Signaling by FGFR3 (Homo sapiens)

From WikiPathways

Jump to: navigation, search
2, 12, 28, 35, 46...54, 9470, 9740, 48, 57, 795433, 75, 86, 12822, 114, 12845, 1193922, 33, 68, 865, 6, 19, 29, 44...19, 44, 72, 100, 111...10931, 33, 68, 77, 7821, 53, 75, 7795, 11221, 75, 7715, 49, 56, 85, 96...10613, 20, 67, 107, 111...1, 8, 23, 41, 51...13, 17, 61, 76, 88...90, 101, 13440, 48, 57, 798, 23, 41, 81, 104...18, 86, 8938, 1052436, 50, 65, 10868, 14033, 12949, 1391053, 75, 77, 13322, 8633, 12938, 1051182493, 103, 106, 126, 131118, 13467, 82, 84, 107, 111...1033, 1293, 14, 101423964, 66, 87, 1023, 69, 11268, 78, 864, 5414, 10118, 43, 73, 14036, 65, 114cytosolcytosolGolgi lumenPP2A(A:C):SPRY2GalNAc-T178-FGF23(25-251) S-Farn-Me PalmS NRAS FGF5-1 FGF9 FGF1 FGFR3 K650M FGFR3b-binding FGFsp-6Y-FGFR3 K650E FGF2(10-155) p-6Y-FGFR3 K650N GAB1 p-4Y FGFR3(23-793)-TACC3(143-838) fusion FGF16 UBC(305-380) FGF9 RPS27A(1-76) FGF8-1 FGF18 FGF5-1 p-6Y-FRS2 p6Y-FGFR3c P250R BRAF UBC(381-456) ActivatedFGFR3:p-FRS2:p-PTPN11:GRB2:GAB1:PI3KATPGRB2-1 FGF9 FGF5-1 FGF18 FGF9 ATPADPp-Y239,Y240,Y317-SHC1-2 HS GalNAc-T178-FGF23(25-251) UBB(153-228) GalNAc-T178-FGF23(25-251) FGF9 UBC(77-152) p-6Y-FGFR3b FGF8-1 PLCG1FGF16 FGF16 PPP2CA FGF20 FGFR3 p-Y FGFR3 fusiondimersp-6Y-FGFR3 G380R FGF20 ActivatedFGFR3:p-FRS:p-PTPN11FGF18 UBC(153-228) p-6Y-FGFR3 K650T FGFR3 cysteinemutantsFGF8-1 GalNAc-T178-FGF23(25-251) UBC(381-456) GalNAc-T178-FGF23(25-251) FGF4 UBC(381-456) p-Y194,Y195,Y272-SHC1-3 FGF1 p-6Y-FGFR3c FGFR3 point mutantswith enhancedkinase activityPTPN11 UBB(77-152) FGF20 HS FGF17-1 FGF9 FGF18 FGF5-1 FGF5-1 p-6Y-FGFR3c FGF16 FGF17-1 FGF5-1 FGFR3bFGF17-1 FGF4 GAB1 UBC(457-532) FGF5-1 FGF17-1 p-Y55,Y227-SPRY2 activatedFGFR3:p-4Y-PLCG1PI(3,4,5)P3FGF8-1 PP2A(A:C):Y55/Y227-pSPRY2p-6Y-FGFR3b GalNAc-T178-FGF23(25-251) UBC(77-152) FGF5-1 FGF9 FGF16 SRC-1UBC(609-684) FGF20 FGF20 FGF18 FGFR3(23-758)-TACC3(549-838) fusion FGF17-1 FGFR3 795fs*139 FGFR3 G380R p-6Y-FGFR3b ATPFGF2(10-155) HS UBA52(1-76) PPP2R1A FGFR3 K650N UBC(457-532) FGF17-1 SPRY2 p-6Y-FGFR3c GRB2-1 GRB2-1 FGFR3c P250RFGF4 S-Farn-Me KRAS4B FGF9 FGF20 GalNAc-T178-FGF23(25-251)FGF8-1 RPS27A(1-76) FGFR3c FGF18 p-Y546,Y584-PTPN11 FGF18 FGF16 UBC(1-76) UBA52(1-76) FGF1 UBB(153-228) PI(4,5)P2FGF18 FGF18 Activated FGFR3cysteine mutantsFGF20 PPP2CA FRS2 HS p-6Y-FGFR3 G380R FGFR3 mutant dimersFGFR3 A391E p-6Y-FGFR3c FGF20 HS FRS3PIK3R1 UBC(533-608) UBC(609-684) GTP S-Farn-Me KRAS4B ATPFGF18 p-6Y-FGFR3 G370C FGF1 HS p-6Y-FGFR3c FGF8-1 GalNAc-T178-FGF23(25-251) FGF20 FGF16 FGF4 FGF4 FGF1 FGF1 FGFR3 G382D FGF5-1 FGF16 SOS1 FGF18 FGF8-1 FGF18 GalNAc-T178-FGF23(25-251) FGF1 FGFR3(23-758)-TACC3(649-838) fusion FGF8-1 Activated FGFR3cP250R mutantp-Y55,Y227-SPRY2 CBL GalNAc-T178-FGF23(25-251) p-6Y-FGFR3 S371C p-6Y-FGFR3 Y373C UBC(77-152) PIK3R1 p-Y55,Y227-SPRY2 FGF16 FGF16 pY-FGFR3 FGFR3c P250R FGF1 FGFR3 K650N p-Y371-CBL FGF2(10-155) p-Y546,Y584-PTPN11 FGFR3 (4;14) translocation mutant dimers p-6Y-FGFR3b FGF1 FGF20 FGFR3 K650T HS FGFR3 cysteine mutant dimers FGFR3 G370C CBL FGF4 FGF1 PTPN11FGFR3b G697C FGF16 ActivatedFGFR3:p-FRS2HSGALNT3GRB2:GAB1UBC(609-684) FGF8-1 p-T250,T255,T385,S437-MKNK1UBC(153-228) p-T185,Y187-MAPK1 S-Farn-Me-PalmS KRAS4A FGF2(10-155) p-6Y-FGFR3c FGF20 FGF20 GalNAc-T178-FGF23(25-251) PPP2CB UBC(1-76) FGFR3(23-760)-TACC3(647-838) fusion FGF8-1 FGF17-1 FGF1 FGF17-1 FGF2(10-155) FGF8-1 FGF20 p21 RAS:GTPp-S112,S121-SPRY2 GAB1 FGFR3b G697C FGF9 FGF16 FGF4 FGFR3 S371C p-6Y-FGFR3b FGF20 ActivatedFGFR3:p-FRS2:p-PTPN11:p-CBL:GRB2UDP-GalNAcFGF5-1 GRB2:GAB1:PIK3R1p-S111,S120-SPRY2FGF2(10-155) HS Activated FGFR3:FRS2PIK3R1p-6Y-FRS2 FGFR3cUBC(457-532) ADPATPFGF1 p-6Y-FGFR3 Y373C UBC(305-380) p-Y546,Y584-PTPN11 FGF17-1 FGFR3 K650E ADPGalNAc-T178-FGF23(25-251) FGFR3 R248C ActivatedFGFR3:p-FRS2:p-PTPN11FGF18 FGF8-1 Y55/Y227-pSPRY2:CBLFGFR3 G380R FGF1 GalNAc-T178-FGF23(25-251) p-6Y-FGFR3 795fs*139 FGF20 FGF17-1 GRB2-1 PIK3CAp-6Y-FGFR3c ADPGalNAc-T178-FGF23(25-251)FGF18 FGF5-1 FGF17-1 CBLFGF2(10-155) UBC(77-152) FGFR3 Y373C FGFR3c P250R FGFR3(23-793)-TACC3(143-838) fusion FGFR3 (4;14)translocationmutantsp-6Y-FRS2 p-6Y-FGFR3b FGF2(10-155) UBC(153-228) FGF9 FGFR3(23-760)-BAIAP2L1(18-522) fusion UDPp-5Y-FRS3 FGF4 ActivatedFGFR3:p-FRSFGFR3 point mutant dimers with enhanced kinase activity FGF20 FGF2(10-155) GalNAc-T178-FGF23(25-251) Ubp-Y546,Y584-PTPN11 GalNAc-T178-FGF23(25-251) FGF9 GalNAc-T178-FGF23(25-251) UBB(1-76) FGFR3b homodimerbound to FGFUBA52(1-76) p-Y371-CBL:GRB2UBC(381-456) PPP2CA HS FGF4 FGF8-1 PiFGF17-1 GDP ADPBRAFUBC(533-608) p-8T-FRS2 S-Farn-Me-2xPalmS HRAS FGF18 FGF4 FGF4 PPP2R1A FGFR3 G382D PPP2R1A p-6Y-FGFR3b FGFR3c-binding FGFsp-6Y-FGFR3c FGF4 UBB(1-76) GRB2-1 Activated FGFR3mutants withenhanced kinaseactivityGRB2-1FGF20 FGF20 FGFR3 K650E p-5Y-FRS3 RPS27A(1-76) FGF5-1 FGF18 p-6Y-FGFR3c p-S112,S115-SPRY2 GDPGRB2-1 FGF20 FGF2(10-155) PIK3CA p-6Y-FGFR3b p-Y55,Y227-SPRY2 p-Y239,Y240,Y317-SHC1-2 FGFR3 Y373C FGF16 UBC(153-228) FGF17-1 p-4Y FGFR3(23-760)-TACC3(647-838) fusion FGFR3b S249C FGF20 FGFR3 R248C UBC(533-608) FGF2(10-155) FGFR3(23-760)-TACC3(612-838) fusion FGF9 ADPFGF18 FGF20 FGF20 ADPADPFGF16 FGFR3 fusion dimersFGF17-1 FGFR3c P250R mutantdimer bound to FGFp-6Y-FGFR3b FGF5-1 PPP2CA FGF16 p-6Y-FGFR3c PIK3R1 PP2A(A:C):S112/S121-pSPRY2HS FGF8-1 FGF2(10-155) FGF16 p-6Y-FGFR3b FGF1 FGF1 GAB1 FGFR3 point mutant dimers with enhanced kinase activity FGF2(10-155) FGF2(10-155) p-6Y-FGFR3c FGF16 FGF16 SHC1-2 HS ActivatedFGFR3:p-FRS2:GRB2:GAB1:PIK3R1FGF18 p21 RAS:GDPADPFGF9 PPP2R1A UBC(153-228) FGFR3 G382D ADPFGF8-1 FGF18 HS PPA2A(A:C):S112/S115p-SPRY2UBA52(1-76) p-5Y-FRS3 GRB2-1 p-6Y-FGFR3b FGF20 p-6Y-FGFR3b ActivatedFGFR3:p-FRS2:p-PTPN11:GRB2:GAB1:PIK3R1FGF18 FGF20 FGF18 FGF20 UBC(229-304) FGF9 p-6Y-FRS2 activatedFGFR3:PLCG1p-6Y-FGFR3c PPA2A(A:C):SPRY2FGF16 FGF9 GRB2-1 FGFR3 G380R Ub-Activated FGFR3complex:Ub-p-FRS2FGF8-1 PPP2CB FGFR3 G370C ATPFGF17-1 FGF5-1 p-6Y-FRS2 FGF9 FGF17-1 FGF20 RPS27A(1-76) FGF9 HS FGF18 UBB(77-152) FGF1 S-Farn-Me PalmS NRAS FGF9 FGF17-1 p-4Y-PLCG1ADPFGFR3b ATPFGF8-1 RAF/MAP kinasecascadep-6Y-FGFR3 G382D FGF2(10-155) GalNAc-T178-FGF23(25-251) HS FGF20 ATPFGF17-1 FGF17-1 UBB(77-152) FGF2(10-155) FGF5-1 FGFR3 (4;14) translocation mutant dimers FGF2(10-155) FGF4 FGF2(10-155) FGF8-1 ATPFGFR3 R248C UBC(305-380) FGF17-1 FGF9 HS FGF16 FGF8-1 PPP2CA FGFR3 FGF4 p-6Y-FGFR3c FGF9 UBC(609-684) p-6Y-FGFR3b GalNAc-T178-FGF23(25-251) SPRY2:B-RAFGalNAc-T178-FGF23(25-251) FGF8-1 SPRY2 FGFR3 (4;14)translocationmutant dimersPI(3,4,5)P3 FGF17-1 FGF1 FGF17-1 p-6Y-FGFR3 K650E p-6Y-FGFR3b GRB2-1 p-6Y-FRS2 FGFR3 G382D SHC1-2,SHC1-3p-6Y-FRS2 FGF8-1 FGF20 HS GRB2-1 FGF2(10-155) FGF1 FGF18 FGF20 FGF4 PPA2A (A:C):Y55/Y227p-SPRY2:GRB2Activated FGFR3t(4;14)translocationmutantsp-6Y-FGFR3b FGFR3(23-760)-BAIAP2L1(18-522) fusion ActivatedFGFR3:pY-SHC1:GRB2:SOS1FGFR3c homodimerbound to FGFp-Y194,Y195,Y272-SHC1-3 GalNAc-T178-FGF23(25-251) GTPPPP2CB ATPFGF17-1 FGF16 FGF5-1 GalNAc-T178-FGF23(25-251) FGF8-1 FGF4 SHC1-2 FGF1 FGF4 PPP2CB FGF4 UBC(1-76) p-6Y-FGFR3b G697C FGF17-1 PIK3R1 FGF18 FGF17-1 FGF5-1 FGFR3(23-758)-TACC3(649-838) fusion p-4Y-PLCG1 FGF9 FGF5-1 FGFR3 795fs*139 FGF17-1 p-S111,S120-SPRY2 FGFR3(23-758)-TACC3(549-838) fusion FGF9 HS Activated FGFR3chomodimerp-6Y-FGFR3b UBC(533-608) FGF5-1 UBC(229-304) UBB(1-76) FGF5-1 p-6Y-FGFR3c p-6Y-FGFR3 K650M PPP2R1A FGFR3 K650Q FGF5-1 GalNAc-T178-FGF23(25-251) HS SHC1-3 p-6Y-FGFR3c FGF8-1 p-6Y-FGFR3c Ub:Y55/Y227-pSPRY2:CBLUBC(381-456) HS UBC(77-152) FGFR3b S249C GalNAc-T178-FGF23(25-251) p-6Y-FGFR3c FGF4 FGF9 FGF4 RPS27A(1-76) GAB1 UBC(457-532) BRAF PPP2R1A FGF5-1 S-Farn-Me-PalmS KRAS4A FGF4 Activated FGFR3UBC(229-304) FGF18 p-6Y-FRS2 FGF1 UbADPUBB(1-76) FGF4 GalNAc-T178-FGF23(25-251) FGFR3 cysteine mutant dimers FGF8-1 p-6Y-FRS2 p-6Y-FGFR3b FGF9 FGF17-1 FGF2(10-155) GalNAc-T178-FGF23(25-251) FGF17-1 FGF2(10-155) HS FGFR3 Y373C HS GRB2-1 UBC(1-76) p-6Y-FRS2 FGF16 FGF8-1 FGFR3 K650T ATPFGFR3 R248C FGF1 SOS1 FGF4 FGF2(10-155) SOS1 FGF9 FGF5-1 FGF17-1 FGF9 FGF9 FGF2(10-155) FGFR3 K650E HS Activated FGFR3:FRS3HS FGFR3(23-760)-TACC3(612-838) fusion UBC(305-380) Tyrosine kinaseinhibitors of FGFR3mutantsActivatedFGFR3:pY-SHC1HS FGF1 FGF8-1 p-6Y-FGFR3b FGF8-1 UBB(153-228) FGFR3 Y373C FGF1 FGF9 FGF1 FGF4 FGF17-1 ActivatedFGFR3:p-FRS2:GRB2:GAB1:PI3KFGF2(10-155) FGF16 PIP3 activates AKTsignalingActivated FGFR3:SHC1FGF8-1 p-6Y-FGFR3c FGF2(10-155) FGF18 PI(3,4,5)P3SPRY2 HS p-6Y-FGFR3 G382D FGF18 FGF9 ADPFGF16 GalNAc-T178-FGF23(25-251) FGF16 UBC(533-608) FGF1 S-Farn-Me-2xPalmS HRAS GAB1 p-4Y FGFR3(23-758)-TACC3(649-838) fusion FGF17-1 PPP2CB FGF5-1 UBB(153-228) FGFR3 K650Q ATPPLCG1 p-4Y FGFR3(23-760)-TACC3(612-838) fusion UBB(153-228) ActivatedFGFR3:p-8T-FRS2p-5Y-FRS3 FGFR3 G380R FGF8-1 FGF1 SHC1-3 GalNAc-T178-FGF23(25-251) p-6Y-FRS2 ATPFGF5-1 ADPActivatedFGFR3:p-FRS:PTPN11ATPp-T,Y MAPK dimersp-6Y-FGFR3 R248C PPP2CA FGF9 FGF2(10-155) FGF4 FGF8-1 GRB2-1 FGFR3 fusionsp-4Y FGFR3(23-758)-TACC3(549-838) fusion UBC(229-304) FGF18 p-4Y FGFR3(23-760)-BAIAP2L1(18-522) fusion p-6Y-FGFR3b FGF9 GalNAc-T178-FGF23(25-251) PP2A (A:C)p-5Y-FRS3 ActivatedFGFR3:p-FRS3UBB(1-76) FGF18 FGF4 FGF16 PPP2R1A p-6Y-FRS2 UBC(609-684) HS UBC(1-76) p-6Y-FGFR3c p-Y55,Y227-SPRY2 FGFR3(23-760)-TACC3(647-838) fusion p-6Y-FGFR3 A391E HS FGFR3c P250R PPP2CA FGF17-1 p-Y371-CBL FGF20 FGF5-1 FGF8-1 PIK3R1 p-Y371-CBL FGF4 FGFR3 A391E GalNAc-T178-FGF23(25-251) HS FGF9 HSActivated FGFR3bhomodimerFGF2(10-155) PIK3CA Ub-(Y55/Y227)p-SPRY2FGF20 UBC(305-380) HS FGF18 p-6Y-FGFR3c UBB(77-152) p-6Y-FGFR3c FRS3 p-6Y-FGFR3c FGFR3 cysteinemutant dimersFGF4 FGF5-1 FGF1 GalNAc-T178-FGF23(25-251) p-6Y-FGFR3 K650Q UBC(229-304) ActivatedFGFR3:p-FRS:GRB2:SOS1GRB2-1 FGF18 FGF16 FGF16 FGF1 FGF1 UBC(457-532) FGF1 FGF5-1 FGF16 FGFR3 S371C p-6Y-FGFR3 R248C DAG and IP3signalingPPP2CB FGF18 FGF20 FGF4 p-6Y-FGFR3b p-6Y-FGFR3b FGF8-1 p-T202,Y204-MAPK3 FGF1 ATPHS p-6Y-FGFR3c GRB2-1:SOS1FGF2(10-155) FGFR3 mutantdimers:TKIsFGF2(10-155) FRS2FGFR3 K650E p-6Y-FGFR3b S249C mutant p-Y546,Y584-PTPN11 p-Y546,Y584-PTPN11 FGF1 UBA52(1-76) UBB(77-152) FGF5-1 FGFR3(23-793)-TACC3(143-838) fusion FGFR3 K650M FGF23(25-251)p-6Y-FGFR3b S111/S120p-SPRY2:B-RAFPPP2CB FGFR3 point mutantdimers withenhanced kinaseactivityADPp-6Y-FGFR3b 10647, 9619, 67, 80100111, 13015, 16, 30, 34, 96...116, 121116, 12144, 72, 8054116, 12144, 67, 72, 98, 99, 111...44, 67, 72, 98, 99, 111...19, 80, 136127106116, 1215, 19, 80116, 12111, 82, 11310647, 966739, 12555, 80, 8367111, 130116, 12155, 80, 831, 63, 121111, 13044, 67, 72, 98, 99, 111...11, 82, 11363119, 67, 8044, 67, 72, 98, 138116, 12115, 16, 30, 34, 96...1, 63, 121116, 12114044, 67, 72, 98, 99, 111...44, 67, 72, 98, 13855, 80, 8315, 58, 71, 9854116, 121116, 12111, 82, 113144, 7215, 16, 30, 34, 96...72, 809, 25, 26, 32, 37...44, 72, 8044, 80, 99, 111116, 1215419, 80, 1361001186315, 58, 71, 9861, 76, 88, 122, 123119, 80, 13627, 7219, 67, 8044, 72, 8011, 47, 1131061, 63, 12119, 67, 806344, 72, 9954544, 72, 9927, 7211, 82, 11319, 80, 1365, 19, 8027, 7211, 47, 11315, 16, 30, 34, 96...44, 80, 99, 1117116, 121 >


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: 5654741
Reactome-version 
Reactome version: 75
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. Capelletti M, Dodge ME, Ercan D, Hammerman PS, Park SI, Kim J, Sasaki H, Jablons DM, Lipson D, Young L, Stephens PJ, Miller VA, Lindeman NI, Munir KJ, Richards WG, Jänne PA.; ''Identification of recurrent FGFR3-TACC3 fusion oncogenes from lung adenocarcinoma.''; PubMed Europe PMC Scholia
  2. Ornitz DM, Marie PJ.; ''FGF signaling pathways in endochondral and intramembranous bone development and human genetic disease.''; PubMed Europe PMC Scholia
  3. 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
  4. DaSilva J, Xu L, Kim HJ, Miller WT, Bar-Sagi D.; ''Regulation of sprouty stability by Mnk1-dependent phosphorylation.''; PubMed Europe PMC Scholia
  5. Zhang Y, Hiraishi Y, Wang H, Sumi KS, Hayashido Y, Toratani S, Kan M, Sato JD, Okamoto T.; ''Constitutive activating mutation of the FGFR3b in oral squamous cell carcinomas.''; PubMed Europe PMC Scholia
  6. Greulich H, Pollock PM.; ''Targeting mutant fibroblast growth factor receptors in cancer.''; PubMed Europe PMC Scholia
  7. 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
  8. 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
  9. Turjanski AG, Vaqué JP, Gutkind JS.; ''MAP kinases and the control of nuclear events.''; PubMed Europe PMC Scholia
  10. Kato K, Jeanneau C, Tarp MA, Benet-Pagès A, Lorenz-Depiereux B, Bennett EP, Mandel U, Strom TM, Clausen H.; ''Polypeptide GalNAc-transferase T3 and familial tumoral calcinosis. Secretion of fibroblast growth factor 23 requires O-glycosylation.''; PubMed Europe PMC Scholia
  11. Tomlinson DC, Baldo O, Harnden P, Knowles MA.; ''FGFR3 protein expression and its relationship to mutation status and prognostic variables in bladder cancer.''; PubMed Europe PMC Scholia
  12. Beenken A, Mohammadi M.; ''The FGF family: biology, pathophysiology and therapy.''; PubMed Europe PMC Scholia
  13. Trudel S, Stewart AK, Rom E, Wei E, Li ZH, Kotzer S, Chumakov I, Singer Y, Chang H, Liang SB, Yayon A.; ''The inhibitory anti-FGFR3 antibody, PRO-001, is cytotoxic to t(4;14) multiple myeloma cells.''; PubMed Europe PMC Scholia
  14. 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
  15. Tavormina PL, Rimoin DL, Cohn DH, Zhu YZ, Shiang R, Wasmuth JJ.; ''Another mutation that results in the substitution of an unpaired cysteine residue in the extracellular domain of FGFR3 in thanatophoric dysplasia type I.''; PubMed Europe PMC Scholia
  16. Intini D, Baldini L, Fabris S, Lombardi L, Ciceri G, Maiolo AT, Neri A.; ''Analysis of FGFR3 gene mutations in multiple myeloma patients with t(4;14).''; PubMed Europe PMC Scholia
  17. Bernard-Pierrot I, Gruel N, Stransky N, Vincent-Salomon A, Reyal F, Raynal V, Vallot C, Pierron G, Radvanyi F, Delattre O.; ''Characterization of the recurrent 8p11-12 amplicon identifies PPAPDC1B, a phosphatase protein, as a new therapeutic target in breast cancer.''; PubMed Europe PMC Scholia
  18. Gotoh N.; ''Regulation of growth factor signaling by FRS2 family docking/scaffold adaptor proteins.''; PubMed Europe PMC Scholia
  19. Webster MK, Donoghue DJ.; ''Constitutive activation of fibroblast growth factor receptor 3 by the transmembrane domain point mutation found in achondroplasia.''; PubMed Europe PMC Scholia
  20. Krejci P, Murakami S, Prochazkova J, Trantirek L, Chlebova K, Ouyang Z, Aklian A, Smutny J, Bryja V, Kozubik A, Wilcox WR.; ''NF449 is a novel inhibitor of fibroblast growth factor receptor 3 (FGFR3) signaling active in chondrocytes and multiple myeloma cells.''; PubMed Europe PMC Scholia
  21. 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
  22. 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
  23. Singh P, Thomas GE, Gireesh KK, Manna TK.; ''TACC3 protein regulates microtubule nucleation by affecting γ-tubulin ring complexes.''; PubMed Europe PMC Scholia
  24. Carpenter G, Ji Q.; ''Phospholipase C-gamma as a signal-transducing element.''; PubMed Europe PMC Scholia
  25. Plotnikov A, Zehorai E, Procaccia S, Seger R.; ''The MAPK cascades: signaling components, nuclear roles and mechanisms of nuclear translocation.''; PubMed Europe PMC Scholia
  26. Wellbrock C, Karasarides M, Marais R.; ''The RAF proteins take centre stage.''; PubMed Europe PMC Scholia
  27. Sibley K, Cuthbert-Heavens D, Knowles MA.; ''Loss of heterozygosity at 4p16.3 and mutation of FGFR3 in transitional cell carcinoma.''; PubMed Europe PMC Scholia
  28. 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
  29. Lindgren D, Liedberg F, Andersson A, Chebil G, Gudjonsson S, Borg A, Månsson W, Fioretos T, Höglund M.; ''Molecular characterization of early-stage bladder carcinomas by expression profiles, FGFR3 mutation status, and loss of 9q.''; PubMed Europe PMC Scholia
  30. Chou A, Dekker N, Jordan RC.; ''Identification of novel fibroblast growth factor receptor 3 gene mutations in actinic cheilitis and squamous cell carcinoma of the lip.''; PubMed Europe PMC Scholia
  31. 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
  32. Brown MD, Sacks DB.; ''Protein scaffolds in MAP kinase signalling.''; PubMed Europe PMC Scholia
  33. 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
  34. Soverini S, Terragna C, Testoni N, Ruggeri D, Tosi P, Zamagni E, Cellini C, Cavo M, Baccarani M, Tura S, Martinelli G.; ''Novel mutation and RNA splice variant of fibroblast growth factor receptor 3 in multiple myeloma patients at diagnosis.''; PubMed Europe PMC Scholia
  35. Dailey L, Ambrosetti D, Mansukhani A, Basilico C.; ''Mechanisms underlying differential responses to FGF signaling.''; PubMed Europe PMC Scholia
  36. 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
  37. Roskoski R.; ''RAF protein-serine/threonine kinases: structure and regulation.''; PubMed Europe PMC Scholia
  38. Mohammadi M, Olsen SK, Ibrahimi OA.; ''Structural basis for fibroblast growth factor receptor activation.''; PubMed Europe PMC Scholia
  39. 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
  40. 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
  41. Yuan L, Liu ZH, Lin ZR, Xu LH, Zhong Q, Zeng MS.; ''Recurrent FGFR3-TACC3 fusion gene in nasopharyngeal carcinoma.''; PubMed Europe PMC Scholia
  42. 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
  43. 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
  44. Goriely A, Hansen RM, Taylor IB, Olesen IA, Jacobsen GK, McGowan SJ, Pfeifer SP, McVean GA, Rajpert-De Meyts E, Wilkie AO.; ''Activating mutations in FGFR3 and HRAS reveal a shared genetic origin for congenital disorders and testicular tumors.''; PubMed Europe PMC Scholia
  45. 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
  46. Schlessinger J.; ''Common and distinct elements in cellular signaling via EGF and FGF receptors.''; PubMed Europe PMC Scholia
  47. 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
  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. d'Avis PY, Robertson SC, Meyer AN, Bardwell WM, Webster MK, Donoghue DJ.; ''Constitutive activation of fibroblast growth factor receptor 3 by mutations responsible for the lethal skeletal dysplasia thanatophoric dysplasia type I.''; PubMed Europe PMC Scholia
  50. 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
  51. Burgess SG, Peset I, Joseph N, Cavazza T, Vernos I, Pfuhl M, Gergely F, Bayliss R.; ''Aurora-A-Dependent Control of TACC3 Influences the Rate of Mitotic Spindle Assembly.''; PubMed Europe PMC Scholia
  52. Cargnello M, Roux PP.; ''Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases.''; PubMed Europe PMC Scholia
  53. 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
  54. 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
  55. Li E, You M, Hristova K.; ''FGFR3 dimer stabilization due to a single amino acid pathogenic mutation.''; PubMed Europe PMC Scholia
  56. Wesche J, Haglund K, Haugsten EM.; ''Fibroblast growth factors and their receptors in cancer.''; PubMed Europe PMC Scholia
  57. 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
  58. Shotelersuk V, Ittiwut C, Shotelersuk K, Triratanachat S, Poovorawan Y, Mutirangura A.; ''Fibroblast growth factor receptor 3 S249C mutation in virus associated squamous cell carcinomas.''; PubMed Europe PMC Scholia
  59. 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
  60. Roskoski R.; ''ERK1/2 MAP kinases: structure, function, and regulation.''; PubMed Europe PMC Scholia
  61. Trudel S, Ely S, Farooqi Y, Affer M, Robbiani DF, Chesi M, Bergsagel PL.; ''Inhibition of fibroblast growth factor receptor 3 induces differentiation and apoptosis in t(4;14) myeloma.''; PubMed Europe PMC Scholia
  62. 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
  63. 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
  64. 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
  65. 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
  66. 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
  67. Ronchetti D, Greco A, Compasso S, Colombo G, Dell'Era P, Otsuki T, Lombardi L, Neri A.; ''Deregulated FGFR3 mutants in multiple myeloma cell lines with t(4;14): comparative analysis of Y373C, K650E and the novel G384D mutations.''; PubMed Europe PMC Scholia
  68. 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
  69. 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
  70. 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
  71. Hernández S, de Muga S, Agell L, Juanpere N, Esgueva R, Lorente JA, Mojal S, Serrano S, Lloreta J.; ''FGFR3 mutations in prostate cancer: association with low-grade tumors.''; PubMed Europe PMC Scholia
  72. Bellus GA, Spector EB, Speiser PW, Weaver CA, Garber AT, Bryke CR, Israel J, Rosengren SS, Webster MK, Donoghue DJ, Francomano CA.; ''Distinct missense mutations of the FGFR3 lys650 codon modulate receptor kinase activation and the severity of the skeletal dysplasia phenotype.''; PubMed Europe PMC Scholia
  73. 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
  74. Roskoski R.; ''MEK1/2 dual-specificity protein kinases: structure and regulation.''; PubMed Europe PMC Scholia
  75. 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
  76. Trudel S, Li ZH, Wei E, Wiesmann M, Chang H, Chen C, Reece D, Heise C, Stewart AK.; ''CHIR-258, a novel, multitargeted tyrosine kinase inhibitor for the potential treatment of t(4;14) multiple myeloma.''; PubMed Europe PMC Scholia
  77. 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
  78. 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
  79. 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
  80. van Rhijn BW, van Tilborg AA, Lurkin I, Bonaventure J, de Vries A, Thiery JP, van der Kwast TH, Zwarthoff EC, Radvanyi F.; ''Novel fibroblast growth factor receptor 3 (FGFR3) mutations in bladder cancer previously identified in non-lethal skeletal disorders.''; PubMed Europe PMC Scholia
  81. Carter EP, Fearon AE, Grose RP.; ''Careless talk costs lives: fibroblast growth factor receptor signalling and the consequences of pathway malfunction.''; PubMed Europe PMC Scholia
  82. Chesi M, Nardini E, Brents LA, Schröck E, Ried T, Kuehl WM, Bergsagel PL.; ''Frequent translocation t(4;14)(p16.3;q32.3) in multiple myeloma is associated with increased expression and activating mutations of fibroblast growth factor receptor 3.''; PubMed Europe PMC Scholia
  83. Meyers GA, Orlow SJ, Munro IR, Przylepa KA, Jabs EW.; ''Fibroblast growth factor receptor 3 (FGFR3) transmembrane mutation in Crouzon syndrome with acanthosis nigricans.''; PubMed Europe PMC Scholia
  84. Otsuki T, Yamada O, Yata K, Sakaguchi H, Kurebayashi J, Nakazawa N, Taniwaki M, Yawata Y, Ueki A.; ''Expression of fibroblast growth factor and FGF-receptor family genes in human myeloma cells, including lines possessing t(4;14)(q16.3;q32. 3) and FGFR3 translocation.''; PubMed Europe PMC Scholia
  85. Webster MK, Donoghue DJ.; ''FGFR activation in skeletal disorders: too much of a good thing.''; PubMed Europe PMC Scholia
  86. 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
  87. 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
  88. Paterson JL, Li Z, Wen XY, Masih-Khan E, Chang H, Pollett JB, Trudel S, Stewart AK.; ''Preclinical studies of fibroblast growth factor receptor 3 as a therapeutic target in multiple myeloma.''; PubMed Europe PMC Scholia
  89. Mohi MG, Neel BG.; ''The role of Shp2 (PTPN11) in cancer.''; PubMed Europe PMC Scholia
  90. Wong ES, Lim J, Low BC, Chen Q, Guy GR.; ''Evidence for direct interaction between Sprouty and Cbl.''; PubMed Europe PMC Scholia
  91. Carneiro BA, Elvin JA, Kamath SD, Ali SM, Paintal AS, Restrepo A, Berry E, Giles FJ, Johnson ML.; ''FGFR3-TACC3: A novel gene fusion in cervical cancer.''; PubMed Europe PMC Scholia
  92. Kyriakis JM, Avruch J.; ''Mammalian MAPK signal transduction pathways activated by stress and inflammation: a 10-year update.''; PubMed Europe PMC Scholia
  93. 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
  94. 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
  95. 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
  96. Tavormina PL, Shiang R, Thompson LM, Zhu YZ, Wilkin DJ, Lachman RS, Wilcox WR, Rimoin DL, Cohn DH, Wasmuth JJ.; ''Thanatophoric dysplasia (types I and II) caused by distinct mutations in fibroblast growth factor receptor 3.''; PubMed Europe PMC Scholia
  97. 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
  98. Cappellen D, De Oliveira C, Ricol D, de Medina S, Bourdin J, Sastre-Garau X, Chopin D, Thiery JP, Radvanyi F.; ''Frequent activating mutations of FGFR3 in human bladder and cervix carcinomas.''; PubMed Europe PMC Scholia
  99. 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
  100. Naski MC, Wang Q, Xu J, Ornitz DM.; ''Graded activation of fibroblast growth factor receptor 3 by mutations causing achondroplasia and thanatophoric dysplasia.''; PubMed Europe PMC Scholia
  101. 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
  102. 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
  103. 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
  104. Parker BC, Annala MJ, Cogdell DE, Granberg KJ, Sun Y, Ji P, Li X, Gumin J, Zheng H, Hu L, Yli-Harja O, Haapasalo H, Visakorpi T, Liu X, Liu CG, Sawaya R, Fuller GN, Chen K, Lang FF, Nykter M, Zhang W.; ''The tumorigenic FGFR3-TACC3 gene fusion escapes miR-99a regulation in glioblastoma.''; PubMed Europe PMC Scholia
  105. 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
  106. 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
  107. Qing J, Du X, Chen Y, Chan P, Li H, Wu P, Marsters S, Stawicki S, Tien J, Totpal K, Ross S, Stinson S, Dornan D, French D, Wang QR, Stephan JP, Wu Y, Wiesmann C, Ashkenazi A.; ''Antibody-based targeting of FGFR3 in bladder carcinoma and t(4;14)-positive multiple myeloma in mice.''; PubMed Europe PMC Scholia
  108. 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
  109. 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
  110. McKay MM, Morrison DK.; ''Integrating signals from RTKs to ERK/MAPK.''; PubMed Europe PMC Scholia
  111. Chesi M, Brents LA, Ely SA, Bais C, Robbiani DF, Mesri EA, Kuehl WM, Bergsagel PL.; ''Activated fibroblast growth factor receptor 3 is an oncogene that contributes to tumor progression in multiple myeloma.''; PubMed Europe PMC Scholia
  112. 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
  113. Rousseau F, el Ghouzzi V, Delezoide AL, Legeai-Mallet L, Le Merrer M, Munnich A, Bonaventure J.; ''Missense FGFR3 mutations create cysteine residues in thanatophoric dwarfism type I (TD1).''; PubMed Europe PMC Scholia
  114. 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
  115. Roberts PJ, Der CJ.; ''Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer.''; PubMed Europe PMC Scholia
  116. Williams SV, Hurst CD, Knowles MA.; ''Oncogenic FGFR3 gene fusions in bladder cancer.''; PubMed Europe PMC Scholia
  117. Avet-Loiseau H, Li JY, Facon T, Brigaudeau C, Morineau N, Maloisel F, Rapp MJ, Talmant P, Trimoreau F, Jaccard A, Harousseau JL, Bataille R.; ''High incidence of translocations t(11;14)(q13;q32) and t(4;14)(p16;q32) in patients with plasma cell malignancies.''; PubMed Europe PMC Scholia
  118. 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
  119. 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
  120. Burke D, Wilkes D, Blundell TL, Malcolm S.; ''Fibroblast growth factor receptors: lessons from the genes.''; PubMed Europe PMC Scholia
  121. 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
  122. 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
  123. Grand EK, Chase AJ, Heath C, Rahemtulla A, Cross NC.; ''Targeting FGFR3 in multiple myeloma: inhibition of t(4;14)-positive cells by SU5402 and PD173074.''; PubMed Europe PMC Scholia
  124. Cantwell-Dorris ER, O'Leary JJ, Sheils OM.; ''BRAFV600E: implications for carcinogenesis and molecular therapy.''; PubMed Europe PMC Scholia
  125. 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
  126. Reardon W, Wilkes D, Rutland P, Pulleyn LJ, Malcolm S, Dean JC, Evans RD, Jones BM, Hayward R, Hall CM, Nevin NC, Baraister M, Winter RM.; ''Craniosynostosis associated with FGFR3 pro250arg mutation results in a range of clinical presentations including unisutural sporadic craniosynostosis.''; PubMed Europe PMC Scholia
  127. 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
  128. 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
  129. Eswarakumar VP, Lax I, Schlessinger J.; ''Cellular signaling by fibroblast growth factor receptors.''; PubMed Europe PMC Scholia
  130. Rousseau F, Saugier P, Le Merrer M, Munnich A, Delezoide AL, Maroteaux P, Bonaventure J, Narcy F, Sanak M.; ''Stop codon FGFR3 mutations in thanatophoric dwarfism type 1.''; PubMed Europe PMC Scholia
  131. Bellus GA, Gaudenz K, Zackai EH, Clarke LA, Szabo J, Francomano CA, Muenke M.; ''Identical mutations in three different fibroblast growth factor receptor genes in autosomal dominant craniosynostosis syndromes.''; PubMed Europe PMC Scholia
  132. Webster MK, D'Avis PY, Robertson SC, Donoghue DJ.; ''Profound ligand-independent kinase activation of fibroblast growth factor receptor 3 by the activation loop mutation responsible for a lethal skeletal dysplasia, thanatophoric dysplasia type II.''; PubMed Europe PMC Scholia
  133. Wang JK, Gao G, Goldfarb M.; ''Fibroblast growth factor receptors have different signaling and mitogenic potentials.''; PubMed Europe PMC Scholia
  134. 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
  135. Lemmon MA, Schlessinger J.; ''Cell signaling by receptor tyrosine kinases.''; PubMed Europe PMC Scholia
  136. Reddy PL, Grewal RP.; ''The G1138A mutation rate in the fibroblast growth factor receptor 3 (FGFR3) gene is increased in cells carrying the t (4; 14) translocation.''; PubMed Europe PMC Scholia
  137. Li Z, Zhu YX, Plowright EE, Bergsagel PL, Chesi M, Patterson B, Hawley TS, Hawley RG, Stewart AK.; ''The myeloma-associated oncogene fibroblast growth factor receptor 3 is transforming in hematopoietic cells.''; PubMed Europe PMC Scholia
  138. Greenman C, Stephens P, Smith R, Dalgliesh GL, Hunter C, Bignell G, Davies H, Teague J, Butler A, Stevens C, Edkins S, O'Meara S, Vastrik I, Schmidt EE, Avis T, Barthorpe S, Bhamra G, Buck G, Choudhury B, Clements J, Cole J, Dicks E, Forbes S, Gray K, Halliday K, Harrison R, Hills K, Hinton J, Jenkinson A, Jones D, Menzies A, Mironenko T, Perry J, Raine K, Richardson D, Shepherd R, Small A, Tofts C, Varian J, Webb T, West S, Widaa S, Yates A, Cahill DP, Louis DN, Goldstraw P, Nicholson AG, Brasseur F, Looijenga L, Weber BL, Chiew YE, DeFazio A, Greaves MF, Green AR, Campbell P, Birney E, Easton DF, Chenevix-Trench G, Tan MH, Khoo SK, Teh BT, Yuen ST, Leung SY, Wooster R, Futreal PA, Stratton MR.; ''Patterns of somatic mutation in human cancer genomes.''; PubMed Europe PMC Scholia
  139. Adar R, Monsonego-Ornan E, David P, Yayon A.; ''Differential activation of cysteine-substitution mutants of fibroblast growth factor receptor 3 is determined by cysteine localization.''; PubMed Europe PMC Scholia
  140. 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
  141. Cseh B, Doma E, Baccarini M.; ''"RAF" neighborhood: protein-protein interaction in the Raf/Mek/Erk pathway.''; PubMed Europe PMC Scholia

History

View all...
CompareRevisionActionTimeUserComment
114753view16:24, 25 January 2021ReactomeTeamReactome version 75
113197view11:26, 2 November 2020ReactomeTeamReactome version 74
112422view15:36, 9 October 2020ReactomeTeamReactome version 73
101326view11:21, 1 November 2018ReactomeTeamreactome version 66
100864view20:54, 31 October 2018ReactomeTeamreactome version 65
100405view19:27, 31 October 2018ReactomeTeamreactome version 64
99953view16:12, 31 October 2018ReactomeTeamreactome version 63
99509view14:45, 31 October 2018ReactomeTeamreactome version 62 (2nd attempt)
94046view13:53, 16 August 2017ReactomeTeamreactome version 61
93671view11:30, 9 August 2017ReactomeTeamreactome version 61
87127view18:46, 18 July 2016EgonwOntology Term : 'signaling pathway' added !
86795view09:26, 11 July 2016ReactomeTeamreactome version 56
83307view10:43, 18 November 2015ReactomeTeamVersion54
81444view12:58, 21 August 2015ReactomeTeamNew pathway

External references

DataNodes

View all...
NameTypeDatabase referenceComment
ADPMetaboliteCHEBI:456216 (ChEBI)
ATPMetaboliteCHEBI:30616 (ChEBI)
AZD4547
Activated FGFR3:p-8T-FRS2ComplexR-HSA-5654301 (Reactome)
Activated FGFR3:p-FRS2:GRB2:GAB1:PI3KComplexR-HSA-5654195 (Reactome)
Activated FGFR3:p-FRS2:GRB2:GAB1:PIK3R1ComplexR-HSA-5654203 (Reactome)
Activated FGFR3:p-FRS2:p-PTPN11:GRB2:GAB1:PI3KComplexR-HSA-5654197 (Reactome)
Activated FGFR3:p-FRS2:p-PTPN11:GRB2:GAB1:PIK3R1ComplexR-HSA-5654199 (Reactome)
Activated FGFR3:p-FRS2:p-PTPN11:p-CBL:GRB2ComplexR-HSA-5654307 (Reactome)
Activated FGFR3:p-FRS2:p-PTPN11ComplexR-HSA-5654198 (Reactome)
Activated FGFR3:p-FRS2ComplexR-HSA-5654196 (Reactome)
Activated FGFR3:p-FRS3ComplexR-HSA-5654303 (Reactome)
Activated FGFR3:p-FRS:GRB2:SOS1ComplexR-HSA-5654308 (Reactome)
Activated FGFR3:p-FRS:PTPN11ComplexR-HSA-5654310 (Reactome)
Activated FGFR3:p-FRS:p-PTPN11ComplexR-HSA-5654312 (Reactome)
Activated FGFR3:p-FRSComplexR-HSA-5654305 (Reactome)
Activated FGFR3:pY-SHC1:GRB2:SOS1ComplexR-HSA-5654316 (Reactome)
Activated FGFR3:pY-SHC1ComplexR-HSA-5654314 (Reactome)
Activated FGFR3 cysteine mutantsComplexR-HSA-2012020 (Reactome)
Activated FGFR3

mutants with enhanced kinase

activity
ComplexR-HSA-2033358 (Reactome)
Activated FGFR3

t(4;14) translocation

mutants
ComplexR-HSA-2038377 (Reactome)
Activated FGFR3:FRS2ComplexR-HSA-5654180 (Reactome)
Activated FGFR3:FRS3ComplexR-HSA-5654297 (Reactome)
Activated FGFR3:SHC1ComplexR-HSA-5654300 (Reactome)
Activated FGFR3ComplexR-HSA-5654146 (Reactome)
Activated FGFR3b homodimerComplexR-HSA-190380 (Reactome)
Activated FGFR3c P250R mutantComplexR-HSA-2011989 (Reactome)
Activated FGFR3c homodimerComplexR-HSA-190389 (Reactome)
BRAF ProteinP15056 (Uniprot-TrEMBL)
BRAFProteinP15056 (Uniprot-TrEMBL)
CBL ProteinP22681 (Uniprot-TrEMBL)
CBLProteinP22681 (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.
FGF1 ProteinP05230 (Uniprot-TrEMBL)
FGF16 ProteinO43320 (Uniprot-TrEMBL)
FGF17-1 ProteinO60258-1 (Uniprot-TrEMBL)
FGF18 ProteinO76093 (Uniprot-TrEMBL)
FGF2(10-155) ProteinP09038 (Uniprot-TrEMBL)
FGF20 ProteinQ9NP95 (Uniprot-TrEMBL)
FGF23(25-251)ProteinQ9GZV9 (Uniprot-TrEMBL)
FGF4 ProteinP08620 (Uniprot-TrEMBL)
FGF5-1 ProteinP12034-1 (Uniprot-TrEMBL)
FGF8-1 ProteinP55075-1 (Uniprot-TrEMBL)
FGF9 ProteinP31371 (Uniprot-TrEMBL)
FGFR3 (4;14)

translocation

mutant dimers
ComplexR-HSA-2038375 (Reactome)
FGFR3 (4;14)

translocation

mutants
ComplexR-HSA-2038374 (Reactome)
FGFR3 (4;14) translocation mutant dimers R-HSA-2038375 (Reactome)
FGFR3 795fs*139 ProteinP22607 (Uniprot-TrEMBL)
FGFR3 A391E ProteinP22607 (Uniprot-TrEMBL)
FGFR3 G370C ProteinP22607 (Uniprot-TrEMBL)
FGFR3 G380R ProteinP22607 (Uniprot-TrEMBL)
FGFR3 G382D ProteinP22607 (Uniprot-TrEMBL)
FGFR3 K650E ProteinP22607 (Uniprot-TrEMBL)
FGFR3 K650M ProteinP22607 (Uniprot-TrEMBL)
FGFR3 K650N ProteinP22607 (Uniprot-TrEMBL)
FGFR3 K650Q ProteinP22607 (Uniprot-TrEMBL)
FGFR3 K650T ProteinP22607 (Uniprot-TrEMBL)
FGFR3 ProteinP22607 (Uniprot-TrEMBL)
FGFR3 R248C ProteinP22607 (Uniprot-TrEMBL)
FGFR3 S371C ProteinP22607 (Uniprot-TrEMBL)
FGFR3 Y373C ProteinP22607 (Uniprot-TrEMBL)
FGFR3 cysteine mutant dimersComplexR-HSA-2012041 (Reactome)
FGFR3 cysteine mutantsComplexR-HSA-2012043 (Reactome)
FGFR3 cysteine mutant dimers R-HSA-2012041 (Reactome)
FGFR3 fusion dimersComplexR-HSA-8853268 (Reactome)
FGFR3 fusionsComplexR-HSA-8853269 (Reactome)
FGFR3 mutant dimers:TKIsComplexR-HSA-2077417 (Reactome)
FGFR3 mutant dimersComplexR-HSA-2077412 (Reactome)
FGFR3 point mutant

dimers with enhanced kinase

activity
ComplexR-HSA-2033383 (Reactome)
FGFR3 point mutant dimers with enhanced kinase activity R-HSA-2033383 (Reactome)
FGFR3 point mutants

with enhanced

kinase activity
ComplexR-HSA-2018770 (Reactome)
FGFR3(23-758)-TACC3(549-838) fusion ProteinP22607 (Uniprot-TrEMBL)
FGFR3(23-758)-TACC3(649-838) fusion ProteinP22607 (Uniprot-TrEMBL)
FGFR3(23-760)-BAIAP2L1(18-522) fusion ProteinP22607 (Uniprot-TrEMBL)
FGFR3(23-760)-TACC3(612-838) fusion ProteinP22607 (Uniprot-TrEMBL)
FGFR3(23-760)-TACC3(647-838) fusion ProteinP22607 (Uniprot-TrEMBL)
FGFR3(23-793)-TACC3(143-838) fusion ProteinP22607 (Uniprot-TrEMBL)
FGFR3b G697C ProteinP22607-2 (Uniprot-TrEMBL) Note that residue G697C is numbered according to the FGFR3c isoform, (UniProt 22607-1) and actually corresponds to G699C in the FGFR3b isoform (Uniprot 22607-2).
FGFR3b ProteinP22607-2 (Uniprot-TrEMBL)
FGFR3b S249C ProteinP22607-2 (Uniprot-TrEMBL)
FGFR3b homodimer bound to FGFComplexR-HSA-190232 (Reactome)
FGFR3b-binding FGFsComplexR-HSA-189964 (Reactome)
FGFR3bProteinP22607-2 (Uniprot-TrEMBL)
FGFR3c ProteinP22607-1 (Uniprot-TrEMBL)
FGFR3c P250R ProteinP22607-1 (Uniprot-TrEMBL) Muenke Syndrome
FGFR3c P250R mutant dimer bound to FGFComplexR-HSA-2011952 (Reactome)
FGFR3c P250RProteinP22607-1 (Uniprot-TrEMBL) Muenke Syndrome
FGFR3c homodimer bound to FGFComplexR-HSA-190234 (Reactome)
FGFR3c-binding FGFsComplexR-HSA-189955 (Reactome)
FGFR3cProteinP22607-1 (Uniprot-TrEMBL)
FRS2 ProteinQ8WU20 (Uniprot-TrEMBL)
FRS2ProteinQ8WU20 (Uniprot-TrEMBL)
FRS3 ProteinO43559 (Uniprot-TrEMBL)
FRS3ProteinO43559 (Uniprot-TrEMBL)
GAB1 ProteinQ13480 (Uniprot-TrEMBL)
GALNT3ProteinQ14435 (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)
GTP MetaboliteCHEBI:15996 (ChEBI)
GTPMetaboliteCHEBI:15996 (ChEBI)
GalNAc-T178-FGF23(25-251) ProteinQ9GZV9 (Uniprot-TrEMBL)
GalNAc-T178-FGF23(25-251)ProteinQ9GZV9 (Uniprot-TrEMBL)
HS MetaboliteCHEBI:28815 (ChEBI)
HSMetaboliteCHEBI:28815 (ChEBI)
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:43474 (ChEBI)
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)
S-Farn-Me KRAS4B ProteinP01116-2 (Uniprot-TrEMBL)
S-Farn-Me PalmS NRAS ProteinP01111 (Uniprot-TrEMBL)
S-Farn-Me-2xPalmS HRAS ProteinP01112 (Uniprot-TrEMBL)
S-Farn-Me-PalmS KRAS4A ProteinP01116-1 (Uniprot-TrEMBL)
S111/S120 p-SPRY2:B-RAFComplexR-HSA-1295587 (Reactome)
SHC1-2 ProteinP29353-2 (Uniprot-TrEMBL)
SHC1-2,SHC1-3ComplexR-HSA-1169480 (Reactome) SHC1 isoforms p46 and p52 are found in B cells (Smit et al. 1994).
SHC1-3 ProteinP29353-3 (Uniprot-TrEMBL)
SOS1 ProteinQ07889 (Uniprot-TrEMBL)
SPRY2 ProteinO43597 (Uniprot-TrEMBL)
SPRY2:B-RAFComplexR-HSA-1295598 (Reactome)
SRC-1ProteinP12931-1 (Uniprot-TrEMBL)
Tyrosine kinase

inhibitors of FGFR3

mutants
ComplexR-ALL-2077416 (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)
UDP-GalNAcMetaboliteCHEBI:16650 (ChEBI)
UDPMetaboliteCHEBI:17659 (ChEBI)
Ub-(Y55/Y227)p-SPRY2ComplexR-HSA-1370875 (Reactome)
Ub-Activated FGFR3 complex:Ub-p-FRS2ComplexR-HSA-5654362 (Reactome)
Ub:Y55/Y227-pSPRY2:CBLComplexR-HSA-934572 (Reactome)
UbComplexR-HSA-113595 (Reactome)
Y55/Y227-pSPRY2:CBLComplexR-HSA-934576 (Reactome)
activated FGFR3:PLCG1ComplexR-HSA-5654226 (Reactome)
activated FGFR3:p-4Y-PLCG1ComplexR-HSA-5654156 (Reactome)
p-4Y FGFR3(23-758)-TACC3(549-838) fusion ProteinP22607 (Uniprot-TrEMBL)
p-4Y FGFR3(23-758)-TACC3(649-838) fusion ProteinP22607 (Uniprot-TrEMBL)
p-4Y FGFR3(23-760)-BAIAP2L1(18-522) fusion ProteinP22607 (Uniprot-TrEMBL)
p-4Y FGFR3(23-760)-TACC3(612-838) fusion ProteinP22607 (Uniprot-TrEMBL)
p-4Y FGFR3(23-760)-TACC3(647-838) fusion ProteinP22607 (Uniprot-TrEMBL)
p-4Y FGFR3(23-793)-TACC3(143-838) fusion ProteinP22607 (Uniprot-TrEMBL)
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-FGFR3 795fs*139 ProteinP22607 (Uniprot-TrEMBL)
p-6Y-FGFR3 A391E ProteinP22607 (Uniprot-TrEMBL)
p-6Y-FGFR3 G370C ProteinP22607 (Uniprot-TrEMBL)
p-6Y-FGFR3 G380R ProteinP22607 (Uniprot-TrEMBL)
p-6Y-FGFR3 G382D ProteinP22607 (Uniprot-TrEMBL)
p-6Y-FGFR3 K650E ProteinP22607 (Uniprot-TrEMBL)
p-6Y-FGFR3 K650M ProteinP22607 (Uniprot-TrEMBL)
p-6Y-FGFR3 K650N ProteinP22607 (Uniprot-TrEMBL)
p-6Y-FGFR3 K650Q ProteinP22607 (Uniprot-TrEMBL)
p-6Y-FGFR3 K650T ProteinP22607 (Uniprot-TrEMBL)
p-6Y-FGFR3 R248C ProteinP22607 (Uniprot-TrEMBL)
p-6Y-FGFR3 S371C ProteinP22607 (Uniprot-TrEMBL)
p-6Y-FGFR3 Y373C ProteinP22607 (Uniprot-TrEMBL)
p-6Y-FGFR3b G697C ProteinP22607-2 (Uniprot-TrEMBL) Note that residue G697C is numbered according to the FGFR3c isoform, (UniProt 22607-1) and actually corresponds to G699C in the FGFR3b isoform (Uniprot 22607-2).
p-6Y-FGFR3b ProteinP22607-2 (Uniprot-TrEMBL)
p-6Y-FGFR3b S249C mutant ProteinP22607-2 (Uniprot-TrEMBL)
p-6Y-FGFR3c ProteinP22607-1 (Uniprot-TrEMBL)
p-6Y-FRS2 ProteinQ8WU20 (Uniprot-TrEMBL)
p-8T-FRS2 ProteinQ8WU20 (Uniprot-TrEMBL)
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-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 FGFR3 fusion dimersComplexR-HSA-8853232 (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)
p21 RAS:GDPComplexR-HSA-109796 (Reactome)
p21 RAS:GTPComplexR-HSA-109783 (Reactome)
p6Y-FGFR3c P250R ProteinP22607-1 (Uniprot-TrEMBL) Muenke Syndrome
pY-FGFR3 ProteinP22607 (Uniprot-TrEMBL)

Annotated Interactions

View all...
SourceTargetTypeDatabase referenceComment
ADPArrowR-HSA-1295609 (Reactome)
ADPArrowR-HSA-190385 (Reactome)
ADPArrowR-HSA-190388 (Reactome)
ADPArrowR-HSA-2012073 (Reactome)
ADPArrowR-HSA-2012082 (Reactome)
ADPArrowR-HSA-2033485 (Reactome)
ADPArrowR-HSA-2038387 (Reactome)
ADPArrowR-HSA-5654222 (Reactome)
ADPArrowR-HSA-5654408 (Reactome)
ADPArrowR-HSA-5654565 (Reactome)
ADPArrowR-HSA-5654628 (Reactome)
ADPArrowR-HSA-5654631 (Reactome)
ADPArrowR-HSA-5654634 (Reactome)
ADPArrowR-HSA-5654705 (Reactome)
ADPArrowR-HSA-5654709 (Reactome)
ADPArrowR-HSA-8853309 (Reactome)
ADPArrowR-HSA-934559 (Reactome)
ATPR-HSA-1295609 (Reactome)
ATPR-HSA-190385 (Reactome)
ATPR-HSA-190388 (Reactome)
ATPR-HSA-2012073 (Reactome)
ATPR-HSA-2012082 (Reactome)
ATPR-HSA-2033485 (Reactome)
ATPR-HSA-2038387 (Reactome)
ATPR-HSA-5654222 (Reactome)
ATPR-HSA-5654408 (Reactome)
ATPR-HSA-5654565 (Reactome)
ATPR-HSA-5654628 (Reactome)
ATPR-HSA-5654631 (Reactome)
ATPR-HSA-5654634 (Reactome)
ATPR-HSA-5654705 (Reactome)
ATPR-HSA-5654709 (Reactome)
ATPR-HSA-8853309 (Reactome)
ATPR-HSA-934559 (Reactome)
Activated FGFR3:p-8T-FRS2ArrowR-HSA-5654565 (Reactome)
Activated FGFR3:p-8T-FRS2TBarR-HSA-5654408 (Reactome)
Activated FGFR3:p-FRS2:GRB2:GAB1:PI3KArrowR-HSA-5654640 (Reactome)
Activated FGFR3:p-FRS2:GRB2:GAB1:PI3Kmim-catalysisR-HSA-5654705 (Reactome)
Activated FGFR3:p-FRS2:GRB2:GAB1:PIK3R1ArrowR-HSA-5654637 (Reactome)
Activated FGFR3:p-FRS2:GRB2:GAB1:PIK3R1R-HSA-5654640 (Reactome)
Activated FGFR3:p-FRS2:p-PTPN11:GRB2:GAB1:PI3KArrowR-HSA-5654643 (Reactome)
Activated FGFR3:p-FRS2:p-PTPN11:GRB2:GAB1:PI3Kmim-catalysisR-HSA-5654709 (Reactome)
Activated FGFR3:p-FRS2:p-PTPN11:GRB2:GAB1:PIK3R1ArrowR-HSA-5654641 (Reactome)
Activated FGFR3:p-FRS2:p-PTPN11:GRB2:GAB1:PIK3R1R-HSA-5654643 (Reactome)
Activated FGFR3:p-FRS2:p-PTPN11:p-CBL:GRB2ArrowR-HSA-5654730 (Reactome)
Activated FGFR3:p-FRS2:p-PTPN11:p-CBL:GRB2R-HSA-5654679 (Reactome)
Activated FGFR3:p-FRS2:p-PTPN11:p-CBL:GRB2mim-catalysisR-HSA-5654679 (Reactome)
Activated FGFR3:p-FRS2:p-PTPN11R-HSA-5654641 (Reactome)
Activated FGFR3:p-FRS2:p-PTPN11R-HSA-5654730 (Reactome)
Activated FGFR3:p-FRS2ArrowR-HSA-5654408 (Reactome)
Activated FGFR3:p-FRS2R-HSA-5654637 (Reactome)
Activated FGFR3:p-FRS3ArrowR-HSA-5654628 (Reactome)
Activated FGFR3:p-FRS:GRB2:SOS1ArrowR-HSA-5654416 (Reactome)
Activated FGFR3:p-FRS:GRB2:SOS1mim-catalysisR-HSA-5654413 (Reactome)
Activated FGFR3:p-FRS:PTPN11ArrowR-HSA-5654633 (Reactome)
Activated FGFR3:p-FRS:PTPN11R-HSA-5654631 (Reactome)
Activated FGFR3:p-FRS:PTPN11mim-catalysisR-HSA-5654631 (Reactome)
Activated FGFR3:p-FRS:p-PTPN11ArrowR-HSA-5654631 (Reactome)
Activated FGFR3:p-FRS:p-PTPN11ArrowR-HSA-8941628 (Reactome)
Activated FGFR3:p-FRSR-HSA-5654416 (Reactome)
Activated FGFR3:p-FRSR-HSA-5654633 (Reactome)
Activated FGFR3:pY-SHC1:GRB2:SOS1ArrowR-HSA-5654646 (Reactome)
Activated FGFR3:pY-SHC1:GRB2:SOS1mim-catalysisR-HSA-5654647 (Reactome)
Activated FGFR3:pY-SHC1ArrowR-HSA-5654634 (Reactome)
Activated FGFR3:pY-SHC1R-HSA-5654646 (Reactome)
Activated FGFR3 cysteine mutantsArrowR-HSA-2012082 (Reactome)
Activated FGFR3

mutants with enhanced kinase

activity
ArrowR-HSA-2033485 (Reactome)
Activated FGFR3

t(4;14) translocation

mutants
ArrowR-HSA-2038387 (Reactome)
Activated FGFR3:FRS2ArrowR-HSA-5654409 (Reactome)
Activated FGFR3:FRS2R-HSA-5654408 (Reactome)
Activated FGFR3:FRS2R-HSA-5654565 (Reactome)
Activated FGFR3:FRS2mim-catalysisR-HSA-5654408 (Reactome)
Activated FGFR3:FRS3ArrowR-HSA-5654623 (Reactome)
Activated FGFR3:FRS3R-HSA-5654628 (Reactome)
Activated FGFR3:FRS3mim-catalysisR-HSA-5654628 (Reactome)
Activated FGFR3:SHC1ArrowR-HSA-5654625 (Reactome)
Activated FGFR3:SHC1R-HSA-5654634 (Reactome)
Activated FGFR3:SHC1mim-catalysisR-HSA-5654634 (Reactome)
Activated FGFR3ArrowR-HSA-5654148 (Reactome)
Activated FGFR3R-HSA-5654224 (Reactome)
Activated FGFR3R-HSA-5654409 (Reactome)
Activated FGFR3R-HSA-5654623 (Reactome)
Activated FGFR3R-HSA-5654625 (Reactome)
Activated FGFR3b homodimerArrowR-HSA-190385 (Reactome)
Activated FGFR3c P250R mutantArrowR-HSA-2012073 (Reactome)
Activated FGFR3c homodimerArrowR-HSA-190388 (Reactome)
BRAFArrowR-HSA-1295604 (Reactome)
CBLArrowR-HSA-1295621 (Reactome)
CBLR-HSA-1295622 (Reactome)
FGF23(25-251)R-HSA-8851619 (Reactome)
FGFR3 (4;14)

translocation

mutant dimers
ArrowR-HSA-2038386 (Reactome)
FGFR3 (4;14)

translocation

mutant dimers
R-HSA-2038387 (Reactome)
FGFR3 (4;14)

translocation

mutant dimers
mim-catalysisR-HSA-2038387 (Reactome)
FGFR3 (4;14)

translocation

mutants
R-HSA-2038386 (Reactome)
FGFR3 cysteine mutant dimersArrowR-HSA-2012084 (Reactome)
FGFR3 cysteine mutant dimersR-HSA-2012082 (Reactome)
FGFR3 cysteine mutant dimersmim-catalysisR-HSA-2012082 (Reactome)
FGFR3 cysteine mutantsR-HSA-2012084 (Reactome)
FGFR3 fusion dimersArrowR-HSA-8853317 (Reactome)
FGFR3 fusion dimersR-HSA-8853309 (Reactome)
FGFR3 fusion dimersmim-catalysisR-HSA-8853309 (Reactome)
FGFR3 fusionsR-HSA-8853317 (Reactome)
FGFR3 mutant dimers:TKIsArrowR-HSA-2077420 (Reactome)
FGFR3 mutant dimersR-HSA-2077420 (Reactome)
FGFR3 point mutant

dimers with enhanced kinase

activity
ArrowR-HSA-2033476 (Reactome)
FGFR3 point mutant

dimers with enhanced kinase

activity
R-HSA-2033485 (Reactome)
FGFR3 point mutant

dimers with enhanced kinase

activity
mim-catalysisR-HSA-2033485 (Reactome)
FGFR3 point mutants

with enhanced

kinase activity
R-HSA-2033476 (Reactome)
FGFR3b homodimer bound to FGFArrowR-HSA-190263 (Reactome)
FGFR3b homodimer bound to FGFR-HSA-190385 (Reactome)
FGFR3b homodimer bound to FGFmim-catalysisR-HSA-190385 (Reactome)
FGFR3b-binding FGFsR-HSA-190263 (Reactome)
FGFR3bR-HSA-190263 (Reactome)
FGFR3c P250R mutant dimer bound to FGFArrowR-HSA-2012074 (Reactome)
FGFR3c P250R mutant dimer bound to FGFR-HSA-2012073 (Reactome)
FGFR3c P250R mutant dimer bound to FGFmim-catalysisR-HSA-2012073 (Reactome)
FGFR3c P250RR-HSA-2012074 (Reactome)
FGFR3c homodimer bound to FGFArrowR-HSA-190261 (Reactome)
FGFR3c homodimer bound to FGFR-HSA-190388 (Reactome)
FGFR3c homodimer bound to FGFmim-catalysisR-HSA-190388 (Reactome)
FGFR3c-binding FGFsR-HSA-190261 (Reactome)
FGFR3c-binding FGFsR-HSA-2012074 (Reactome)
FGFR3cR-HSA-190261 (Reactome)
FRS2R-HSA-5654409 (Reactome)
FRS3R-HSA-5654623 (Reactome)
GALNT3mim-catalysisR-HSA-8851619 (Reactome)
GDPArrowR-HSA-5654413 (Reactome)
GDPArrowR-HSA-5654647 (Reactome)
GDPArrowR-HSA-8941628 (Reactome)
GRB2-1:SOS1R-HSA-5654416 (Reactome)
GRB2-1:SOS1R-HSA-5654646 (Reactome)
GRB2-1ArrowR-HSA-1549564 (Reactome)
GRB2-1R-HSA-1295613 (Reactome)
GRB2:GAB1:PIK3R1ArrowR-HSA-177931 (Reactome)
GRB2:GAB1:PIK3R1R-HSA-5654637 (Reactome)
GRB2:GAB1:PIK3R1R-HSA-5654641 (Reactome)
GRB2:GAB1R-HSA-177931 (Reactome)
GTPR-HSA-5654413 (Reactome)
GTPR-HSA-5654647 (Reactome)
GTPR-HSA-8941628 (Reactome)
GalNAc-T178-FGF23(25-251)ArrowR-HSA-8851619 (Reactome)
GalNAc-T178-FGF23(25-251)ArrowR-HSA-8851632 (Reactome)
GalNAc-T178-FGF23(25-251)R-HSA-8851632 (Reactome)
HSArrowR-HSA-190261 (Reactome)
HSArrowR-HSA-190263 (Reactome)
HSR-HSA-190261 (Reactome)
HSR-HSA-190263 (Reactome)
HSR-HSA-2012074 (Reactome)
PI(3,4,5)P3ArrowR-HSA-5654705 (Reactome)
PI(3,4,5)P3ArrowR-HSA-5654709 (Reactome)
PI(3,4,5)P3R-HSA-5654224 (Reactome)
PI(4,5)P2R-HSA-5654705 (Reactome)
PI(4,5)P2R-HSA-5654709 (Reactome)
PIK3CAR-HSA-5654640 (Reactome)
PIK3CAR-HSA-5654643 (Reactome)
PIK3R1R-HSA-177931 (Reactome)
PLCG1R-HSA-5654224 (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-5654633 (Reactome)
PTPN11mim-catalysisR-HSA-1549564 (Reactome)
PiArrowR-HSA-1295632 (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-190261 (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-190263 (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-190385 (Reactome) The intrinsic protein tyrosine kinase activity of activated FGF receptor 3 catalyzes multiple phosphorylation events, creating a number of binding sites on its cytoplasmic tail for membrane bound docking proteins to gather intracellular signaling mediators. Based on sequence alignment, FGFR3 contains 6 of the 8 cytoplasmic tyrosine residues identified in FGFR1. Mutagenesis studies highlight the importance of tyrosine residue 724 in signaling mediated by FGFR3, including transformation, PPTN11/SHP2 phosphorylation, and activation of MAPK, PI3K and STAT pathways. These studies also identified a role for the PLCG1-binding tyrosine residue, Y760, in STAT activation, and a potential role for tyrosine 770 as a negative regulator of FGFR3 signaling.
R-HSA-190388 (Reactome) The intrinsic protein tyrosine kinase activity of activated FGF receptor 3 catalyzes multiple phosphorylation events, creating a number of binding sites on its cytoplasmic tail for membrane bound docking proteins to gather intracellular signaling mediators. Based on sequence alignment, FGFR3 contains 6 of the 8 cytoplasmic tyrosine residues identified in FGFR1. Mutagenesis studies highlight the importance of tyrosine residue 724 in signaling mediated by FGFR3, including transformation, PPTN11/SHP2 phosphorylation, and activation of MAPK, PI3K and STAT pathways. These studies also identified a role for the PLCG1-binding tyrosine residue, Y760, in STAT activation, and a potential role for tyrosine 770 as a negative regulator of FGFR3 signaling.
R-HSA-2012073 (Reactome) After high-affinity ligand binding, FGFR3 P250R is believed to undergo trans-autophosphorylation in a manner analogous to the wild-type receptor, although this remains to be experimentally validated (Ibrahimi, 2004a).
R-HSA-2012074 (Reactome) FGFR3 P350R is associated with the development of Muenke syndrome, a milder craniosynostotic condition than Apert Syndrome (Bellus, 1996; Reardon, 1997). This mutation, which falls in the highly conserved Ser-Pro dipeptide in the IgII-IgIII linker, has been shown to increase the affinity of the receptor for its natural ligands, particularly for FGF9 (Ibrahimi, 2004a), without expanding the ligand-binding range of the receptor. This difference, compared to the paralogous FGFR2 S252W and P253R mutations, which bind an expanded range of ligands, is thought to account for the milder phenotype of Muenke Syndrome (Yu, 2000; Ibrahimi, 2004a, b).
R-HSA-2012082 (Reactome) Although each of FGFR3 R248C, S249C, G370C, S371C and Y373C have been shown to undergo ligand-independent dimerization and receptor autophosphorylation, there is conflicting evidence about which mutants also show increased phosphorylation upon ligand stimulation. Mutants showed elevated levels of ligand-independent MAPK pathway activation and supported expression of an in vivo reporter gene (d'Avis, 1998; Adar, 2009).
R-HSA-2012084 (Reactome) Activating mutations in FGFR3 that introduce a mutant cysteine residue to the Ig2-Ig3 linker domain or the extracellular juxtamembrane region have been identified in the lethal neonatal disorder thanatophoric dysplasia (Tavormina, 1995a, b; Rousseau, 1996; reviewed in Webster and Donoghue, 1997; Burke, 1998). The presence of the mutant cysteine residue causes ligand-independent dimerization of the receptor through Cys-mediated intramolecular disulphide bonds and leads to increased biological signaling without changing the intrinsic kinase activity of the receptor (d'Avis, 1998; Adar, 2002). More recently, the same mutations, arising somatically, have been identified in a range of cancers including bladder, prostrate and cervical cancer, as well as in multiple myeloma and head and neck squamous cell carcinoma (reviewed in Wesche, 2011).
R-HSA-2033476 (Reactome) Activating point mutations G380R, N540K and K650E/M/N/Q in FGFR3 have been identified in achondroplasia, hypochondroplasia and thanatophoric dysplasia I and II (reviewed in Webster and Donoghue, 1997, Burke, 1998). These mutants, which occur in the transmembrane and the kinase domain, have been shown to undergo ligand-independent dimerization and autophosphorylation when transfected into NIH 3T3 cells, although the extent of constitutive activation varies depending on the precise mutation (Webster and Donoghue, 1996; Webster, 1996; Naski, 1996; Bellus, 2000). In addition, some of the mutants retain the ability to respond to exogenous ligand, while others appear to be completely ligand-independent (Naski, 1996; Goriely, 2009). Interestingly, the extent of kinase activation correlates with the severity of the resulting condition, with the K650M and E mutations associated with thanatophoric dysplasia showing the higher levels of kinase activity than the G380R mutation associated with achondroplasia (Naski, 1996; Bellus, 2000; Goriely, 2009). More recently, these same mutations, along with G382D, N540S, K650T, and G97C, have also been identified in a range of cancers, most notably in bladder cancer and multiple myeloma (Zhang, 2005; Ronchetti, 2001; van Rhijn, 2002; Lindgren, 2006; reveiewed in Wesche, 2011; Greulich and Pollock, 2011).
R-HSA-2033485 (Reactome) Activated point mutants in the transmembrane and kinase domains of FGFR3 have been shown to undergo constitutive autophosphorylation in a ligand-independent manner (Naski, 1996; Webster, 1996 and Donoghue, 1996; Webster, 1996; Bellus, 2000; Goriely, 2009). Some of the point mutants, including K650E and G380R, may also be able to further respond after exposure to ligand (Naski, 1996). Dimerization and activation of the FGFR3 transmembrane mutants is thought to occur via the formation of non-native hydrogen bonds that promote intermolecular interactions (Webster and Donoghue 1996), while the kinase domain mutants activate phosphorylation by mimicking conformational changes in the activation loop (Webster, 1996). Mutants with enhanced kinase activity appear to be activated to differing extents that, for the most part, correlate with the severity of the disease phenotype (Webster, 1996; Bellus, 2000; Goriely, 2009), although the results of in vitro kinase assays with immunoprecipitated proteins do not fully recapitulate the pathological consequences of the mutation (Goriely, 2009). K650E has also been shown to transform NIH 3T3 cells (Chesi, 2001).


R-HSA-2038386 (Reactome) ~15% of multiple myelomas contain translocations that put the FGFR3 gene under the control of the strong IGH locus (Chesi, 1997; Avet-Loiseau, 1998). This translocation results in the overexpresssion of FGFR3 (Chesi, 1997), which leads to aberrant signaling in either a ligand-dependent (Otsuki, 1999; Qing, 2009) or independent fashion (Chesi, 2001). Overexpression of WT FGFR3 results in a low level of FGF-independent MAPK activation, suggesting that overexpression can lead to ligand-independent dimerization; however this response is more pronounced after ligand-stimulation (Chesi, 2001; Qing, 2009). ~5% of multiple myelomas with FGFR3 translocations also contain coding sequence activating mutations (Chesi, 1997; Avet-Loiseau, 1998). These mutations (R248C, Y373C, K650E and K650M) mimic activating mutations seen in bone development disoders, are believed to arise later in tumor progression than the translocation event and contribute to ligand-independent signaling (Chesi, 1997; Chesi, 2001; Li, 2001; Ronchetti, 2001).
R-HSA-2038387 (Reactome) Overexpression of WT FGFR3 is weakly transforming when expressed in a mouse haematopoietic model, while expression of translocated FGFR3 carrying activating point mutations in the coding sequence is strongly transforming in both NIH 3T3 cells and the haematopoietic mouse model (Chesi, 2001; Ronchetti, 2001; Li, 2001). Activating mutations in FGFR3 are mutually exclusive with activating Ras mutations, and focus formation in NIH 3T3 cells is inhibited by cotransfection with dominant negative forms of ras or raf, suggesting that activation of the MAPK pathway is the primary oncogenic event in translocated myeloma lines (Chesi, 2001). Inhibition of FGFR3 in multiple myeloma lines and tumors has been shown to inhibit proliferation (Grand, 2004; Qing, 2009; Trudel, 2009; Krejci, 2010)
R-HSA-2077420 (Reactome) FGFR3 has been shown to be a target of a range of different tyrosine kinase inhibitors, including those restricted to in vitro use as well as a number that are currently in clinical trials for therapeutic use (see for instance, Paterson, 2004; Trudel, 2004; Trudel, 2005; Grand, 2004, Chen, 2005; Bernard-Pierrot, 2006; http::/clinicaltrials.gov). There are also two anti-FGFR3 antibodies that have shown preliminary promise in cancer cell lines or mouse models (Qing, 2009; Trudel, 2006).
R-HSA-5654148 (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-5654222 (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-5654224 (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-5654408 (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-5654409 (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-5654413 (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-5654416 (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-5654565 (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-5654623 (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-5654625 (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-5654628 (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-5654631 (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-5654633 (Reactome) p-FRS2 has two PPTN11/SHP2-binding sites at pY436 and pY471.
R-HSA-5654634 (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-5654637 (Reactome) The direct GRB2-binding sites of FRS2 have a major role in activation of the PI3K pathway.
R-HSA-5654640 (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(Rodrigues et al. 2000, Onishi-Haraikawa et al. 2001). In unstimulated cells, PI3K class IA exists as an inactive heterodimer of a p85 regulatory subunit (encoded by PIK3R1, PIK3R2 or PIK3R3) and a p110 catalytic subunit (encoded by PIK3CA, PIK3CB or PIK3CD). Binding of the iSH2 domain of the p85 regulatory subunit to the ABD and C2 domains of the p110 catalytic subunit both stabilizes p110 and inhibits its catalytic activity. This inhibition is relieved when the SH2 domains of p85 bind phosphorylated tyrosines on activated RTKs or their adaptor proteins. Binding to membrane-associated receptors brings activated PI3K in proximity to its membrane-localized substrate, PIP2 (Mandelker et al. 2009, Burke et al. 2011).
R-HSA-5654641 (Reactome) p-PPTN11 recruits GRB2-GAB1 to the activated receptor.
R-HSA-5654643 (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(Rodrigues et al. 2000, Onishi-Haraikawa et al. 2001). In unstimulated cells, PI3K class IA exists as an inactive heterodimer of a p85 regulatory subunit (encoded by PIK3R1, PIK3R2 or PIK3R3) and a p110 catalytic subunit (encoded by PIK3CA, PIK3CB or PIK3CD). Binding of the iSH2 domain of the p85 regulatory subunit to the ABD and C2 domains of the p110 catalytic subunit both stabilizes p110 and inhibits its catalytic activity. This inhibition is relieved when the SH2 domains of p85 bind phosphorylated tyrosines on activated RTKs or their adaptor proteins. Binding to membrane-associated receptors brings activated PI3K in proximity to its membrane-localized substrate, PIP2 (Mandelker et al. 2009, Burke et al. 2011).
R-HSA-5654646 (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-5654647 (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-5654679 (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-5654705 (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-5654709 (Reactome) Once recruited to the membrane, PI3K catalyzes the phosphorylation of PI(4,5)P2 to PI(3,4,5)P3.
R-HSA-5654730 (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-8851619 (Reactome) Fibroblast growth factor 23 (FGF23, aka phosphatonin) is a regulator of phosphate homeostasis and vitamin D metabolism. Its effects are thought to be mediated via the FGFR3c receptor. Glycosylation is necessary for FGF23 secretion from the cell and that is mediated by polypeptide N-acetylgalactosaminyltransferase 3 (GALNT3), which transfers an N-acetylgalactosaminyl (GalNAc) moiety from a high energy donor to threonine 178 on FGF23 (Kato et al. 2006). Competition between proprotein convertase cleavage and O-glycosylation determines the level of secreted active FGF23.
R-HSA-8851632 (Reactome) Once fibroblast growth factor 23 (FGF23, aka phosphatonin) is glycosylated by GALNT3, it is secreted from the cell (Kato et al. 2006).
R-HSA-8853309 (Reactome) FGFR3 fusions promote cellular proliferation and tumorigenesis that can be inhibited by tyrosine kinase inhibitors, suggesting that signaling is dependent on autophosphorylation of tyrosine residues in the intracellular region as is the case for WT FGFR3 (Singh et al, 2012; Parker et al, 2013; Williams et al, 2013; Wu et al, 2013; Yuan et al, 2014). FGFR3 fusions are reported to activate the ERK , STAT and AKT pathways, but not the PLC gamma pathway as the fusions generally lack the tyrosine residue required for PLC gamma recruitment (Parker et al, 2013; Williams et al, 2013; Wu et al, 2013; reviewed in Parker et al, 2014; Carter et al, 2015).
R-HSA-8853317 (Reactome) Constitutively active fusions of FGFR3 have been identified in glioblastoma, non-small cell lung cancer and bladder cancer, among others (Singh et al, 2012; Williams et al, 2013; Wu et al, 2013; Wang et al, 2014; Capelletti et al, 2014; Yuan et al, 2015; Carneiro et al, 2015; reviewed in Parker et al, 2014). The most prevalent fusion partner is TACC3 (transforming acidic coiled-coil-containing protein 3), a microtubule binding protein with roles in microtubule spindle assembly and chromosome segregation (Singh et al, 2014; Burgess et al, 2015; reviewed in Parker et al, 2014). There are conflicting reports about whether FGFR3 fusions form constitutive dimers, however ligand-independent autophosphorylation and downstream signaling has been demonstrated. FGFR3 fusions promote cellular proliferation and tumorigenesis and appear to escaped miRNA-mediated downregulation (Singh et al, 2012; Williams et al, 2013; Wu et al, 2013; Parker et al, 2013; reviewed in Parker et al, 2014).
R-HSA-8941628 (Reactome) RAS nucleotide is stimulated downstream of activated FGFR3 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)
SHC1-2,SHC1-3R-HSA-5654625 (Reactome)
SPRY2:B-RAFR-HSA-1295634 (Reactome)
SRC-1mim-catalysisR-HSA-1295609 (Reactome)
Tyrosine kinase

inhibitors of FGFR3

mutants
R-HSA-2077420 (Reactome)
UDP-GalNAcR-HSA-8851619 (Reactome)
UDPArrowR-HSA-8851619 (Reactome)
Ub-(Y55/Y227)p-SPRY2ArrowR-HSA-1295621 (Reactome)
Ub-Activated FGFR3 complex:Ub-p-FRS2ArrowR-HSA-5654679 (Reactome)
Ub:Y55/Y227-pSPRY2:CBLArrowR-HSA-934604 (Reactome)
Ub:Y55/Y227-pSPRY2:CBLR-HSA-1295621 (Reactome)
UbR-HSA-5654679 (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 FGFR3:PLCG1ArrowR-HSA-5654224 (Reactome)
activated FGFR3:PLCG1R-HSA-5654222 (Reactome)
activated FGFR3:PLCG1mim-catalysisR-HSA-5654222 (Reactome)
activated FGFR3:p-4Y-PLCG1ArrowR-HSA-5654222 (Reactome)
activated FGFR3:p-4Y-PLCG1R-HSA-5654148 (Reactome)
p-4Y-PLCG1ArrowR-HSA-5654148 (Reactome)
p-S111,S120-SPRY2ArrowR-HSA-1295604 (Reactome)
p-T,Y MAPK dimersArrowR-HSA-1295634 (Reactome)
p-T,Y MAPK dimersmim-catalysisR-HSA-5654565 (Reactome)
p-T250,T255,T385,S437-MKNK1mim-catalysisR-HSA-934559 (Reactome)
p-Y FGFR3 fusion dimersArrowR-HSA-8853309 (Reactome)
p-Y371-CBL:GRB2R-HSA-5654730 (Reactome)
p21 RAS:GDPR-HSA-5654413 (Reactome)
p21 RAS:GDPR-HSA-5654647 (Reactome)
p21 RAS:GDPR-HSA-8941628 (Reactome)
p21 RAS:GTPArrowR-HSA-5654413 (Reactome)
p21 RAS:GTPArrowR-HSA-5654647 (Reactome)
p21 RAS:GTPArrowR-HSA-8941628 (Reactome)
Personal tools