1c homodimerComplexR-HSA-190425 (Reactome)
BAG4(1-126):FGFR1(208-822) fusion ProteinO95429 (Uniprot-TrEMBL)
BAG4(1-126):p-8Y FGFR1(208-822) fusion ProteinO95429 (Uniprot-TrEMBL)
BCR-FGFR1 fusion ProteinP11274 (Uniprot-TrEMBL)
BCR-p-FGFR1 fusion mutant dimerComplexR-HSA-1838980 (Reactome)
BCR-p-FGFR1 fusion ProteinP11274 (Uniprot-TrEMBL)
BGJ398 MetaboliteCHEBI:63451 (ChEBI) A pan-FGFR ATP-competitive inhibitor that is in phase I clinical trials for advanced solid malignancies with amplification or activation of FGFR1 and 2 or activation of FGFR3 (NCT01004224).
BRAF ProteinP15056 (Uniprot-TrEMBL)
BRAFProteinP15056 (Uniprot-TrEMBL)
Brivanib MetaboliteCHEBI:443041 (ChEBI) Brivanib, from Bristol Myers Squibb, is an FGFR and VEGFR inhibitor that has anti-angiogenic effects and that is in clinical trials for endometrial cancer (NCT00888173) (Huynh, 2008). No specific data pertaining to the activity of Brivanib against activated or amplified FGFRs is available, however.
Brivanib alaninate MetaboliteCHEBI:270995 (ChEBI) Brivanib alaninate is the pro-drug of brivanib, a VEGFR- and FGFR-tyrosine kinase inhibitor that is in Phase I clinical trials for metastatic solid tumors and in patients with endometrial cancer (NCT0088173). No specific data pertaining to the activity of Brivanib against activated or amplified FGFRs is available, however.
CBL ProteinP22681 (Uniprot-TrEMBL)
CBLProteinP22681 (Uniprot-TrEMBL)
CNTRL-FGFR1 fusion ProteinQ7Z7A1-3 (Uniprot-TrEMBL)
CNTRL-p-2Y-FGFR1 fusion ProteinQ7Z7A1-3 (Uniprot-TrEMBL)
CPSF6-FGFR1 fusion ProteinQ16630 (Uniprot-TrEMBL)
CPSF6-p-2Y-FGFR1 fusion ProteinQ16630 (Uniprot-TrEMBL)
CUX1-FGFR1 fusion ProteinP39880 (Uniprot-TrEMBL)
CUX1-p-2Y-FGFR1 fusion ProteinP39880 (Uniprot-TrEMBL)
DAG and IP3 signalingPathwayR-HSA-1489509 (Reactome) This pathway describes the generation of DAG and IP3 by the PLCgamma-mediated hydrolysis of PIP2 and the subsequent downstream signaling events.
Dovitinib MetaboliteCHEBI:594834 (ChEBI) A Novartis tyrosine kinase inhibitor with activity against multiple tyrosine kinase receptors including FGFRs, VEGFRs, PDGFRs, KIT, FLT3 and CSFR. TKI258 is in Phase II clinical trials for advanced breast cancer in patients with and without FGFR1 amplification (NCT00958971), for endometrial cancer with WT or activated FGFR2 mutants (NCT01379534), for relapsed myeloma with and without the t4:14 FGFR3 translocation/amplification (NCT01058434), and in bladder cancer in cases where archived material is available to check for correlation with FGFR3 mutation status (NCT00790426).
E3810 MetaboliteCHEBI:65138 (ChEBI) E-3810 is a dual VEGFR and FGFR inhibitor that has anti-angiogenic and anti-tumorigenic effects in preclinical studies (Bello, 2011). It is in Phase I clinical trials for patients with solid tumors (NCT01283945).
ERLIN2(1-185):FGFR1(c.-88-822) fusion ProteinO94905 (Uniprot-TrEMBL)
ERLIN2(1-185):p-8Y FGFR1(c.-88-822) fusion ProteinO94905 (Uniprot-TrEMBL)
FGF1 ProteinP05230 (Uniprot-TrEMBL)
FGF10 ProteinO15520 (Uniprot-TrEMBL)
FGF17-1 ProteinO60258-1 (Uniprot-TrEMBL)
FGF18 ProteinO76093 (Uniprot-TrEMBL)
FGF2(10-155) ProteinP09038 (Uniprot-TrEMBL)
FGF20 ProteinQ9NP95 (Uniprot-TrEMBL)
FGF22 ProteinQ9HCT0 (Uniprot-TrEMBL)
FGF23 bound to Klotho and FGFR1cComplexR-HSA-190218 (Reactome)
FGF23(25-251) ProteinQ9GZV9 (Uniprot-TrEMBL)
FGF3 ProteinP11487 (Uniprot-TrEMBL)
FGF4 ProteinP08620 (Uniprot-TrEMBL)
FGF5-1 ProteinP12034-1 (Uniprot-TrEMBL)
FGF6 ProteinP10767 (Uniprot-TrEMBL)
FGF8-1 ProteinP55075-1 (Uniprot-TrEMBL)
FGF9 ProteinP31371 (Uniprot-TrEMBL)
FGFR1 K656E ProteinP11362 (Uniprot-TrEMBL)
FGFR1 N546K ProteinP11362 (Uniprot-TrEMBL) weak constitutive activation of ligand independent binding based on analogy of FGFR3 N540 mutation in hypochondroplasia (Rand 2005)
FGFR1 P252S ProteinP11362 (Uniprot-TrEMBL) thought to increase # hydrogen bonds, increase ligand affinity and ligand binding range (Ruhe 2007)
FGFR1 P252T ProteinP11362 (Uniprot-TrEMBL) thought to increase # hydrogen bonds, increase ligand affinity and ligand binding range (Ruhe 2007)
FGFR1 P252X mutant

dimers bound to

FGFs
ComplexR-HSA-2050638 (Reactome)
FGFR1 P252X mutantsComplexR-HSA-2012029 (Reactome)
FGFR1 R576W ProteinP11362 (Uniprot-TrEMBL)
FGFR1 fusion mutant dimers:TKIsComplexR-HSA-1839036 (Reactome)
FGFR1 mutant dimers

with enhanced

kinase activity
ComplexR-HSA-2023433 (Reactome)
FGFR1 mutants with

enhanced kinase

activity
ComplexR-HSA-2012030 (Reactome)
FGFR1(22-763):TACC1(571-805) fusion ProteinP11362 (Uniprot-TrEMBL)
FGFR1OP-FGFR1 fusion ProteinO95684 (Uniprot-TrEMBL)
FGFR1OP-p-FGFR1 fusion mutant dimerComplexR-HSA-1838997 (Reactome)
FGFR1OP-p-FGFR1 fusion ProteinO95684 (Uniprot-TrEMBL)
FGFR1OP2-FGFR1 fusion ProteinQ9NVK5 (Uniprot-TrEMBL)
FGFR1OP2-p-2Y-FGFR1 fusion ProteinQ9NVK5 (Uniprot-TrEMBL)
FGFR1b ProteinP11362-19 (Uniprot-TrEMBL) While the existence of a "b" isoform of fibroblast growth factor receptor 1 is well established and its biochemical and functional properties have been extensively characterized (e.g., Mohammadi et al. 2005; Zhang et al. 2006), its amino acid sequence is not represented in reference protein sequence databases, except as the 47-residue polypeptide (deposited in GenBank as accession AAB19502) first used by Johnson et al. (1991) to distinguish the "b" and "c" isoforms of the receptor.
FGFR1b homodimer bound to FGFComplexR-HSA-190226 (Reactome)
FGFR1b homodimerComplexR-HSA-190228 (Reactome)
FGFR1b-binding FGFsComplexR-HSA-189963 (Reactome)
FGFR1bProteinP11362-19 (Uniprot-TrEMBL) While the existence of a "b" isoform of fibroblast growth factor receptor 1 is well established and its biochemical and functional properties have been extensively characterized (e.g., Mohammadi et al. 2005; Zhang et al. 2006), its amino acid sequence is not represented in reference protein sequence databases, except as the 47-residue polypeptide (deposited in GenBank as accession AAB19502) first used by Johnson et al. (1991) to distinguish the "b" and "c" isoforms of the receptor.
FGFR1c ProteinP11362-1 (Uniprot-TrEMBL)
FGFR1c P252R ProteinP11362-1 (Uniprot-TrEMBL)
FGFR1c homodimer bound to FGFComplexR-HSA-190233 (Reactome)
FGFR1c homodimerComplexR-HSA-190222 (Reactome)
FGFR1c-binding FGFsComplexR-HSA-189953 (Reactome)
FGFR1c:KAL1ComplexR-HSA-5654346 (Reactome)
FGFR1cProteinP11362-1 (Uniprot-TrEMBL)
FGFRL1

dimer:SPRED1/2

dimer
ComplexR-HSA-5654350 (Reactome)
FGFRL1 ProteinQ8N441 (Uniprot-TrEMBL)
FGFRL1 dimerComplexR-HSA-5654348 (Reactome)
FGFRL1-binding FGFs:FGFRL1 dimerComplexR-HSA-5654354 (Reactome)
FGFRL1-binding FGFsComplexR-HSA-5654351 (Reactome)
FLRT1 ProteinQ9NZU1 (Uniprot-TrEMBL)
FLRT1,2,3ComplexR-HSA-5656056 (Reactome)
FLRT2 ProteinO43155 (Uniprot-TrEMBL)
FLRT3 ProteinQ9NZU0 (Uniprot-TrEMBL)
FRS2 ProteinQ8WU20 (Uniprot-TrEMBL)
FRS2ProteinQ8WU20 (Uniprot-TrEMBL)
FRS3 ProteinO43559 (Uniprot-TrEMBL)
FRS3ProteinO43559 (Uniprot-TrEMBL)
GAB1 ProteinQ13480 (Uniprot-TrEMBL)
GAB2 ProteinQ9UQC2 (Uniprot-TrEMBL)
GDP MetaboliteCHEBI:17552 (ChEBI)
GDPMetaboliteCHEBI:17552 (ChEBI)
GRB2-1 ProteinP62993-1 (Uniprot-TrEMBL)
GRB2-1:SOS1ComplexR-HSA-109797 (Reactome)
GRB2-1ProteinP62993-1 (Uniprot-TrEMBL)
GRB2:GAB1:PIK3R1ComplexR-HSA-179864 (Reactome)
GRB2:GAB1ComplexR-HSA-179849 (Reactome)
GRB2:GAB2ComplexR-HSA-912522 (Reactome)
GTP MetaboliteCHEBI:15996 (ChEBI)
GTPMetaboliteCHEBI:15996 (ChEBI)
GalNAc-T178-FGF23(25-251) ProteinQ9GZV9 (Uniprot-TrEMBL)
HRAS ProteinP01112 (Uniprot-TrEMBL)
HS MetaboliteCHEBI:28815 (ChEBI)
HSMetaboliteCHEBI:28815 (ChEBI)
KAL1 ProteinP23352 (Uniprot-TrEMBL)
KAL1:HSComplexR-HSA-5654355 (Reactome)
KAL1ProteinP23352 (Uniprot-TrEMBL)
KL-1 ProteinQ9UEF7-1 (Uniprot-TrEMBL)
KL-2 ProteinQ9UEF7-2 (Uniprot-TrEMBL)
KRAS ProteinP01116 (Uniprot-TrEMBL)
Klotho bound to FGF23ComplexR-HSA-190208 (Reactome)
LRRFIP1-FGFR1 fusion ProteinQ32MZ4 (Uniprot-TrEMBL)
LRRFIP1-p-2Y-FGFR1 fusion ProteinQ32MZ4 (Uniprot-TrEMBL)
MYO18A-FGFR1 fusion ProteinQ92614 (Uniprot-TrEMBL)
MYO18A-p-2Y-FGFR1 fusion ProteinQ92614 (Uniprot-TrEMBL)
Midostaurin MetaboliteCHEBI:63452 (ChEBI) PKC412 is a multi-tyrosine kinase inhibitor that has been shown to be active against FGFR1 fusion proteins (Wasag, 2011; Chen,2004) and against multiple myeloma (Chen, 2005).
NRAS ProteinP01111 (Uniprot-TrEMBL)
Overexpressed FGFR1:TKIsComplexR-HSA-2023442 (Reactome)
Overexpressed FGFR1 homodimersComplexR-HSA-1982053 (Reactome)
PD173074 MetaboliteCHEBI:63448 (ChEBI) PD173074 is potent pan-FGFR reversible inhibitor that interacts with residues in the ATP-binding pocket and inhibits tyrosine kinase activity and autophosphorylation (Mohammadi, 1998; Ezzat, 2005). PD173074 is not suitable for therapeutic use due to issues with toxicity.
PI(3,4,5)P3 MetaboliteCHEBI:16618 (ChEBI)
PI(3,4,5)P3MetaboliteCHEBI:16618 (ChEBI)
PI(4,5)P2MetaboliteCHEBI:18348 (ChEBI)
PIK3CA ProteinP42336 (Uniprot-TrEMBL)
PIK3CAProteinP42336 (Uniprot-TrEMBL)
PIK3R1 ProteinP27986 (Uniprot-TrEMBL)
PIK3R1ProteinP27986 (Uniprot-TrEMBL)
PIP3 activates AKT signalingPathwayR-HSA-1257604 (Reactome) Signaling by AKT is one of the key outcomes of receptor tyrosine kinase (RTK) activation. AKT is activated by the cellular second messenger PIP3, a phospholipid that is generated by PI3K. In ustimulated cells, PI3K class IA enzymes reside in the cytosol as inactive heterodimers composed of p85 regulatory subunit and p110 catalytic subunit. In this complex, p85 stabilizes p110 while inhibiting its catalytic activity. Upon binding of extracellular ligands to RTKs, receptors dimerize and undergo autophosphorylation. The regulatory subunit of PI3K, p85, is recruited to phosphorylated cytosolic RTK domains either directly or indirectly, through adaptor proteins, leading to a conformational change in the PI3K IA heterodimer that relieves inhibition of the p110 catalytic subunit. Activated PI3K IA phosphorylates PIP2, converting it to PIP3; this reaction is negatively regulated by PTEN phosphatase. PIP3 recruits AKT to the plasma membrane, allowing TORC2 to phosphorylate a conserved serine residue of AKT. Phosphorylation of this serine induces a conformation change in AKT, exposing a conserved threonine residue that is then phosphorylated by PDPK1 (PDK1). Phosphorylation of both the threonine and the serine residue is required to fully activate AKT. The active AKT then dissociates from PIP3 and phosphorylates a number of cytosolic and nuclear proteins that play important roles in cell survival and metabolism. For a recent review of AKT signaling, please refer to Manning and Cantley, 2007.
PLCG1 ProteinP19174 (Uniprot-TrEMBL)
PLCG1ProteinP19174 (Uniprot-TrEMBL)
PP2A (A:C)ComplexR-HSA-934544 (Reactome)
PP2A(A:C):S112/S121-pSPRY2ComplexR-HSA-934578 (Reactome)
PP2A(A:C):SPRY2ComplexR-HSA-934550 (Reactome)
PP2A(A:C):Y55/Y227-pSPRY2ComplexR-HSA-934598 (Reactome)
PPA2A

(A:C):S112/S115

p-SPRY2
ComplexR-HSA-1295605 (Reactome)
PPA2A (A:C):Y55/Y227 p-SPRY2:GRB2ComplexR-HSA-1295625 (Reactome)
PPA2A(A:C):SPRY2ComplexR-HSA-1295593 (Reactome)
PPP2CA ProteinP67775 (Uniprot-TrEMBL)
PPP2CB ProteinP62714 (Uniprot-TrEMBL)
PPP2R1A ProteinP30153 (Uniprot-TrEMBL)
PTPN11 ProteinQ06124 (Uniprot-TrEMBL)
PTPN11ProteinQ06124 (Uniprot-TrEMBL)
PiMetaboliteCHEBI:18367 (ChEBI)
Plasma membrane FGFR1 fusion dimersComplexR-HSA-8853271 (Reactome)
Plasma membrane p-Y FGFR1 fusion dimersComplexR-HSA-8853273 (Reactome)
RAF/MAP kinase cascadePathwayR-HSA-5673001 (Reactome) The RAS-RAF-MEK-ERK pathway regulates processes such as proliferation, differentiation, survival, senescence and cell motility in response to growth factors, hormones and cytokines, among others. Binding of these stimuli to receptors in the plasma membrane promotes the GEF-mediated activation of RAS at the plasma membrane and initiates the three-tiered kinase cascade of the conventional MAPK cascades. GTP-bound RAS recruits RAF (the MAPK kinase kinase), and promotes its dimerization and activation (reviewed in Cseh et al, 2014; Roskoski, 2010; McKay and Morrison, 2007; Wellbrock et al, 2004). Activated RAF phosphorylates the MAPK kinase proteins MEK1 and MEK2 (also known as MAP2K1 and MAP2K2), which in turn phophorylate the proline-directed kinases ERK1 and 2 (also known as MAPK3 and MAPK1) (reviewed in Roskoski, 2012a, b; Kryiakis and Avruch, 2012). Activated ERK proteins may undergo dimerization and have identified targets in both the nucleus and the cytosol; consistent with this, a proportion of activated ERK protein relocalizes to the nucleus in response to stimuli (reviewed in Roskoski 2012b; Turjanski et al, 2007; Plotnikov et al, 2010; Cargnello et al, 2011). Although initially seen as a linear cascade originating at the plasma membrane and culminating in the nucleus, the RAS/RAF MAPK cascade is now also known to be activated from various intracellular location. Temporal and spatial specificity of the cascade is achieved in part through the interaction of pathway components with numerous scaffolding proteins (reviewed in McKay and Morrison, 2007; Brown and Sacks, 2009).
The importance of the RAS/RAF MAPK cascade is highlighted by the fact that components of this pathway are mutated with high frequency in a large number of human cancers. Activating mutations in RAS are found in approximately one third of human cancers, while ~8% of tumors express an activated form of BRAF (Roberts and Der, 2007; Davies et al, 2002; Cantwell-Dorris et al, 2011).
RPS27A(1-76) ProteinP62979 (Uniprot-TrEMBL)
S111/S120 p-SPRY2:B-RAFComplexR-HSA-1295587 (Reactome)
SHC1 p46,p52ComplexR-HSA-1169480 (Reactome) SHC1 isoforms p46 and p52 are found in B cells (Smit et al. 1994).
SHC1-2 ProteinP29353-2 (Uniprot-TrEMBL)
SHC1-3 ProteinP29353-3 (Uniprot-TrEMBL)
SOS1 ProteinQ07889 (Uniprot-TrEMBL)
SPRED1 ProteinQ7Z699 (Uniprot-TrEMBL)
SPRED1/2 dimerComplexR-HSA-5654215 (Reactome)
SPRED2 ProteinQ7Z698 (Uniprot-TrEMBL)
SPRY2 ProteinO43597 (Uniprot-TrEMBL)
SPRY2:B-RAFComplexR-HSA-1295598 (Reactome)
SRC-1ProteinP12931-1 (Uniprot-TrEMBL)
STAT1 ProteinP42224 (Uniprot-TrEMBL)
STAT1,3ComplexR-HSA-1888196 (Reactome)
STAT3 ProteinP40763 (Uniprot-TrEMBL)
STAT5A ProteinP42229 (Uniprot-TrEMBL)
STAT5A,STAT5BComplexR-HSA-452094 (Reactome)
STAT5B ProteinP51692 (Uniprot-TrEMBL)
SU5402 MetaboliteCHEBI:63449 (ChEBI) SU5402 is an ATP-competitive FGFR and VEGFR inhibitor that is used as an in vitro reagent. Su5402 is not suitable for therapeutic use due to toxicity issues.
TRIM24-FGFR1 fusion ProteinO15164 (Uniprot-TrEMBL)
TRIM24-p-2Y-FGFR1 fusion ProteinO15164 (Uniprot-TrEMBL)
Tyrosine kinase

inhibitors of

overexpressed FGFR1
ComplexR-ALL-2023441 (Reactome)
Tyrosine kinase

inhibitors of FGFR1

fusion mutants
ComplexR-ALL-2045079 (Reactome)
UBA52(1-76) ProteinP62987 (Uniprot-TrEMBL)
UBB(1-76) ProteinP0CG47 (Uniprot-TrEMBL)
UBB(153-228) ProteinP0CG47 (Uniprot-TrEMBL)
UBB(77-152) ProteinP0CG47 (Uniprot-TrEMBL)
UBC(1-76) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(153-228) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(229-304) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(305-380) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(381-456) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(457-532) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(533-608) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(609-684) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(77-152) ProteinP0CG48 (Uniprot-TrEMBL)
Ub-(Y55/Y227)p-SPRY2ComplexR-HSA-1370875 (Reactome)
Ub-Activated FGFR1 complex:Ub-p-FRS2ComplexR-HSA-5654357 (Reactome)
Ub:Y55/Y227-pSPRY2:CBLComplexR-HSA-934572 (Reactome)
UbComplexR-HSA-113595 (Reactome)
Y55/Y227-pSPRY2:CBLComplexR-HSA-934576 (Reactome)
ZMYM2-FGFR1 fusion ProteinQ9UBW7 (Uniprot-TrEMBL)
ZMYM2-p-2Y-FGFR1 fusion ProteinQ9UBW7 (Uniprot-TrEMBL)
activated FGFR1:PLCG1ComplexR-HSA-5654155 (Reactome)
activated FGFR1:p-4Y-PLCG1ComplexR-HSA-5654154 (Reactome)
cytosolic FGFR1

fusion mutant

dimers
ComplexR-HSA-1839026 (Reactome)
cytosolic FGFR1 fusion mutantsComplexR-HSA-1839029 (Reactome)
cytosolic p-FGFR1

fusion mutant

dimers
ComplexR-HSA-1839020 (Reactome)
cytosolic p-FGFR1

fusion mutant

dimers
ComplexR-HSA-1839023 (Reactome)
p-4Y-PLCG1 ProteinP19174 (Uniprot-TrEMBL)
p-4Y-PLCG1ProteinP19174 (Uniprot-TrEMBL)
p-5Y-FRS3 ProteinO43559 (Uniprot-TrEMBL) The phospho-tyrosine positions for FRS2-beta were inferred by similarity to the analogous positions in FRS2-alpha. Five out of six tyrosine positions in alpha are present in beta.
p-6Y FGFR1(22-763):TACC1(571-805) fusion ProteinP11362 (Uniprot-TrEMBL)
p-6Y-FRS2 ProteinQ8WU20 (Uniprot-TrEMBL)
p-8T-FRS2 ProteinQ8WU20 (Uniprot-TrEMBL)
p-8Y- FGFR1 R576W ProteinP11362 (Uniprot-TrEMBL)
p-8Y-FGFR1 K656E ProteinP11362 (Uniprot-TrEMBL)
p-8Y-FGFR1 N546K ProteinP11362 (Uniprot-TrEMBL)
p-8Y-FGFR1 P252S ProteinP11362 (Uniprot-TrEMBL) thought to increase # hydrogen bonds, increase ligand affinity and ligand binding range (Ruhe 2007)
p-8Y-FGFR1 P252T ProteinP11362 (Uniprot-TrEMBL) thought to increase # hydrogen bonds, increase ligand affinity and ligand binding range (Ruhe 2007)
p-8Y-FGFR1b ProteinP11362-19 (Uniprot-TrEMBL) While the existence of a "b" isoform of fibroblast growth factor receptor 1 is well established and its biochemical and functional properties have been extensively characterized (e.g., Mohammadi et al. 2005; Zhang et al. 2006), its amino acid sequence is not represented in reference protein sequence databases, except as the 47-residue polypeptide (deposited in GenBank as accession AAB19502) first used by Johnson et al. (1991) to distinguish the "b" and "c" isoforms of the receptor.
p-8Y-FGFR1c ProteinP11362-1 (Uniprot-TrEMBL)
p-8Y-FGFR1c P252R ProteinP11362-1 (Uniprot-TrEMBL)
p-FGFR1 fusion

mutant

dimers:PIK3R1
ComplexR-HSA-1839052 (Reactome)
p-FGFR1 mutant fusions:PI3KComplexR-HSA-1839055 (Reactome)
p-S111,S120-SPRY2 ProteinO43597 (Uniprot-TrEMBL)
p-S111,S120-SPRY2ProteinO43597 (Uniprot-TrEMBL)
p-S112,S115-SPRY2 ProteinO43597 (Uniprot-TrEMBL)
p-S112,S121-SPRY2 ProteinO43597 (Uniprot-TrEMBL)
p-STAT5A, p-STAT5BComplexR-HSA-507929 (Reactome)
p-T,Y MAPK dimersComplexR-HSA-1268261 (Reactome)
p-T185,Y187-MAPK1 ProteinP28482 (Uniprot-TrEMBL)
p-T202,Y204-MAPK3 ProteinP27361 (Uniprot-TrEMBL)
p-T250,T255,T385,S437-MKNK1ProteinQ9BUB5 (Uniprot-TrEMBL)
p-Y-GAB2 ProteinQ9UQC2 (Uniprot-TrEMBL)
p-Y177-BCR-pY-FGFR1 mutant:GRB2:p-GAB2:PIK3R1ComplexR-HSA-1839048 (Reactome)
p-Y194,Y195,Y272-SHC1-3 ProteinP29353-3 (Uniprot-TrEMBL)
p-Y239,Y240,Y317-SHC1-2 ProteinP29353-2 (Uniprot-TrEMBL)
p-Y371-CBL ProteinP22681 (Uniprot-TrEMBL)
p-Y371-CBL:GRB2ComplexR-HSA-182964 (Reactome)
p-Y546,Y584-PTPN11 ProteinQ06124 (Uniprot-TrEMBL)
p-Y55,Y227-SPRY2 ProteinO43597 (Uniprot-TrEMBL)
p-Y694-STAT5A ProteinP42229 (Uniprot-TrEMBL)
p-Y699-STAT5B ProteinP51692 (Uniprot-TrEMBL)
p-Y701-STAT1 ProteinP42224 (Uniprot-TrEMBL)
p-Y705-STAT3 ProteinP40763 (Uniprot-TrEMBL)
p21 RAS:GDPComplexR-HSA-109796 (Reactome)
p21 RAS:GTPComplexR-HSA-109783 (Reactome)
pY-STAT1,3ComplexR-HSA-1888197 (Reactome)
pY177-BCR-p-FGFR1 fusion mutant dimerComplexR-HSA-1839043 (Reactome)
pY177-BCR-p-FGFR1 fusion ProteinP11274 (Uniprot-TrEMBL)
pY177-BCR-pY-FGFR1 mutant:GRB2:p-GAB1:PI3KComplexR-HSA-1839051 (Reactome)
pY177-BCR-pY-FGFR1 mutant:GRB2:p-GAB2ComplexR-HSA-1839047 (Reactome)
pY177-BCR1-p-FGFR1 mutant:GRB2:GAB2ComplexR-HSA-1839045 (Reactome)
plasma membrane FGFR1 fusionsComplexR-HSA-8853277 (Reactome)

Annotated Interactions

View all...
SourceTargetTypeDatabase referenceComment
ADPArrowR-HSA-1295609 (Reactome)
ADPArrowR-HSA-1839065 (Reactome)
ADPArrowR-HSA-1839067 (Reactome)
ADPArrowR-HSA-1839091 (Reactome)
ADPArrowR-HSA-1839107 (Reactome)
ADPArrowR-HSA-1839110 (Reactome)
ADPArrowR-HSA-1839112 (Reactome)
ADPArrowR-HSA-1888198 (Reactome)
ADPArrowR-HSA-190427 (Reactome)
ADPArrowR-HSA-190429 (Reactome)
ADPArrowR-HSA-191062 (Reactome)
ADPArrowR-HSA-1982066 (Reactome)
ADPArrowR-HSA-2023455 (Reactome)
ADPArrowR-HSA-2023460 (Reactome)
ADPArrowR-HSA-5654149 (Reactome)
ADPArrowR-HSA-5654545 (Reactome)
ADPArrowR-HSA-5654560 (Reactome)
ADPArrowR-HSA-5654575 (Reactome)
ADPArrowR-HSA-5654578 (Reactome)
ADPArrowR-HSA-5654582 (Reactome)
ADPArrowR-HSA-5654587 (Reactome)
ADPArrowR-HSA-5654690 (Reactome)
ADPArrowR-HSA-5654692 (Reactome)
ADPArrowR-HSA-8853325 (Reactome)
ADPArrowR-HSA-934559 (Reactome)
ATPR-HSA-1295609 (Reactome)
ATPR-HSA-1839065 (Reactome)
ATPR-HSA-1839067 (Reactome)
ATPR-HSA-1839091 (Reactome)
ATPR-HSA-1839107 (Reactome)
ATPR-HSA-1839110 (Reactome)
ATPR-HSA-1839112 (Reactome)
ATPR-HSA-1888198 (Reactome)
ATPR-HSA-190427 (Reactome)
ATPR-HSA-190429 (Reactome)
ATPR-HSA-191062 (Reactome)
ATPR-HSA-1982066 (Reactome)
ATPR-HSA-2023455 (Reactome)
ATPR-HSA-2023460 (Reactome)
ATPR-HSA-5654149 (Reactome)
ATPR-HSA-5654545 (Reactome)
ATPR-HSA-5654560 (Reactome)
ATPR-HSA-5654575 (Reactome)
ATPR-HSA-5654578 (Reactome)
ATPR-HSA-5654582 (Reactome)
ATPR-HSA-5654587 (Reactome)
ATPR-HSA-5654690 (Reactome)
ATPR-HSA-5654692 (Reactome)
ATPR-HSA-8853325 (Reactome)
ATPR-HSA-934559 (Reactome)
Activated FGFR1:p-8T-FRS2ArrowR-HSA-5654560 (Reactome)
Activated FGFR1:p-FRS2:GRB2:GAB1:PI3KArrowR-HSA-5654591 (Reactome)
Activated FGFR1:p-FRS2:GRB2:GAB1:PI3Kmim-catalysisR-HSA-5654690 (Reactome)
Activated FGFR1:p-FRS2:GRB2:GAB1:PIK3R1ArrowR-HSA-5654592 (Reactome)
Activated FGFR1:p-FRS2:GRB2:GAB1:PIK3R1R-HSA-5654591 (Reactome)
Activated FGFR1:p-FRS2:GRB2:SOS1ArrowR-HSA-5654586 (Reactome)
Activated FGFR1:p-FRS2:GRB2:SOS1mim-catalysisR-HSA-5654392 (Reactome)
Activated FGFR1:p-FRS2:p-PTPN11:GRB2:GAB1:PI3KArrowR-HSA-5654596 (Reactome)
Activated FGFR1:p-FRS2:p-PTPN11:GRB2:GAB1:PI3Kmim-catalysisR-HSA-5654692 (Reactome)
Activated FGFR1:p-FRS2:p-PTPN11:p-CBL:GRB2ArrowR-HSA-5654673 (Reactome)
Activated FGFR1:p-FRS2:p-PTPN11:p-CBL:GRB2R-HSA-5654672 (Reactome)
Activated FGFR1:p-FRS2:p-PTPN11:p-CBL:GRB2mim-catalysisR-HSA-5654672 (Reactome)
Activated FGFR1:p-FRS2:p-PTPN11R-HSA-5654594 (Reactome)
Activated FGFR1:p-FRS2:p-PTPN11R-HSA-5654673 (Reactome)
Activated FGFR1:p-FRS2ArrowR-HSA-5654575 (Reactome)
Activated FGFR1:p-FRS2R-HSA-5654592 (Reactome)
Activated FGFR1:p-FRS3ArrowR-HSA-5654578 (Reactome)
Activated FGFR1:p-FRS:PTPN11ArrowR-HSA-5654584 (Reactome)
Activated FGFR1:p-FRS:PTPN11R-HSA-5654587 (Reactome)
Activated FGFR1:p-FRS:PTPN11mim-catalysisR-HSA-5654587 (Reactome)
Activated FGFR1:p-FRS:p-PTPN11ArrowR-HSA-5654587 (Reactome)
Activated FGFR1:p-FRS:p-PTPN11ArrowR-HSA-8941623 (Reactome)
Activated FGFR1:p-FRSR-HSA-5654584 (Reactome)
Activated FGFR1:p-FRSR-HSA-5654586 (Reactome)
Activated FGFR1:p-SHC1:GRB2:SOS1ArrowR-HSA-5654597 (Reactome)
Activated FGFR1:p-SHC1:GRB2:SOS1mim-catalysisR-HSA-5654600 (Reactome)
Activated FGFR1:p-SHC1ArrowR-HSA-5654582 (Reactome)
Activated FGFR1:p-SHC1R-HSA-5654597 (Reactome)
Activated FGFR:p-FRS2:p-PTPN11:GRB2:GAB1:PIK3R1ArrowR-HSA-5654594 (Reactome)
Activated FGFR:p-FRS2:p-PTPN11:GRB2:GAB1:PIK3R1R-HSA-5654596 (Reactome)
Activated

overexpressed

FGFR1b homodimer
ArrowR-HSA-1982066 (Reactome)
Activated

overexpressed

FGFR1c homodimer
ArrowR-HSA-5654545 (Reactome)
Activated FGFR1 P252X mutantsArrowR-HSA-2023455 (Reactome)
Activated FGFR1

mutant dimers with enhanced kinase

activity
ArrowR-HSA-2023460 (Reactome)
Activated FGFR1:FLRTArrowR-HSA-5656064 (Reactome)
Activated FGFR1:FRS2ArrowR-HSA-5654569 (Reactome)
Activated FGFR1:FRS2R-HSA-5654560 (Reactome)
Activated FGFR1:FRS2R-HSA-5654575 (Reactome)
Activated FGFR1:FRS2mim-catalysisR-HSA-5654575 (Reactome)
Activated FGFR1:FRS3ArrowR-HSA-5654571 (Reactome)
Activated FGFR1:FRS3R-HSA-5654578 (Reactome)
Activated FGFR1:FRS3mim-catalysisR-HSA-5654578 (Reactome)
Activated FGFR1:SHC1ArrowR-HSA-5654573 (Reactome)
Activated FGFR1:SHC1R-HSA-5654582 (Reactome)
Activated FGFR1:SHC1mim-catalysisR-HSA-5654582 (Reactome)
Activated FGFR1ArrowR-HSA-5654165 (Reactome)
Activated FGFR1R-HSA-5654167 (Reactome)
Activated FGFR1R-HSA-5654569 (Reactome)
Activated FGFR1R-HSA-5654571 (Reactome)
Activated FGFR1R-HSA-5654573 (Reactome)
Activated FGFR1R-HSA-5656064 (Reactome)
Activated FGFR1b homodimerArrowR-HSA-190427 (Reactome)
Activated FGFR1c

bound to

FGF23:Klotho
ArrowR-HSA-191062 (Reactome)
Activated FGFR1c homodimerArrowR-HSA-190429 (Reactome)
BCR-p-FGFR1 fusion mutant dimerR-HSA-1839067 (Reactome)
BCR-p-FGFR1 fusion mutant dimermim-catalysisR-HSA-1839067 (Reactome)
BRAFArrowR-HSA-1295604 (Reactome)
CBLArrowR-HSA-1295621 (Reactome)
CBLR-HSA-1295622 (Reactome)
FGF23 bound to Klotho and FGFR1cArrowR-HSA-190268 (Reactome)
FGF23 bound to Klotho and FGFR1cR-HSA-191062 (Reactome)
FGF23 bound to Klotho and FGFR1cmim-catalysisR-HSA-191062 (Reactome)
FGFR1 P252X mutant

dimers bound to

FGFs
ArrowR-HSA-2023451 (Reactome)
FGFR1 P252X mutant

dimers bound to

FGFs
R-HSA-2023455 (Reactome)
FGFR1 P252X mutant

dimers bound to

FGFs
mim-catalysisR-HSA-2023455 (Reactome)
FGFR1 P252X mutantsR-HSA-2023451 (Reactome)
FGFR1 fusion mutant dimers:TKIsArrowR-HSA-1839039 (Reactome)
FGFR1 mutant dimers

with enhanced

kinase activity
ArrowR-HSA-2023456 (Reactome)
FGFR1 mutant dimers

with enhanced

kinase activity
R-HSA-2023460 (Reactome)
FGFR1 mutant dimers

with enhanced

kinase activity
mim-catalysisR-HSA-2023460 (Reactome)
FGFR1 mutants with

enhanced kinase

activity
R-HSA-2023456 (Reactome)
FGFR1OP-p-FGFR1 fusion mutant dimermim-catalysisR-HSA-1888198 (Reactome)
FGFR1b homodimer bound to FGFArrowR-HSA-190245 (Reactome)
FGFR1b homodimer bound to FGFR-HSA-190427 (Reactome)
FGFR1b homodimer bound to FGFmim-catalysisR-HSA-190427 (Reactome)
FGFR1b homodimerArrowR-HSA-1982065 (Reactome)
FGFR1b homodimerR-HSA-1982066 (Reactome)
FGFR1b homodimermim-catalysisR-HSA-1982066 (Reactome)
FGFR1b-binding FGFsR-HSA-190245 (Reactome)
FGFR1bR-HSA-190245 (Reactome)
FGFR1bR-HSA-1982065 (Reactome)
FGFR1c homodimer bound to FGFArrowR-HSA-190256 (Reactome)
FGFR1c homodimer bound to FGFR-HSA-190429 (Reactome)
FGFR1c homodimer bound to FGFmim-catalysisR-HSA-190429 (Reactome)
FGFR1c homodimerArrowR-HSA-5654544 (Reactome)
FGFR1c homodimerR-HSA-5654545 (Reactome)
FGFR1c homodimermim-catalysisR-HSA-5654545 (Reactome)
FGFR1c-binding FGFsR-HSA-190256 (Reactome)
FGFR1c-binding FGFsR-HSA-2023451 (Reactome)
FGFR1c:KAL1ArrowR-HSA-5654514 (Reactome)
FGFR1c:KAL1TBarR-HSA-190256 (Reactome)
FGFR1cR-HSA-190256 (Reactome)
FGFR1cR-HSA-190268 (Reactome)
FGFR1cR-HSA-5654514 (Reactome)
FGFR1cR-HSA-5654544 (Reactome)
FGFRL1

dimer:SPRED1/2

dimer
ArrowR-HSA-5654510 (Reactome)
FGFRL1 dimerR-HSA-5654510 (Reactome)
FGFRL1 dimerR-HSA-5654511 (Reactome)
FGFRL1-binding FGFs:FGFRL1 dimerArrowR-HSA-5654511 (Reactome)
FGFRL1-binding FGFsR-HSA-5654511 (Reactome)
FLRT1,2,3R-HSA-5656064 (Reactome)
FRS2R-HSA-5654569 (Reactome)
FRS3R-HSA-5654571 (Reactome)
GDPArrowR-HSA-5654392 (Reactome)
GDPArrowR-HSA-5654600 (Reactome)
GDPArrowR-HSA-8941623 (Reactome)
GRB2-1:SOS1R-HSA-5654586 (Reactome)
GRB2-1:SOS1R-HSA-5654597 (Reactome)
GRB2-1ArrowR-HSA-1549564 (Reactome)
GRB2-1R-HSA-1295613 (Reactome)
GRB2:GAB1:PIK3R1ArrowR-HSA-177931 (Reactome)
GRB2:GAB1:PIK3R1R-HSA-5654592 (Reactome)
GRB2:GAB1:PIK3R1R-HSA-5654594 (Reactome)
GRB2:GAB1R-HSA-177931 (Reactome)
GRB2:GAB2R-HSA-1839095 (Reactome)
GTPR-HSA-5654392 (Reactome)
GTPR-HSA-5654600 (Reactome)
GTPR-HSA-8941623 (Reactome)
HSArrowR-HSA-190245 (Reactome)
HSArrowR-HSA-190256 (Reactome)
HSArrowR-HSA-190268 (Reactome)
HSR-HSA-190245 (Reactome)
HSR-HSA-190256 (Reactome)
HSR-HSA-190268 (Reactome)
HSR-HSA-2023451 (Reactome)
HSR-HSA-5654511 (Reactome)
HSR-HSA-5654515 (Reactome)
KAL1:HSArrowR-HSA-190256 (Reactome)
KAL1:HSArrowR-HSA-5654515 (Reactome)
KAL1R-HSA-5654514 (Reactome)
KAL1R-HSA-5654515 (Reactome)
Klotho bound to FGF23R-HSA-190268 (Reactome)
Overexpressed FGFR1:TKIsArrowR-HSA-2023462 (Reactome)
Overexpressed FGFR1 homodimersR-HSA-2023462 (Reactome)
PI(3,4,5)P3ArrowR-HSA-1839091 (Reactome)
PI(3,4,5)P3ArrowR-HSA-1839107 (Reactome)
PI(3,4,5)P3ArrowR-HSA-5654690 (Reactome)
PI(3,4,5)P3ArrowR-HSA-5654692 (Reactome)
PI(3,4,5)P3R-HSA-5654167 (Reactome)
PI(4,5)P2R-HSA-1839091 (Reactome)
PI(4,5)P2R-HSA-1839107 (Reactome)
PI(4,5)P2R-HSA-5654690 (Reactome)
PI(4,5)P2R-HSA-5654692 (Reactome)
PIK3CAR-HSA-1839080 (Reactome)
PIK3CAR-HSA-1839102 (Reactome)
PIK3CAR-HSA-5654591 (Reactome)
PIK3CAR-HSA-5654596 (Reactome)
PIK3R1R-HSA-177931 (Reactome)
PIK3R1R-HSA-1839078 (Reactome)
PIK3R1R-HSA-1839114 (Reactome)
PLCG1R-HSA-5654167 (Reactome)
PP2A (A:C)ArrowR-HSA-1295622 (Reactome)
PP2A(A:C):S112/S121-pSPRY2ArrowR-HSA-934559 (Reactome)
PP2A(A:C):S112/S121-pSPRY2TBarR-HSA-1295609 (Reactome)
PP2A(A:C):SPRY2ArrowR-HSA-1295599 (Reactome)
PP2A(A:C):SPRY2R-HSA-1295609 (Reactome)
PP2A(A:C):SPRY2R-HSA-934559 (Reactome)
PP2A(A:C):Y55/Y227-pSPRY2ArrowR-HSA-1295609 (Reactome)
PP2A(A:C):Y55/Y227-pSPRY2ArrowR-HSA-1549564 (Reactome)
PP2A(A:C):Y55/Y227-pSPRY2R-HSA-1295613 (Reactome)
PP2A(A:C):Y55/Y227-pSPRY2R-HSA-1295622 (Reactome)
PPA2A

(A:C):S112/S115

p-SPRY2
R-HSA-1295632 (Reactome)
PPA2A

(A:C):S112/S115

p-SPRY2
mim-catalysisR-HSA-1295632 (Reactome)
PPA2A (A:C):Y55/Y227 p-SPRY2:GRB2ArrowR-HSA-1295613 (Reactome)
PPA2A (A:C):Y55/Y227 p-SPRY2:GRB2R-HSA-1549564 (Reactome)
PPA2A(A:C):SPRY2ArrowR-HSA-1295632 (Reactome)
PPA2A(A:C):SPRY2R-HSA-1295599 (Reactome)
PTPN11R-HSA-5654584 (Reactome)
PTPN11mim-catalysisR-HSA-1549564 (Reactome)
PiArrowR-HSA-1295632 (Reactome)
Plasma membrane FGFR1 fusion dimersArrowR-HSA-8853322 (Reactome)
Plasma membrane FGFR1 fusion dimersR-HSA-8853325 (Reactome)
Plasma membrane FGFR1 fusion dimersmim-catalysisR-HSA-8853325 (Reactome)
Plasma membrane p-Y FGFR1 fusion dimersArrowR-HSA-8853325 (Reactome)
R-HSA-1295599 (Reactome) SPRY2 translocates to the plasma membrane upon activation of cells with FGF, and translocation is required for the inhibition of growth factor-stimulated cell migration, proliferation and differentiation. Translocation may be mediated by interactions with PIP2 in the membrane, palmitoylation of the C-terminal region of SPRY2 and/or interactions with caveolin-1.
R-HSA-1295604 (Reactome) MAPK-dependent serine phosphorylation of SPRY2 disrupts complex formation with B-RAF.
R-HSA-1295609 (Reactome) Sprouty 2 protein is phosphorylated on tyrosine residue 55. The ability of SRC kinase to catalyze this reaction has been demonstrated with purified proteins in vitro (Li et al. 2004) and in cultured cells with studies of the effects of SRC-family pharmacological inhibitors and of dominant-negative mutant SRC proteins (Mason et al. 2004). SRC kinase also phosphorylates numerous tyrosine residues in the C terminal region of SPRY2 including Y227, in response to FGF but not EGF stimulation.
R-HSA-1295613 (Reactome) Some evidence suggests that SPRY2 may exert its negative effect by binding to GRB2 and competing with the GRB2:SOS1 interaction that is required for MAPK activation. SPRY2 phosphorylation at Y55 is stimulated in response to both FGF and EGF, and is required for SPRY2 to act as a negative regulator of FGF signaling. Y55 is not thought to be a GRB2 binding site, however. Instead, phosphorylation at Y55 is thought to cause a conformational change in SPRY2 that reveals a cryptic PXXPXPR GRB2-docking site in the C-terminal of SPRY2.
SPRY2 has also been shown to be phosphorylated at multiple tyrosine residues in its C-terminal in response to FGF, but not EGF, stimulation. This phosphorylation, in particular at residue 227, is thought to augment the ability of SPRY2 to inhibit FGF signaling through the MAPK cascade, although the mechanism remains to be elucidated.
R-HSA-1295621 (Reactome) After ubiquitination, CBL dissociates from SPRY2
R-HSA-1295622 (Reactome) The N terminal TKB domain of CBL binds to the phospho-tyrosine 55 of SPRY2, targeting SPRY2 for degradation by the 26S proteasome. Y55 is also a binding site for PP2A, which dephosphorylates numerous serine and threonine residues on SPRY2, allowing a conformational change that may promote a SPRY2:GRB2 interaction and limit the extent of MAPK activation following FGF stimulation.
R-HSA-1295632 (Reactome) In unstimulated cells, SPRY2 has been shown to be phosphorylated on multiple serine and threonine residues. In these cells, SPRY2 exists in a complex with the regulatory and catalytic subunits (A and C, respectively) of the serine/threonine phosphatase PP2A. After stimulation with FGF, the catalytic activity of PP2A increases and the phosphatase dephophorylates SPRY at serine 112 and serine 115. This is thought to promote changes in tertiary structure that promote GRB2 binding and phosphorylation of Y55 and Y227.
R-HSA-1295634 (Reactome) Some evidence suggests that SPRY2 can exert its negative role on FGF signaling at the level of RAF activation. Hypophosphorylated SPRY2 binds to inactive B-RAF, preventing it from activating ERK signaling. MAPK activation results in phosphorylation of SPRY2 on six serine residues (S7, S42, S111, S120, S140 and S167), and inhibits B-RAF binding. Phosphorylation at S111 and S120 directly affects B-RAF binding while the remaining four sites appear to contribute indirectly. Oncogenic forms of B-RAF such as B-RAF V600E, which adopt active kinase conformations, do not associate with SPRY2, regardless of its phosphorylation status. This suggests that two mechanisms affect the SPRY2:B-RAF interaction: SPRY2 phosphorylation and B-RAF conformation.
R-HSA-1549564 (Reactome) PPTN11 (also known as SHP2) may exert its positive effects on MAPK activation in response to FGF stimulation by catalyzing the dephosphorylation of tyrosine resides on SPRY2. This dephosphorylation promotes dissociation of the GRB2/SPRY2 complex and as a consequence stimulates GRB2 association with the activated receptor, leading to sustained MAPK signaling.
R-HSA-177931 (Reactome) The Src homology 2 (SH2) domain of the phosphatidylinositol 3-kinase (PIK3) regulatory subunit (PIK3R1, i.e. PI3Kp85) binds to GAB1 in a phosphorylation-independent manner. GAB1 serves as a docking protein which recruits a number of downstream signalling proteins. PIK3R1 can bind to either GAB1 or phosphorylated GAB1.
R-HSA-1839031 (Reactome) 8p11 myeloproliferative syndrome (EMS) is a myeloproliferative disorder that rapidly progresses to acute myeloid leukemia if not treated (reviewed in Jackson, 2010, Knights and Cook, 2010). A characteristic feature of EMS is the presence of fusion proteins that contain the kinase domain of FGFR1 and the oligomerization domain of an unrelated protein. This is believed to promote the ligand independent dimerization and activation of the kinase domain. To date, there are 11 identified partners that form fusion proteins with FGFR1 in EMS: ZMYM2 (Xiao, 1998; Popovici, 1998; Reiter, 1998; Ollendorff, 1999; Xiao, 2000), FGFR1OP1 (Popovici, 1999), CNTRL (Guasch, 2000), BCR (Demiroglu, 2001), FGFR1OP2 (Grand, 2004), TRIM24 (Belloni, 2005), CUX1 (Wasag, 2011), MYO18A (Walz, 2005), CPSF6 (Hidalgo-Curtis, 2008), HERV-K (Guasch, 2003) and LRRFIP1 (Soler, 2009).
R-HSA-1839039 (Reactome) In a murine mouse model of ZNF198-FGFR1-induced EMS, treatment with the FGFR-inhibitor Midostaurin (PKC412) resulted in prolonged survival (Chen, 2004). Similarly, growth of ZNF-198-FGFR1-, FGFR1OP2-FGFR1-, and BCR-FGFR1-expressing lines is blocked by treatment with FGFR-inhibitors (Demiroglu, 2001; Gu, 2006; Chase, 2007; Zhen, 2007; Wasag, 2011).
R-HSA-1839065 (Reactome) After ligand-independent dimerization, FGFR1 fusions are trans-autophosphorylated on tyrosine residues (see for instance Popovici, 1998; Ollendorff, 1999; Guasch, 2000). Although the sites of tyrosine phosphorylation have not been mapped in the context of the fusion proteins, at least some of the same residues appear to be phosphorylated as in full length FGFR1. For instance, phospho-specific antibodies have demonstrated that TRIM24 is phosphorylated on Y653 and Y654, the activation loop tyrosines of FGFR1 (Belloni, 2005). Likewise, FGFR1 fusions with ZMYM2, BCR, FGFR1OP and TRIM24 all result in recruitment and phosphorylation of PLCgamma, and where mutational studies have been performed, mutation of the PLCgamma binding site Y766 has been shown to abrogate this signaling (Roumiantsev, 2004, Lelievre, 2008, Chase, 2007). In the case of BCR-FGFR1, the BCR moiety of the fusion protein has also been shown to be phosphorylated on at least one tyrosine residue, Y177, which results in the recruitment of GRB2 (Roumiantsev, 2004).
R-HSA-1839067 (Reactome) Unique among FGFR1 fusion proteins, which generally give rise to an atypical myeloproliferative syndrome (EMS) (reviewed in Jackson, 2010), the BCR-FGFR1 fusion results in a more typical chronic myeloid leukemia (CML). Although both EMS and CML activate PLCgamma signaling, and mutation of the FGFR1 Y766 PLCgamma binding site attenuates both diseases, CML-specific signaling also appears to be mediated through the BCR portion of the fusion protein. BCR Y177 binds GRB2-GAB1 and induces CML-like leukemia in mice, while expression of a Y177F BCR-FGFR1 fusion induces EMS-like disease (Roumiantsev, 2004).
R-HSA-1839078 (Reactome) Activation of the PI3K signaling pathway has been demonstrated for a number of FGFR1 fusion proteins and inhibitors of this pathway impair the proliferative and survival function of the fusions (Guasch, 2001; Demiroglu, 2001; Chen, 2004; Lelievre, 2008). FGFR1 fusions lack the FRS2-binding site of the full length protein, so the mechanism of PI3K recruitment is unclear. Unlike BCR-FGFR1, which has been shown to recruit GRB2 through the BCR Y177 site, GRB2 did not co-precipitate with the ZMYM2-FGFR1 fusion (Roumianetsev, 2004). In the case of FOP-FGFR1, Y730 has been shown to be required for the recruitment of the p85 subunit of PI3K; however, CEP110-FGFR1, which contains Y730 in the context of the same pYXXM motif, was not shown to recruit p85 at the centrosome (Guasch, 2001).
R-HSA-1839080 (Reactome) Activation of the PI3K pathway has been demonstrated in the case of ZMYM2-FGFR1 (Chen, 2004), BCR-FGFR1 (Demiroglu, 2001) and FOP-FGFR1 (Guasch, 2001), and is presumed to occur to a greater or lesser extent in other FGFR1 fusions as well (reviewed in Jackson, 2010). Activation of the PI3K pathway suggests that the PIK3CA catalytic subunit must be recruited to the fusion protein.
R-HSA-1839091 (Reactome) Once recruited to the activated receptor, PI3K phosphorylates PIP2 to PIP3, leading to activation of AKT signaling. PI3K signaling has been demonstrated in ZMYM2-, FOP- and BCR-FGFR1 fusions (Chen, 2004; Demiroglu, 2001; Guasch, 2001), as well as downstream of a number of other FGFR mutants (see for instance, Byron, 2008; Kunii, 2008; Agazie, 2003; Takeda, 2007).
R-HSA-1839095 (Reactome) Proliferation of BCR-FGFR1 fusion proteins is blocked by treatment with the PI3K inhibitor LY294002, suggesting the activation of this pathway downstream of BCR-FGFR1 phosphorylation. Y177 has been shown to be a binding site for GRB2 and to be required for the both the phosphorylation of GAB2 and the development of CML-like disease (Roumiantsev, 2004, Demiroglu, 2001). By analogy with studies in BCR-ABL, where mutation of Y177 abrogates recruitment of PI3K activity to the fusion protein (Sattler, 2002), this suggests that Y177 may serve as a docking site for a complex of GRB2:GAB1:PI3K in the context of BCR-FGFR1 as well.
R-HSA-1839102 (Reactome) Activation of the PI3K pathway has been demonstrated in the case of ZMYM2-FGFR1 (Chen, 2004), BCR-FGFR1 (Demiroglu, 2001) and FOP-FGFR1 (Guasch, 2001), and is presumed to occur to a greater or lesser extent in other FGFR1 fusions as well (reviewed in Jackson, 2010). Activation of the PI3K pathway suggests that the PIK3CA catalytic subunit must be recruited to the fusion protein.
R-HSA-1839107 (Reactome) Once recruited to the activated BCR-FGFR1 fusion PI3K phosphorylates PIP2 to PIP3, leading to activation of AKT signaling (Roumiantsev, 2004; Demiroglu, 2001).
R-HSA-1839110 (Reactome) Recruitment of GAB2 to the BCR-FGFR1 fusion protein results in GAB2 phosphorylation (Roumiatnetsev, 2004). As in the case of BCR-ABL (Sattler, 2002), recruitment and phosphorylation of GAB2 is dependent on BCR residue Y177. Deletion of Y177 abolishes GRB2 recruitment and converts the more aggressive MPD disorder induced by BCR-FGFR1 to the EMS characteristic of other FGFR1 fusions (Demiroglu, 2001; Roumianetsev, 2004)
R-HSA-1839112 (Reactome) Activation of a subset of FGFR1-fusions (ZMYM2, BCR, FGFR1OP2 and CUX) has been shown to result in downstream phosphorylation of STAT5 proteins at Y694. This phosphorylation is dependent on the FGFR1 fusion, as the STAT5 phosphorylation is abrogated in the presence of an FGFR1-kinase dead fusion (Heath and Cross, 2004; Smedley, 1999; Chase, 2007; Wasage, 2011).
R-HSA-1839114 (Reactome) Based on analogy with studies of the BCR-ABL fusion, phosphorylated GAB2 recruits the regulatory subunit of PI3K to the BCR-FGFR1 fusion (Sattler, 2002; Demiroglu, 2001; Roumiantsev, 2004).
R-HSA-1888198 (Reactome) Expression of FGFR1OP-FGFR1 in both Ba/F3 and Cos-1 cells leads to phosphorylation of STAT1 and STAT3 but not STAT5, and to activation of a STAT1/3-responsive reporter when expressed in NIH3T3 cells (Guasch, 2001). Activation of STAT proteins has also been shown to be oncogenic in the context of derivatives of FGFR1, 3 and 4 that lack the extracellular domain and are are targetted to the plasma membrane by a myristylation signal (Hart et al, 2000).
R-HSA-190245 (Reactome) In this reaction, FGF receptor in the plasma membrane binds an associating extracellular ligand, a requisite step for subsequent activation. The resulting complex consists of dimerized receptor, two ligand molecules, and heparan sulfate. NCAM and other members of the CAM protein family directly or indirectly modulate this interaction in a variety of neural tissues. The details of this interaction in vivo have not been definitively established at the molecular level, but are thought to play a central role in the regulation of the development of these tissues.
R-HSA-190256 (Reactome) In this reaction, FGF receptor in the plasma membrane binds an associating extracellular ligand, a requisite step for subsequent activation. The resulting complex consists of dimerized receptor, two ligand molecules, and heparan sulfate.
R-HSA-190268 (Reactome) In this reaction, FGF receptor in the plasma membrane binds an associating extracellular ligand, a requisite step for subsequent activation. The resulting complex consists of dimerized receptor, two ligand molecules, and heparan sulfate.
R-HSA-190427 (Reactome) Studies have mapped 8 tyrosine residues in the cytoplasmic domain of FGFR1 that are important for signaling. Autophosphorylation of residues 653 and 654, located in the activation loop of the kinase, is necessary to maintain the receptor in the active state. Phosphorylation of other tyrosine residues by the intrinsic protein tyrosine kinase activity of the activated receptor creates binding sites on its cytoplasmic tail for membrane bound docking proteins to gather intracellular signaling mediators.
R-HSA-190429 (Reactome) Studies have mapped 8 tyrosine residues in the cytoplasmic domain of FGFR1 that are important for signaling. Autophosphorylation of residues 653 and 654, located in the activation loop of the kinase, is necessary to maintain the receptor in the active state. Phosphorylation of other tyrosine residues by the intrinsic protein tyrosine kinase activity of the activated receptor creates binding sites on its cytoplasmic tail for membrane bound docking proteins to gather intracellular signaling mediators.
R-HSA-191062 (Reactome) Studies have mapped 8 tyrosine residues in the cytoplasmic domain of FGFR1 that are important for signaling. Autophosphorylation of residues 653 and 654, located in the activation loop of the kinase, is necessary to maintain the receptor in the active state. Phosphorylation of other tyrosine residues by the intrinsic protein tyrosine kinase activity of the activated receptor creates binding sites on its cytoplasmic tail for membrane bound docking proteins to gather intracellular signaling mediators.
R-HSA-1982065 (Reactome) FGFR1-amplified lung cancer and breast cancer cells show strong phosphorylation of FGFR1 and do not show elevated levels of FGF ligand, suggesting that these receptors can undergo ligand-independent activation. Phosphorylation is enhanced in the presence of exogenous ligand, supporting the notion that overexpressed FGFR1 can be activated by both ligand- and ligand-independent pathways (Koziczak, 2004; Dutt, 2008; Weiss, 2010). The biochemical consequences of overexpression of FGFR1 in other cancer types remain to be determined (reviewed in Turner and Gross, 2010; Wesche, 2011.
R-HSA-1982066 (Reactome) FGFR1-amplified lung cancer and breast cancer cells show strong phosphorylation of FGFR1 and do not show elevated levels of FGF ligand, suggesting that these receptors can undergo ligand-independent activation. Phosphorylation is enhanced in the presence of exogenous ligand, supporting the notion that overexpressed FGFR1 can be activated by both ligand- and ligand-independent pathways (Koziczak, 2004; Dutt, 2008; Weiss, 2010). The biochemical consequences of overexpression of FGFR1 in other cancer types remain to be determined (reviewed in Turner and Gross, 2010; Wesche, 2011.
R-HSA-2023451 (Reactome) The missense mutation C775G in exon 5 of FGFR1 encodes a Pro252R gain-of-function mutation that causes Pfeiffer syndrome, an autosomal dominant disorder characterized by premature fusion of bones in the skull and syndactyly of the hands and feet (Muenke, 1994). FGFR1 P252R binds to FGF1, FGF2, FGF4, and FGF6 with 2-5 fold-enhanced affinity, and with 30-fold affinity to FGF9. The enhanced ligand-affinity of the mutant receptor is the result of an additional set of ligand-receptor hydrogen bonds; in particular for FGF9, the additional receptor contacts are thought to compete with FGF9 autoinhibitory dimerization (Ibrahimi, 2004a). The increase in ligand-binding affinity in the absence of an expansion of ligand binding range is thought to account for the milder limb phenotype of Pfeiffer syndrome relative to FGFR2-mediated Apert syndrome (Yu, 2000; Ibrahimi, 2004b).

Somatic mutations in FGFR1 at P252 have also been identified in melanoma (P252S; Ruhe, 2007) and in lung cancer (P252T; Davies, 2005). Based on analogy to the FGFR1 P252R mutation that is found in Pfeiffer syndrome, these mutations are also predicted to increase the ligand-binding affinity of the receptor and to result in increased signaling, although this remains to be directly demonstrated for the S/T alleles (Ibrahimi, 2004a).

R-HSA-2023455 (Reactome) FGFR1 gain-of-function mutations at P252 that result in increased binding affinity to ligand are presumed to be phosphorylated on the same sites as the wild-type receptor, although this has not been demonstrated (Ibrahimi, 2004a).


R-HSA-2023456 (Reactome) Large scale genomic characterization of glioblastoma tumors has identified three point mutants in the kinase domain of FGFR1: N546K, R576W and K656E (Rand, 2005, TCGA, 2008), representing the first kinase domain point mutants identified in this gene in any cancer. These mutants are believed or have been shown to have enhanced kinase activity and to be able to function in a ligand-independent manner (Petiot, 2002; Lew, 2009; Raffioni, 1998, Rand, 2005; Hart, 2000)
R-HSA-2023460 (Reactome) The three kinase domain mutants of FGFR1 that have been identified in glioblastoma are predicted or have been shown to result in enhanced kinase activity. The N546K (Rand, 2005) residue lies in a stretch of 9 amino acids that are conserved between all four FGFRs. Mutation of the paralogous residue in FGFR3 (N540K) has been shown to result in weak ligand-independent contstitutive activation in the autosomal disorder hypochodroplasia (Raffioni, 1998). In FGFR2 mutation of the paralogous residue to lysine has been identified in endometrial cancer and been shown to result in enhanced kinase activity (Dutt, 2008; Pollock, 2008); germline mutations at this site in FGFR2 are also associated with the development of Crouzon and Pfeiffer syndromes (Kan, 2002). The FGFR1 N546K mutations has accelerated rates of autophosphorylation and supports transformation when transfected into Rat-1 cells (Lew, 2009).


The FGFR1 K656E (TCGA, 2008) mutation is paralogous to activating mutations in FGFR3 kinase domain associated with the development of thanatophoric dysplasias (Tavormina, 1999; Bellus, 2000; Hart, 2000), and has itself been shown to activating when expressed in neural crest cells (Petiot, 2002).


The FGFR1 R576W (Rand, 2005) mutation increases the hydrophobicity of the receptor, and is postulated to enhance protein-protein interactions and thereby increase the likelihood of autophosphorylation of adjacent tyrosine residues, although this has not been explicitly demonstrated.



R-HSA-2023462 (Reactome) Treatment of FGFR1-amplified lung and breast cancer cell lines with the in vitro reagents PD173704, SU5402 and FIIN-1 inhibits proliferation, while cells expressing wild-type levels of FGFR1 are insensitive to inhibitors, suggesting that amplified FGFR1 may be a suitable therapeutic target in some cancer lines (Weiss, 2010; Reis-Filho, 2006; Dutt, 2011; Turner, 2010). In fact, a number of other small molecule inhibitors, including Dovitinib and AZD4547, are currently in clinical trials for treatment of FGFR1-amplified cancers (reviewed in Turner and Grose, 2010; Wesche, 2011; http://ClinicalTrials.gov)
R-HSA-5654149 (Reactome) PLC gamma is phosphorylated by activated FGFR, resulting in PLC gamma activation, stimulation of phosphatidyl inositol hydrolysis and generation of two second messengers, diacylglycerol and inositol (1,4,5) P3. Tyrosine phosphorylation of PLCgamma by FGFR4 is weaker than that seen by other isoforms of FGFR.
R-HSA-5654165 (Reactome) Dissociation from the activated receptor quickly follows phosphorylation of PLC-gamma. Phosphorylated PLC-gamma catalyzes the hydrolysis of phosphatidylinositol(4, 5)bisphosphate to generate two second messengers, diacylglycerol and inositol (1,4,5) triphosphate.
R-HSA-5654167 (Reactome) Recruitment of PLC-gamma by FGF receptors has been best studied in FGFR1c signaling, where it has been shown that autophosphorylation of Tyr766 in the C-terminal tail of FGFR1c creates a specific binding site for the SH2 domain of PLC-gamma. A mutant FGFR1c in which Y766 is replaced by phenylalanine is unable to activate PI hydrolysis and Ca2+ release in response to FGF stimulation. Membrane recruitment of PLC-gamma is also aided by binding of the Pleckstrin homology (PH) domain of this enzyme to PtIns(3,4,5) P3 molecules that are generated in response to PI-3 kinase stimulation. By sequence comparison, Y766 is conserved in all FGFR isoforms, and PLC-gamma signaling is observed, to a greater or lesser extent, downstream of all FGFR receptors upon stimulation with FGFs.
R-HSA-5654392 (Reactome) SOS, recruited by GRB2:p-FRS2 to activated FGFR, activates RAS nucleotide exchange from the inactive GDP-bound to the active GTP-bound state.
R-HSA-5654510 (Reactome) FGFRL1 binds to SPRED1 and 2 and Sprouty1 as assessed by co-immunoprecipitation, although the exact stoichiometry of the complex remains to be determined. The interaction requires the C-terminal residues of the short intracellular domain of FGFRL1 (Zhuang et al, 2011). The SPRED proteins are members of the Sprouty family, with established roles as negative regulators of the Ras/Raf/Erk signaling pathway (reviewed in McClatchey and Cichowski, 2012).
R-HSA-5654511 (Reactome) FGFRL1 is a fifth member of the FGFR family of receptors that shares 40% sequence similarity with the extracellular region of FGFR1-4, but FGFRL1 lacks the internal kinase domain required for typical downstream FGFR signaling. Instead, FGFRL1 has a short intracellular domain with a C-terminal histidine rich region that has been shown to interact with the MAP kinase regulator SPRED proteins (Sleeman et al, 2001; Zhuang et al, 2011; reviewed in Trueb et al, 2013). FGFRL1 forms constitutive dimers and has been shown to bind to a wide range of FGF ligands, including FGF3,4,8,10, 22 and with lower affinity to FGF2,5,17,18 and 23 (Reickman et al, 2008; Steinberg et al, 2010). FGFRL1 knockout mice die shortly after birth from lung and renal defects (Gerber et al, 2009; Gerber et al, 2012; Trueb et al, 2013). FGFRL1 has been postulated to act as a decoy receptor that sequesters ligand away from canonical FGF receptors; more recently, however, alternate roles for FGFRL1 in enhancing ERK1/2 activation or promoting FGFR1-mediated signaling have been suggested (Sleeman et al, 2001; Steinberg et al, 2010; Silva et al, 2013; Amann and Trueb, 2013). Further work will be required to elucidate the role(s) of FGFRL1.
R-HSA-5654514 (Reactome) KAL1 is an extracellular matrix-associated protein that modulates signaling by FGFR1c. Mutations in the KAL1 gene are associated with Kallman syndrome, a genetic disorder characterized by olfactory bulb dysgenesis and hypogonadotrophic hypogonadism (Dode et al, 2003; Pitteloud et al, 2006; reviewed in Hu and Bouloux, 2010). KAL1 has been shown to interact with both FGFR1c and with heparan sulfate, with opposing effects on downstream signaling. Preformation of an FGFR1c:KAL1 complex inhibits the association of FGF ligand with the complex and subsequent receptor dimerization and in this way negatively regulates FGFR1c ligand-dependent signaling. In contrast, preformation of a KAL1:heparan sulfate complex promotes stable FGF ligand:receptor interaction thereby enhancing FGFR1c signal transduction (Hu et al, 2009; Hu et al, 2004; Soussi-Yanicostas et al, 1998).
KAL1 consists of an N-terminal cysteine rich domain, a whey acidic protein-like (WAP) domain, four fibronectin III (FnIII) repeats and a C-terminal histidine rich region. The N-terminal cysteine rich region, the WAP domain and the first FnIII domain contribute to the interaction with the D2 and D3 Ig-like domains of FGFR1c. D1 and the acid box of the receptor inhibit the interaction with KAL1 in a manner analogous to the inhibition of FGF binding (Hu et al, 2009). Consistent with this, missense mutations in D1 and the acid box that affect the interaction with KAL1 have been identified in patients with Kallmann syndrome (Dode and Hardelin, 2009). Similarly, loss-of function mutations in the FnIII domain of KAL1 that disrupt the interaction with FGFR1c have also been characterized (Hu et al, 2009; Robertson et al, 2001; Gonzalez-Martinez et al 2004; Oliviera et al, 2001).
R-HSA-5654515 (Reactome) KAL1 is an extracellular matrix-associated protein that modulates signaling by FGFR1c. Mutations in the KAL1 gene are associated with Kallmann syndrome, a genetic disorder characterized by olfactory bulb dysgenesis and hypogonadotrophic hypogonadism (Dode et al, 2003; Pitteloud et al, 2006; reviewed in Yu and Bouloux, 2010). KAL1 has been shown to interact with both FGFR1c and with heparan sulfate, with opposing effects on downstream signaling. Preformation of an FGFR1c:KAL1 complex inhibits the association of FGF ligand with the complex and subsequent receptor dimerization and in this way negatively regulates FGFR1c ligand-dependent signaling. In contrast, preformation of a KAL1:heparan sulfate complex promotes stable FGF ligand:receptor interaction thereby enhancing FGFR1c signal transduction (Hu et al, 2009; Hu et al, 2004; Soussi-Yanicostas et al, 1998).
R-HSA-5654544 (Reactome) FGFR1-amplified lung cancer and breast cancer cells show strong phosphorylation of FGFR1 and do not show elevated levels of FGF ligand, suggesting that these receptors can undergo ligand-independent activation. Phosphorylation is enhanced in the presence of exogenous ligand, supporting the notion that overexpressed FGFR1 can be activated by both ligand- and ligand-independent pathways (Koziczak, 2004; Dutt, 2008; Weiss, 2010). The biochemical consequences of overexpression of FGFR1 in other cancer types remain to be determined (reviewed in Turner and Gross, 2010; Wesche, 2011.
R-HSA-5654545 (Reactome) FGFR1-amplified lung cancer and breast cancer cells show strong phosphorylation of FGFR1 and do not show elevated levels of FGF ligand, suggesting that these receptors can undergo ligand-independent activation. Phosphorylation is enhanced in the presence of exogenous ligand, supporting the notion that overexpressed FGFR1 can be activated by both ligand- and ligand-independent pathways (Koziczak, 2004; Dutt, 2008; Weiss, 2010). The biochemical consequences of overexpression of FGFR1 in other cancer types remain to be determined (reviewed in Turner and Gross, 2010; Wesche, 2011.
R-HSA-5654560 (Reactome) FRS2 has 8 canonical MAPK phosphorylation sites which are phosphorylated by activated ERK1/2 after FGF stimulation. Phosphorylation of these 8 threonine residues counteracts the activating effect of tyrosine phosphorylation of FRS2, although the exact mechanism for this negative regulation is not known. Expression of a version of FRS2 in which the 8 threonine residues are mutated to valine results in enhanced tyrosine phosphorylation of FRS2, enhanced GRB2-SOS1 recruitment and a more sustained MAPK response. The 8 threonine residues are not conserved in FRS3; as a result, signaling through FRS3 complexes do not appear to be subject to this downregulation.
R-HSA-5654569 (Reactome) FRS2 (also known as FRS2alpha) is broadly expressed in adult and fetal tissues. Membrane-bound FRS2 interacts with FGFR as a first step in the phosphorylation of this docking protein. The juxtamembrane binding site for FRS2 does not contain tyrosine, so binding may be independent of receptor activation and/or constitutive. Activation of the FGFR receptor is required for FRS2 phosphorylation and subsequent recruitment of downstream effectors.
R-HSA-5654571 (Reactome) FRS3 (also known as FRS2beta) is predominantly expressed in the developing and adult neuroepithelium. As is the case for FRS2 (also known as FRS2alpha), binding of FRS3 to FGFR may be constitutive and/or independent of receptor activation. Elements of the downstream signaling mediated by the two FRS family members appear to be at least partially conserved, as FRS3 is phosphorylated upon FGF stimulation, binds PPTN11/SHP2 and GRB2 and results in ERK activation. Moreover, expression of FRS3 in FRS2-/- MEFs restores ERK activation.
R-HSA-5654573 (Reactome) Although a role for SHC1 in FGF signalling has been implicated in many studies, it is not clear that SHC1 interacts directly with the receptor.
R-HSA-5654575 (Reactome) FRS2 (also known as FRS alpha is activated through tyrosine phosphorylation catalyzed by the protein kinase domain of the activated FGFR. FRS2 contains four binding sites for the adaptor protein GRB2 at residues Y196, Y306, Y349 and Y392, and two binding sites for the protein tyrosine phosphatase PPTN11/SHP2 at residues Y436 and Y471. Different FGFR isoforms may generate different phosphorylation patterns on FRS2 leading to alternate downstream signaling.
R-HSA-5654578 (Reactome) FRS3 (also known as FRS2 beta) is activated through tyrosine phosphorylation catalyzed by the protein kinase domain of the activated FGFR. By sequence comparison, FRS3 has the 2 PPTN11/SHP2-binding sites and has three of the four GRB2-binding sites.
R-HSA-5654582 (Reactome) The p46 and p53 isoforms of SHC1 have been shown to be phosphorylated upon FGF stimulation. Three consensus RTK phosphoryation sites are present in SHC1, although phosphorylation of these specific tyrosine residues has not been explicitly demonstrated in response to FGF stimulation. In contrast, the p66 isoform of SHC1 does not appear to undergo FGF-dependent phosphorylation.
R-HSA-5654584 (Reactome) p-FRS2 has two PPTN11/SHP2-binding sites at pY436 and pY471.
R-HSA-5654586 (Reactome) Tyrosine phosphorylated FRS2 recruits GRB2:SOS1 complex by means of the SH3 domain of GRB2, leading to RAS-MAP kinase activation. The FRS2:GRB2-mediated pathway plays a minor role in the activation of RAS-MAP kinase pathway compared to that mediated by FRS2:PPTN11.
R-HSA-5654587 (Reactome) Tyrosine phosphorylation of PPTN11/SHP2 by FGFR kinase is required for activation of the phosphatase activity of PPTN11 and for downstream signaling. Tyrosine phosphorylated PPTN11 plays a major role in the activation of RAS-MAP kinase pathway, although the precise role is not yet clear.
R-HSA-5654591 (Reactome) The 110 kDa catalytic subunit (PIK3CA) binds to the 85 kDa regulatory subunit (PIK3R1) to create the active PIK3.
R-HSA-5654592 (Reactome) The direct GRB2-binding sites of FRS2 have a major role in activation of the PI3K pathway.
R-HSA-5654594 (Reactome) p-PPTN11 recruits GRB2-GAB1 to the activated receptor.
R-HSA-5654596 (Reactome) The 110 kDa catalytic subunit (PIK3CA) binds to the 85 kDa regulatory subunit (PIK3R1) to create the active PIK3.
R-HSA-5654597 (Reactome) Phosphorylated SHC1 links FGFR to Grb2 (Klint et al. 1995) leading to the formation of a signaling complex including Shc, Grb2 and Sos. Transformation of NIH 3T3 cells with v-Src produced a strong constitutive association of FGFR1 with Shc, Grb2 and Sos (Curto et al. 1998) suggesting Src involvement. Recruitment of Grb2-Sos links FGFR to the Ras pathway.
R-HSA-5654600 (Reactome) SOS, recruited by GRB2:p-FRS2 to activated FGFR, activates RAS nucleotide exchange from the inactive GDP-bound to the active GTP-bound state.
R-HSA-5654672 (Reactome) Grb2 bound to tyrosine phosphorylated FRS2 forms a ternary complex with Cbl through the binding of the SH3 domains of Grb2 to a proline rich region in Cbl. Grb2-mediated recruitment of Cbl results in ubiquitination of FGFR and FRS2. Cbl is a multidomain protein that posses an intrinsic ubiquitin ligase activity and also functions as a platform for recruitment of a variety of signaling proteins. Multiple mechanisms appear to be required for downregulation of FGFR, as internalization of the receptor is reduced but not abolished if recruitment of CBL to FRS2 is compromised by mutation of GRB2-binding sites.
R-HSA-5654673 (Reactome) The ubiquitin ligase CBL exists in a complex with GRB2 and is recruited to tyrosine-phosphorylated FRS2 after FGF stimulation. In addition to promoting the ubiquitination, endocytosis, and degradation of the activated receptor complex, recruitment of the p-CBL:GRB2 complex seems to attenuate FGFR signaling by competing with GRB2:SOS1 for binding to the direct GRB2-binding sites on p-FRS2.
R-HSA-5654690 (Reactome) Once recruited to the membrane, PI3K catalyzes the phosphorylation of PI(4,5)P2 to PI(3,4,5)P3.
R-HSA-5654692 (Reactome) Once recruited to the activated receptor, PI3K phosphorylates PIP2 to PIP3, leading to activation of AKT signaling. PI3K signaling has been demonstrated in ZMYM2-, FOP- and BCR-FGFR1 fusions (Chen, 2004; Demiroglu, 2001; Guasch, 2001), as well as downstream of a number of other FGFR mutants (see for instance, Byron, 2008; Kunii, 2008; Agazie, 2003; Takeda, 2007).
R-HSA-5656064 (Reactome) The three fibronectin-leucine-rich transmembrane (FLRT) proteins were identified as positive regulators of FGFR signaling that enhance FGFR-dependent RAS/MAPK pathway activation. All three FLRT proteins have been shown to interact with FGFR1 by co-immunoprecipitation and, at least in the case of FLRT3, the interaction is mediated by the FLRT fibronectin-like domain (Bottcher et al, 2004; Haines et al, 2006). Each FLRT gene has a distinct expression pattern and the strength of the protein-protein interaction with the FGF receptor varies, allowing for cell-type specific modulation of signaling activity (Haines et al, 2006). How the FLRT proteins act to enhance FGFR-dependent MAPK pathway activation is not clear, however FLRT1 has recently been shown to be phosphorylated in an FGFR1- and Src family kinase (SFK)-dependent manner (Wheldon et al, 2010).
R-HSA-8853322 (Reactome) Although dimerization of the FGFR1 fusions in glioblastoma, breast cancer and non small cell lung cancer hasn't been directly demonstrated, the ability of these proteins to promote transformation and tumorigenesis suggests that they form active oligomers as is the case for WT FGFR1 proteins (Singh et al, 2012; Wang et al, 2013; Wang et al, 2014; reviewed in Parker et al, 2014).
R-HSA-8853325 (Reactome) Although it hasn't been directly demonstrated in all cases, the ability to promote transformation and anchorage independent growth suggests these fusions undergo autophosphorylation similar to WT FGFR1 proteins. Indeed, active kinase activity has been demonstrated for the the ERLIN2-FGFR1 fusion identified in breast cancer (Singh et al, 2012; Wu et al, 2013; Wang et al, 2014; reviewed in Parker et al, 2014)
R-HSA-8941623 (Reactome) RAS nucleotide is stimulated downstream of activated FGFR1 in a p-PTPN11-dependent manner. The phosphatase activity of PTPN11 is required for activation of the RAS-MAP kinase pathway, although the mechanism for RAS pathway activation is not yet clear (Hadari et al, 1998; reviewed in Mohi et al, 2007; Gotoh et al, 2008).
R-HSA-934559 (Reactome) In humans, the phosphorylated MNK1 kinase phosphorylates the adaptor protein Sprouty2 on Ser112 and Ser121, and also at some other serine and threonine residues. MNK1 appears not to form a complex with Sprouty2. Some of these (including the two main sites mentioned above) conform to the serine-containing consensus sites for phosphorylation by MNK1 kinase (K/R-X-X-S, R-X-S). It appears that serine phosphorylation is required to protect Sprouty2 from degradation.

In the absence of serine phosphorylation, phosphorylation of Tyr55 and subsequent binding to E3 ubiquitin ligase, CBL, is enhanced. Serine phosphorylation of Sprouty2 appears to stabilise the protein by interfering with its potential phosphorylation of Tyr55 (Sprouty2 appears to be a poor substrate for c-Src kinase) in response to growth factor stimulation.
R-HSA-934604 (Reactome) In humans, the phosphorylated adaptor protein Sprouty2 is ubiquitinated by the E3 ubiquitin ligase CBL, marking it for degradation by the 26S proteasome.
S111/S120 p-SPRY2:B-RAFArrowR-HSA-1295634 (Reactome)
S111/S120 p-SPRY2:B-RAFR-HSA-1295604 (Reactome)
SHC1 p46,p52R-HSA-5654573 (Reactome)
SPRED1/2 dimerR-HSA-5654510 (Reactome)
SPRY2:B-RAFR-HSA-1295634 (Reactome)
SRC-1mim-catalysisR-HSA-1295609 (Reactome)
STAT1,3R-HSA-1888198 (Reactome)
STAT5A,STAT5BR-HSA-1839112 (Reactome)
Tyrosine kinase

inhibitors of

overexpressed FGFR1
R-HSA-2023462 (Reactome)
Tyrosine kinase

inhibitors of FGFR1

fusion mutants
R-HSA-1839039 (Reactome)
Ub-(Y55/Y227)p-SPRY2ArrowR-HSA-1295621 (Reactome)
Ub-Activated FGFR1 complex:Ub-p-FRS2ArrowR-HSA-5654672 (Reactome)
Ub:Y55/Y227-pSPRY2:CBLArrowR-HSA-934604 (Reactome)
Ub:Y55/Y227-pSPRY2:CBLR-HSA-1295621 (Reactome)
UbR-HSA-5654672 (Reactome)
UbR-HSA-934604 (Reactome)
Y55/Y227-pSPRY2:CBLArrowR-HSA-1295622 (Reactome)
Y55/Y227-pSPRY2:CBLR-HSA-934604 (Reactome)
Y55/Y227-pSPRY2:CBLmim-catalysisR-HSA-934604 (Reactome)
activated FGFR1:PLCG1ArrowR-HSA-5654167 (Reactome)
activated FGFR1:PLCG1R-HSA-5654149 (Reactome)
activated FGFR1:PLCG1mim-catalysisR-HSA-5654149 (Reactome)
activated FGFR1:p-4Y-PLCG1ArrowR-HSA-5654149 (Reactome)
activated FGFR1:p-4Y-PLCG1R-HSA-5654165 (Reactome)
cytosolic FGFR1

fusion mutant

dimers
ArrowR-HSA-1839031 (Reactome)
cytosolic FGFR1

fusion mutant

dimers
R-HSA-1839039 (Reactome)
cytosolic FGFR1

fusion mutant

dimers
R-HSA-1839065 (Reactome)
cytosolic FGFR1

fusion mutant

dimers
mim-catalysisR-HSA-1839065 (Reactome)
cytosolic FGFR1 fusion mutantsR-HSA-1839031 (Reactome)
cytosolic p-FGFR1

fusion mutant

dimers
ArrowR-HSA-1839065 (Reactome)
cytosolic p-FGFR1

fusion mutant

dimers
R-HSA-1839078 (Reactome)
cytosolic p-FGFR1

fusion mutant

dimers
mim-catalysisR-HSA-1839112 (Reactome)
p-4Y-PLCG1ArrowR-HSA-5654165 (Reactome)
p-FGFR1 fusion

mutant

dimers:PIK3R1
ArrowR-HSA-1839078 (Reactome)
p-FGFR1 fusion

mutant

dimers:PIK3R1
R-HSA-1839080 (Reactome)
p-FGFR1 mutant fusions:PI3KArrowR-HSA-1839080 (Reactome)
p-FGFR1 mutant fusions:PI3Kmim-catalysisR-HSA-1839091 (Reactome)
p-S111,S120-SPRY2ArrowR-HSA-1295604 (Reactome)
p-STAT5A, p-STAT5BArrowR-HSA-1839112 (Reactome)
p-T,Y MAPK dimersArrowR-HSA-1295634 (Reactome)
p-T,Y MAPK dimersmim-catalysisR-HSA-5654560 (Reactome)
p-T250,T255,T385,S437-MKNK1mim-catalysisR-HSA-934559 (Reactome)
p-Y177-BCR-pY-FGFR1 mutant:GRB2:p-GAB2:PIK3R1ArrowR-HSA-1839114 (Reactome)
p-Y177-BCR-pY-FGFR1 mutant:GRB2:p-GAB2:PIK3R1R-HSA-1839102 (Reactome)
p-Y371-CBL:GRB2R-HSA-5654673 (Reactome)
p21 RAS:GDPR-HSA-5654392 (Reactome)
p21 RAS:GDPR-HSA-5654600 (Reactome)
p21 RAS:GDPR-HSA-8941623 (Reactome)
p21 RAS:GTPArrowR-HSA-5654392 (Reactome)
p21 RAS:GTPArrowR-HSA-5654600 (Reactome)
p21 RAS:GTPArrowR-HSA-8941623 (Reactome)
pY-STAT1,3ArrowR-HSA-1888198 (Reactome)
pY177-BCR-p-FGFR1 fusion mutant dimerArrowR-HSA-1839067 (Reactome)
pY177-BCR-p-FGFR1 fusion mutant dimerR-HSA-1839095 (Reactome)
pY177-BCR-pY-FGFR1 mutant:GRB2:p-GAB1:PI3KArrowR-HSA-1839102 (Reactome)
pY177-BCR-pY-FGFR1 mutant:GRB2:p-GAB1:PI3Kmim-catalysisR-HSA-1839107 (Reactome)
pY177-BCR-pY-FGFR1 mutant:GRB2:p-GAB2ArrowR-HSA-1839110 (Reactome)
pY177-BCR-pY-FGFR1 mutant:GRB2:p-GAB2R-HSA-1839114 (Reactome)
pY177-BCR1-p-FGFR1 mutant:GRB2:GAB2ArrowR-HSA-1839095 (Reactome)
pY177-BCR1-p-FGFR1 mutant:GRB2:GAB2R-HSA-1839110 (Reactome)
pY177-BCR1-p-FGFR1 mutant:GRB2:GAB2mim-catalysisR-HSA-1839110 (Reactome)
plasma membrane FGFR1 fusionsR-HSA-8853322 (Reactome)
Personal tools