RAF/MAP kinase cascade (Homo sapiens)

From WikiPathways

Revision as of 10:15, 26 July 2016 by Ryanmiller (Talk | contribs)
Jump to: navigation, search
12, 26, 34, 45, 57...132, 207109, 130, 132, 139, 189...1481, 62, 71, 1484, 12, 100, 11768, 91, 26826, 30, 41, 44, 114...12, 26, 27, 40, 63...46, 76, 22125, 143, 20913, 14, 75, 108, 172...45, 8114, 23, 46, 132, 181...142, 177, 26377, 79, 82, 132, 27010, 15, 18, 30, 33...109, 130, 132, 139, 189...135, 149, 24036, 94, 105, 116, 151...69, 102, 133, 137, 205...142, 177, 250, 26381, 13246, 76, 2218, 30, 46, 70, 104...7, 106, 132, 19957, 72, 184, 19937, 53, 66, 76, 90...77, 79, 86142, 177, 26350, 162, 1955, 8, 104, 129, 161...12, 223, 25411, 12, 29, 42, 56...12, 223, 25411, 12, 56, 99, 126...27, 46, 63, 7695, 122, 131, 16725, 143, 2098, 14, 23, 45, 50...cytosolplasma membraneplasma membranenucleoplasmplasma membraneendosome lumenplasma membraneGolgi lumenVCL ARTN NRAS p-S445,T599,S602,S729 BRAF nuclear MAPK DUSPsFGB SPTAN1 p-Y349,Y350,Y427-SHC1 FGF1 p-S222,S226-MAP2K2 WDR83 p-S311,Y552 KSR1 VCL KRAS FGF9 p-T185,Y187-MAPK1 p-S,218, S222,T292 MAP2K1 p-S338,Y341,T491,S494,S621 RAF1p-2S,T MAP2K1:p-2SMAPK2K2 heterodimerPSMB1 PSMA2 NRAS FGF2(10-155) HRAS p-5Y-FRS3 Mn2+ CSK DAB2IP Signaling by SCF-KITUBC(229-304) PEA15 ARRB1 PSMA5 PPP2R5A SPRED dimerDUSP8 Ca2+ p-S311,Y552 KSR1 p-S218,S222-MAP2K1 KL-2 PSMD3 CSK p-6Y-FGFR3c IL17RDGDP YWHAB PSMA7 KRAS p-T,Y MAPKs p-Y1172,Y1226-ERBB4 JM-A CYT-2 isoform PSMB7 GTP ITGB3 p-S445,T599,S602,S729 BRAF NRAS MAPK1 p-S222,S226-MAP2K2 DUSP9 p-T202,Y204-MAPK3 RASA4 p-S218,S222-MAP2K1 RASA4 Cell surfaceinteractions at thevascular wallSPRED2 RBX1 p-S445,S729-BRAF VCL YWHAB p-S259,S621-RAF1 p-S299,S302,T452,T455,S576 ARAF NGF signalling viaTRKA from theplasma membranep-S222,S226-MAP2K2 PSMA1 Ca2+ UBC(533-608) KRAS FGG p-S311,Y552 KSR1 SYNGAP1 GRB2-1 NRAS WDR83:LAMTOR2:LAMTOR3p-T,Y MAPK monomersand dimersPhosphorylated p-Y877-ERBB2 heterodimers RASAL1 p-Y349,Y350-SHC1 RAP1A PSMB4 MARK3 IQGAP1 p-S214,S576 ARAF YWHAB PPP5CDUSP10 p21RAS:GTP:'activator'RAF:YWHAB dimerH2OYWHAB FGB p-S299,S302,T452,T455,S576 ARAF p-T185,Y187-MAPK1 PHBp-S222,S226-MAP2K2 RASGRF2 p-S338,Y341,T491,S494,S621 RAF1 p-8Y-FGFR1b JAK2 p-S299,S302,T452,T455,S576 ARAF GTP KSR1 p21 RAS:GTP:BRAPPDGFB(82-241) KSR2 CNKSR2 WDR83:LAMTOR2:LAMTOR3:activated RAF:p-2S MAP2K:p-T,Y MAPK complexGTP p-S311,Y552 KSR1 PSMC6 Mg2+ p-S445,T599,S602,S729 BRAF p-S338,Y341,T491,S494,S621 RAF1 L-Glu CNKSR2 KBTBD7 RASA1 KLB Ca2+ RASGRF1 HRAS MAPK3 UBC(1-76) NF1(2-2839) IL17RD ATPp-5Y-RET p-Y530-SRC WDR83 PSMB3 KL-1 p-S299,S302,T452,T455,S576 ARAF MAP2K1 YWHAB MAP2K2 PSMB6 p-S214,S576 ARAF p-S311,Y552 KSR1 GRIN2C KRAS PPP2R5D PSMD10 MARK3 PSMB11 GTPp-S222,S226-MAP2K2 p21 RAS:GTP:RAS GAPsp-S445,S729-BRAF ub-NF1(2-2839) F-actin PSMD12 ADPSignaling by FGFR3CNKSR1 CALM1 GRIN2B p-T202,Y204-MAPK3 NRAS p21RAS:GTP:activatedRAF1homo/heterodimer:PEBP1BRAP p-S338,Y341,T491,S494,S621 RAF1 WDR83 Ubp-S445,S729-BRAF GTP HBEGF(63-148) p-S311,Y552 KSR1 PAQR3MAP2K1 H2OGTP p-S311,Y552 KSR1 MAPK3 ATPp-S299,S302,T452,T455,S576 ARAF KRAS GTP RASGEF1A p-2S MAP2K1:p-2SMAPK2K2 heterodimerKRAS HRAS NEFL GTP p-T184-RASGRP1 p-S445,S729-BRAF VWF(23-763) PPP2R5B NRAS p-S311 KSR1 PP2APiMAP2K2 p-S338,Y341,T491,S494,S621 RAF1 Interleukin-3, 5 andGM-CSF signalingp-S338,Y341,T491,S494,S621 RAF1 p-ERBB4cyt1 homodimers MARK3 PSMD1 p-S299,S302,T452,T455,S576 ARAF GFRA1 p-S299,S302,T452,T455,S576 ARAF HRAS HRAS Interleukin-2signalingNCAM1 HS Gly YWHAB p-S338,Y341,T491,S494,S621 RAF1 H2ORASA1 p-S445,S729-BRAF MARK3 KRAS PPP2CB PSMD2 NRAS F-actin FGA PSMD8 MARK3 PSMB8 p-S218,S222-MAP2K1 WDR83:LAMTOR2:LAMTOR3:activated RAF:MAP2K:MAPK complexPhosphorylated ERBB2:ERBB4cyt1 heterodimers RAP1B MAPKsp-S338,Y341,T491,S494,S621 RAF1 UBB(153-228) MAP3K11LAMTOR2 VWF(23-763) 'activator'RAF:YWHAB dimerLAMTOR3 PP2Ap-S311 KSR1:MARK3Signaling by ERBB4IL17RD:p-2SMAP2Ks:p-T,Y MAPKsIL2RA SPTA1 GTP p-S299,S302,T452,T455,S576 ARAF MAPK1 p-Y427-SHC1 MAPK1 HRAS FGF10 YWHAB NRAS ADPGTP PSD-95 PTPRA Activated FGFR2c homodimer bound to FGF NRTN ATPp-S338,Y341,T491,S494,S621 RAF1 Pip-S365,S445,S729 BRAF PPP2R1A UBB(77-152) MAPK1 p-S222,S226-MAP2K2 MAPK1 CNKSR1 DUSP1 PSMD6 HRAS BRAP:KSR1:MARK3p-T,Y MAPKsp-S311,S406KSR1:MARK3APBB1IP UBC(153-228) TLN1 p-S299,S302,T452,T455,S576 ARAF GTP ADPFGA p-T185,Y187-MAPK1 p-T,Y MAPKs SPRED1 p21RAS:GTP:activatedRAF1homo/heterodimerADPYWHAB GTP ACTN2 PPP2R5C p-S311 KSR1 inactive RAFsPhosphorylated ERBB2:ERBB4cyt2 heterodimers UBC(609-684) MARK3 NRAS RAP1A p-Y194,Y195,Y272-SHC1-1(156-583) PSMA3 ATPIL17RD FN1(32-2386) IQGAP1 PPP2R5C RASGRF2 AKAP9 p-Y419-SRC RAS GAPsFN1(32-2386) p-S311,Y552 KSR1 p-S218,S222-MAP2K1 Signaling by ERBB2FGF8-1 PI(4,5)P2 p-7Y-KIT MARK3 p-5Y,S1119-TEK p-T,Y MAPK dimers GTP p-T185,Y187-MAPK1 PSMB10 UBB(77-152) SPTB YWHAB GFRA3 FGF6 RASAL1 MARK3 p-S445,T599,S602,S729 BRAF p-S445,T599,S602,S729 BRAF FGF17-1 RASAL3 p-T202,Y204-MAPK3 p-S445,T599,S602,S729 BRAF p-T286-CAMK2A p-S311,Y552 KSR1 p-6Y-FGFR3b PSMA6 p-S338,Y341,T491,S494,S621 RAF1 ub-BRAP Mg2+ PPP2R5E p-S576 ARAF RASAL2 MARK3 p-S311,Y552 KSR1 PiUBC(381-456) p21 RAS:GTPYWHAB MAPKs KRAS p-S621-RAF1 JAK3 p-S299,S302,T452,T455,S576 ARAF p-S445,T599,S602,S729 BRAF p-2S MAP2K homo/heterodimers SPTBN2 RAF activatingkinasesPPP2R5A MAPK3 RASA3 p-T,Y MAPK dimersp-S445,S729-BRAF UBB(1-76) APBB1IP PSME2 dephosphorylatedinactive RAFsFGF16 IL2 High affinity binding complex dimers of cytokine receptors using Bc, inactive JAK2, p-(Y593,628)-Bc:p(427,349,350)-SHC1 FGB SPRED3 RAP1B p-T133-RASGRP3 FGF20 dephosphorylatedinactive RAFS:YWHABdimerdephosphorylated"receiver" RAF/KSR1F-actin FGF18 p-Y530-SRC PPP2CA MARK3 FGF3 NRAS ATPNRAS H2Op-S445,S729-BRAF HRAS PPP2R5D p-S299,S302,T452,T455,S576 ARAF PPP2R5B UBC(305-380) ATPp-S445,S729-BRAF p-6Y-EGFR RAF/MAPK scaffoldsGTP UBC(77-152) p-8Y-FGFR1c PSMF1 DUSP10 NF1(2-2839) p-S445,T599,S602,S729 BRAF RASAL3 PSME4 YWHAB YWHAB dimerITGA2B(32-1039) APBB1IP p-S576 ARAF p-S338,Y341,T491,S494,S621 RAF1 KRAS p-S311,S406KSR1:MARK3:YWHABdimerRAP1B p-S311,Y552 KSR1 RAPGEF2 p-S445,T599,S602,S729 BRAF GTP CNKSR2 ANGPT1 DUSP6 p-T287-CAMK2B p-S338,Y341,T491,S494,S621 RAF1 p-S445,T599,S602,S729 BRAF p21RAS:GTP:activatedRAFhomo/heterodimercomplexesUBA52(1-76) p-S445,S729-BRAF p-T,Y MAPK dimers IL17RD:p-2SMAP2Ks:MAPKsPSME1 p-Y341,T491,S494,S621 RAF1 p-S445,T599,S602,S729 BRAF p-S338,Y341,T491,S494,S621 RAF1 p21RAS:GTP:activatedRAFhomo/heterodimersCa2+ HRAS UBB(153-228) p-Y-IRS2 p-S299,S302,T452,T455,S576 ARAF KRAS PSMD5 ADPUBB(1-76) SPRED1 PSMD9 ARRB1 UBC(457-532) GTP PSMD14 p21 RAS:GTP:ub-BRAPp-S311,Y552 KSR1 p-S222,S226-MAP2K2 FGF22 UBC(229-304) RAP1A UBC(609-684) p-S299,S302,T452,T455,S576 ARAF p-S621-RAF1 PPP2CA DUSP7 p-T287-CAMK2G NRAS KRAS p-2S MAP2Khomo/heterodimersp-S214,S576 ARAF p-T202,Y204-MAPK3 PI(4,5)P2 activatedRAF:scaffold:p-2SMAP2K:MAPK complexHRAS ARRB2 UbAPBB1IP inactive RAFs:YWHABdimerp-S218,S222-MAP2K1 p-S445,T599,S602,S729 BRAF KSR2 LAMTOR2 p-S445,T599,S602,S729 BRAF MARK3 p-S311,Y552 KSR1 p-S338,Y341,T491,S494,S621 RAF1 CSK PSMC2 ATPFGB PSMA8 Fc epsilon receptor(FCERI) signalingPPP2R1A YWHAB p-S621-RAF1 YWHAB p-S299,S302,T452,T455,S576 ARAF p-S445,T599,S602,S729 BRAF p-S445,S729-BRAF DUSP16 PSMD7 PSMC4 RAP1A NCAM signaling forneurite out-growthGFRA2 p-Y-JAK1 PSMC5 IL2RG KRAS p-S299,S302,T452,T455,S576 ARAF p-Y-SHC2 MAPK monomers anddimersp-S299,S302,T452,T455,S576 ARAF FGF4 p-S311,S406 KSR1 CNKSR2 UBC(305-380) UBC(77-152) p-5Y-LAT-2 SPRED1 Signaling by InsulinreceptorGFRA4 GTP p-Y-SHC1 SPRED dimer:ub-NF1RPS27A(1-76) p-S576 ARAF FGG DUSP16 HRAS PSMA4 FN1(32-2386) GDNF p-T185,Y187-MAPK1 Signaling by FGFR4Mn2+ p-S445,T599,S602,S729 BRAF CNKSR1 DUSP4 p-Y-SHC3 SPRED dimer:NF1NF1(2-2839) NF1(2-2839)NRAS DUSP5 p-T,Y MAPK monomersand dimersHRAS RASAL2 p-Y-IRS1 p-Y420-FYN p-S621-RAF1 p-S445,S729-BRAF p-5Y-FGFR4 H2OMn2+ PSMC1 PSMD4 NRAS PAQR3 MARK3 SPRED3 MAP2K1 NRAS Signaling by the BCell Receptor (BCR)Signaling by FGFR2KITLG-1(26-190) MAPK monomers anddimersp-S311 KSR1 PPP2R5E LAMTOR2 F-actin UBA52(1-76) p-12Y-PDGFRB ADPHRAS KRAS Signaling by PDGFPip-S299,S302,T452,T455,S576 ARAF p-S311,S406 KSR1 PDGFA-1 p21 RAS:GDPhyperphosphorylatedRAF1p-Y530-SRC DUSP8 p-S445,T599,S602,S729 BRAF p-S445,T599,S602,S729 BRAF hyperphosphorylatedBRAFp-S338,Y341,T491,S494,S621 RAF1 p-S338,Y341,T491,S494,S621 RAF1 p-Y239,Y240,Y317-SHC1-2 p-S445,T599,S602,S729 BRAF FGG KSR2 p-S311 KSR1 Pip-S365,S445,S729 BRAF ITGB3 p-S311,Y552 KSR1 p-S338,Y341,T491,S494,S621 RAF1 ARRB1 SPTBN1 GTP ITGB3 TLN1 UBC(1-76) MAPK3 HRAS SPTBN4 PPP2CB p-S338,Y341,T491,S494,S621 RAF1 GDPMAPK1 p-Y239,Y240,Y317-SHC1-2 PSMC3 p-S621-RAF1 PPP2R1B p-S311,Y552 KSR1 GRIN2A p21RAS:GTP:homo/heterodimerized RAF complexSPRED2 p-S218,S222-MAP2K1 KRAS p-S445,S729-BRAF p-S445,T599,S602,S729 BRAFp-S338,Y341,T491,S494,S621 RAF1 Signaling by Type 1Insulin-like GrowthFactor 1 Receptor(IGF1R)SYNGAP1 GTP H2OKRAS cytosolic MAPK DUSPsRAP1B LAMTOR3 BRAPPSPN MAPK3 MAP2Khomo/heterodimersp-T,Y MAPKs SPRED2 PI(4,5)P2 RPS27A(1-76) FGF5-1 ITGA2B(32-1039) FGF23(25-251) p-S445,T599,S602,S729 BRAF UBC(153-228) Mg2+ UBC(457-532) p-S259,S621-RAF1 activated RAFhomo/heterodimerPDGFB DAB2IP p-S445,T599,S602,S729 BRAF p-S311,Y552 KSR1 VWF(23-763) p-S311 KSR1 p-S338,Y341,T491,S494,S621 RAF1 IQGAP1 VCL ITGA2B(32-1039) p21RAS:GTP:p-S621RAF1homo/heterodimerUBC(381-456) p-S338,Y341,T491,S494,S621 RAF1 MAP2K2 PAQR3:inactive RAFsVWF(23-763) PiactivatedRAF:scaffold:MAP2K:MAPK complexFGG MARK3 PSMB9 NeurotransmitterReceptor BindingAnd DownstreamTransmission In The Postsynaptic CellPI(4,5)P2 FGA PSMB2 BRAP ITGB3 Signaling by EGFRMAPK3 Signaling by FGFR1p-S259,S621-RAF1 p-T,Y MAPK dimers DAP12 interactionsLAMTOR3 Mg2+ YWHAB KSR2 EGF-like ligands GRIN2D ARRB2 NRAS IQGAP1 TLN1 PDGFA-2 p-S311 KSR1 p-S218,S222-MAP2K1 RASA2 p-S365,S445,S729 BRAF MAPK1 activatedRAF:scaffold:p-2SMAP2K:p-2T MAPKcomplexp-S299,S302,T452,T455,S576 ARAF p-S299,S302,T452,T455,S576 ARAF UBC(533-608) Mn2+ KRAS SPRED3 HRAS p-S338,Y341,T491,S494,S621 RAF1 PSME3 SOS1 KSR1 CUL3 FGF19 p-6Y-ERBB2 YWHAB p-S299,S302,T452,T455,S576 ARAF p-S311 KSR1 p-S338,Y341,T491,S494,S621 RAF1 YWHAB KSR1:MARK3ARRB2 p21RAS:GTP:dephosphorylated RAF1 homo/heterodimerDAG ATPITGA2B(32-1039) p-S311,Y552 KSR1 CSK SPTBN5 RASA2 p-S338,Y341,T491,S494,S621 RAF1 GalNAc-T178-FGF23(25-251) PEBP1 HRAS p-T287-CAMK2D RASGRP1 p-7Y-ERBB2 ARRB2 RASA3 p-S311KSR1:MARK3:YWHABdimerPPP2R1B PSMD11 p-T202,Y204-MAPK3 ARRB1 GRIN1 RAS GEFsp-6Y-PTK2 ADPp-S445,S729-BRAF TLN1 RASGRP4 p-Y530-SRC p-T,Y MAPK monomersand dimers:PEA15Activated FGFR2b homodimer bound to FGF ADPp-11Y-PDGFRA MARK3 Ca2+ p-S445,S729-BRAF PSMD13 EGF PEA15PSMB5 p-S311 KSR1 DUSP2 p-S576 ARAF GTP FGA p-S299,S302,T452,T455,S576 ARAF Neuregulins p-Y364,Y418,Y536-IL2RB YWHAB CNKSR1 Phosphorylated p-6Y-ERBB2 heterodimers 26S proteasomeMAPK3 p-6Y-FRS2 KBTBD7:CUL3:RBX1p-S222,S226-MAP2K2 p-S445,S729-BRAF FN1(32-2386) PEBP1p-S445,T599,S602,S729 BRAF 19, 58, 144, 146, 160...84, 136, 141, 25812776123, 25921926632, 74, 112, 118, 163...9, 20, 65, 87, 229...28, 196, 202, 26022, 38, 43, 55, 59...31, 121, 227, 246882353, 6, 24, 35, 49...47, 61, 78, 97, 15092, 138, 2333, 6, 24, 35, 49...16, 21, 52, 168, 197...77773, 6, 24, 35, 49...152119, 2482, 17, 39, 48, 51...3, 6, 24, 35, 49...


Description

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). View original pathway at:Reactome.

Comments

Reactome Converter 
Pathway is converted from Reactome id:

Try the New WikiPathways

View approved pathways at the new wikipathways.org.

Quality Tags

Ontology Terms

 

Bibliography

View all...
  1. Mason CS, Springer CJ, Cooper RG, Superti-Furga G, Marshall CJ, Marais R.; ''Serine and tyrosine phosphorylations cooperate in Raf-1, but not B-Raf activation.''; PubMed Europe PMC Scholia
  2. Chadee DN, Kyriakis JM.; ''MLK3 is required for mitogen activation of B-Raf, ERK and cell proliferation.''; PubMed Europe PMC Scholia
  3. Cohen BD, Green JM, Foy L, Fell HP.; ''HER4-mediated biological and biochemical properties in NIH 3T3 cells. Evidence for HER1-HER4 heterodimers.''; PubMed Europe PMC Scholia
  4. King JA, Corcoran NM, D'Abaco GM, Straffon AF, Smith CT, Poon CL, Buchert M, I S, Hall NE, Lock P, Hovens CM.; ''Eve-3: a liver enriched suppressor of Ras/MAPK signaling.''; PubMed Europe PMC Scholia
  5. Sardi SP, Murtie J, Koirala S, Patten BA, Corfas G.; ''Presenilin-dependent ErbB4 nuclear signaling regulates the timing of astrogenesis in the developing brain.''; PubMed Europe PMC Scholia
  6. Lidke DS, Huang F, Post JN, Rieger B, Wilsbacher J, Thomas JL, Pouysségur J, Jovin TM, Lenormand P.; ''ERK nuclear translocation is dimerization-independent but controlled by the rate of phosphorylation.''; PubMed Europe PMC Scholia
  7. Van Noesel CJ, Brouns GS, van Schijndel GM, Bende RJ, Mason DY, Borst J, van Lier RA.; ''Comparison of human B cell antigen receptor complexes: membrane-expressed forms of immunoglobulin (Ig)M, IgD, and IgG are associated with structurally related heterodimers.''; PubMed Europe PMC Scholia
  8. Weber C, Fraemohs L, Dejana E.; ''The role of junctional adhesion molecules in vascular inflammation.''; PubMed Europe PMC Scholia
  9. Jones FE, Welte T, Fu XY, Stern DF.; ''ErbB4 signaling in the mammary gland is required for lobuloalveolar development and Stat5 activation during lactation.''; PubMed Europe PMC Scholia
  10. Rushworth LK, Hindley AD, O'Neill E, Kolch W.; ''Regulation and role of Raf-1/B-Raf heterodimerization.''; PubMed Europe PMC Scholia
  11. Wellbrock C, Karasarides M, Marais R.; ''The RAF proteins take centre stage.''; PubMed Europe PMC Scholia
  12. Heldin CH, Ostman A, Rönnstrand L.; ''Signal transduction via platelet-derived growth factor receptors.''; PubMed Europe PMC Scholia
  13. Whitehurst AW, Wilsbacher JL, You Y, Luby-Phelps K, Moore MS, Cobb MH.; ''ERK2 enters the nucleus by a carrier-independent mechanism.''; PubMed Europe PMC Scholia
  14. Stewart S, Sundaram M, Zhang Y, Lee J, Han M, Guan KL.; ''Kinase suppressor of Ras forms a multiprotein signaling complex and modulates MEK localization.''; PubMed Europe PMC Scholia
  15. Pritchard CA, Samuels ML, Bosch E, McMahon M.; ''Conditionally oncogenic forms of the A-Raf and B-Raf protein kinases display different biological and biochemical properties in NIH 3T3 cells.''; PubMed Europe PMC Scholia
  16. Peng SB, Henry JR, Kaufman MD, Lu WP, Smith BD, Vogeti S, Rutkoski TJ, Wise S, Chun L, Zhang Y, Van Horn RD, Yin T, Zhang X, Yadav V, Chen SH, Gong X, Ma X, Webster Y, Buchanan S, Mochalkin I, Huber L, Kays L, Donoho GP, Walgren J, McCann D, Patel P, Conti I, Plowman GD, Starling JJ, Flynn DL.; ''Inhibition of RAF Isoforms and Active Dimers by LY3009120 Leads to Anti-tumor Activities in RAS or BRAF Mutant Cancers.''; PubMed Europe PMC Scholia
  17. Park CY, Hoover PJ, Mullins FM, Bachhawat P, Covington ED, Raunser S, Walz T, Garcia KC, Dolmetsch RE, Lewis RS.; ''STIM1 clusters and activates CRAC channels via direct binding of a cytosolic domain to Orai1.''; PubMed Europe PMC Scholia
  18. Yeung K, Seitz T, Li S, Janosch P, McFerran B, Kaiser C, Fee F, Katsanakis KD, Rose DW, Mischak H, Sedivy JM, Kolch W.; ''Suppression of Raf-1 kinase activity and MAP kinase signalling by RKIP.''; PubMed Europe PMC Scholia
  19. Sjölander A, Yamamoto K, Huber BE, Lapetina EG.; ''Association of p21ras with phosphatidylinositol 3-kinase.''; PubMed Europe PMC Scholia
  20. Maki RG.; ''Small is beautiful: insulin-like growth factors and their role in growth, development, and cancer.''; PubMed Europe PMC Scholia
  21. Park S, Rath O, Beach S, Xiang X, Kelly SM, Luo Z, Kolch W, Yeung KC.; ''Regulation of RKIP binding to the N-region of the Raf-1 kinase.''; PubMed Europe PMC Scholia
  22. Zhang D, Sliwkowski MX, Mark M, Frantz G, Akita R, Sun Y, Hillan K, Crowley C, Brush J, Godowski PJ.; ''Neuregulin-3 (NRG3): a novel neural tissue-enriched protein that binds and activates ErbB4.''; PubMed Europe PMC Scholia
  23. Rauen KA.; ''The RASopathies.''; PubMed Europe PMC Scholia
  24. Xiang X, Zang M, Waelde CA, Wen R, Luo Z, Luo Z.; ''Phosphorylation of 338SSYY341 regulates specific interaction between Raf-1 and MEK1.''; PubMed Europe PMC Scholia
  25. Pinkas-Kramarski R, Soussan L, Waterman H, Levkowitz G, Alroy I, Klapper L, Lavi S, Seger R, Ratzkin BJ, Sela M, Yarden Y.; ''Diversification of Neu differentiation factor and epidermal growth factor signaling by combinatorial receptor interactions.''; PubMed Europe PMC Scholia
  26. Agianian B, Gavathiotis E.; ''Current Insights of BRAF Inhibitors in Cancer.''; PubMed Europe PMC Scholia
  27. Maertens O, Johnson B, Hollstein P, Frederick DT, Cooper ZA, Messiaen L, Bronson RT, McMahon M, Granter S, Flaherty K, Wargo JA, Marais R, Cichowski K.; ''Elucidating distinct roles for NF1 in melanomagenesis.''; PubMed Europe PMC Scholia
  28. DeFea KA, Zalevsky J, Thoma MS, Déry O, Mullins RD, Bunnett NW.; ''beta-arrestin-dependent endocytosis of proteinase-activated receptor 2 is required for intracellular targeting of activated ERK1/2.''; PubMed Europe PMC Scholia
  29. Bottorff D, Stang S, Agellon S, Stone JC.; ''RAS signalling is abnormal in a c-raf1 MEK1 double mutant.''; PubMed Europe PMC Scholia
  30. Shao H, Andres DA.; ''A novel RalGEF-like protein, RGL3, as a candidate effector for rit and Ras.''; PubMed Europe PMC Scholia
  31. Shi H, Moriceau G, Kong X, Lee MK, Lee H, Koya RC, Ng C, Chodon T, Scolyer RA, Dahlman KB, Sosman JA, Kefford RF, Long GV, Nelson SF, Ribas A, Lo RS.; ''Melanoma whole-exome sequencing identifies (V600E)B-RAF amplification-mediated acquired B-RAF inhibitor resistance.''; PubMed Europe PMC Scholia
  32. Sun Y, Ikrar T, Davis MF, Gong N, Zheng X, Luo ZD, Lai C, Mei L, Holmes TC, Gandhi SP, Xu X.; ''Neuregulin-1/ErbB4 Signaling Regulates Visual Cortical Plasticity.''; PubMed Europe PMC Scholia
  33. Baljuls A, Schmitz W, Mueller T, Zahedi RP, Sickmann A, Hekman M, Rapp UR.; ''Positive regulation of A-RAF by phosphorylation of isoform-specific hinge segment and identification of novel phosphorylation sites.''; PubMed Europe PMC Scholia
  34. Giroux S, Tremblay M, Bernard D, Cardin-Girard JF, Aubry S, Larouche L, Rousseau S, Huot J, Landry J, Jeannotte L, Charron J.; ''Embryonic death of Mek1-deficient mice reveals a role for this kinase in angiogenesis in the labyrinthine region of the placenta.''; PubMed Europe PMC Scholia
  35. Cacace AM, Michaud NR, Therrien M, Mathes K, Copeland T, Rubin GM, Morrison DK.; ''Identification of constitutive and ras-inducible phosphorylation sites of KSR: implications for 14-3-3 binding, mitogen-activated protein kinase binding, and KSR overexpression.''; PubMed Europe PMC Scholia
  36. Cutler RE, Stephens RM, Saracino MR, Morrison DK.; ''Autoregulation of the Raf-1 serine/threonine kinase.''; PubMed Europe PMC Scholia
  37. Roose JP, Mollenauer M, Gupta VA, Stone J, Weiss A.; ''A diacylglycerol-protein kinase C-RasGRP1 pathway directs Ras activation upon antigen receptor stimulation of T cells.''; PubMed Europe PMC Scholia
  38. Schober A, Weber C.; ''Mechanisms of monocyte recruitment in vascular repair after injury.''; PubMed Europe PMC Scholia
  39. Schaeffer HJ, Catling AD, Eblen ST, Collier LS, Krauss A, Weber MJ.; ''MP1: a MEK binding partner that enhances enzymatic activation of the MAP kinase cascade.''; PubMed Europe PMC Scholia
  40. Lupi C, Giannini R, Ugolini C, Proietti A, Berti P, Minuto M, Materazzi G, Elisei R, Santoro M, Miccoli P, Basolo F.; ''Association of BRAF V600E mutation with poor clinicopathological outcomes in 500 consecutive cases of papillary thyroid carcinoma.''; PubMed Europe PMC Scholia
  41. Saxena M, Williams S, Brockdorff J, Gilman J, Mustelin T.; ''Inhibition of T cell signaling by mitogen-activated protein kinase-targeted hematopoietic tyrosine phosphatase (HePTP).''; PubMed Europe PMC Scholia
  42. Gilmore-Hebert M, Ramabhadran R, Stern DF.; ''Interactions of ErbB4 and Kap1 connect the growth factor and DNA damage response pathways.''; PubMed Europe PMC Scholia
  43. Tsang E, Giannetti AM, Shaw D, Dinh M, Tse JK, Gandhi S, Ho H, Wang S, Papp E, Bradshaw JM.; ''Molecular mechanism of the Syk activation switch.''; PubMed Europe PMC Scholia
  44. Poulikakos PI, Persaud Y, Janakiraman M, Kong X, Ng C, Moriceau G, Shi H, Atefi M, Titz B, Gabay MT, Salton M, Dahlman KB, Tadi M, Wargo JA, Flaherty KT, Kelley MC, Misteli T, Chapman PB, Sosman JA, Graeber TG, Ribas A, Lo RS, Rosen N, Solit DB.; ''RAF inhibitor resistance is mediated by dimerization of aberrantly spliced BRAF(V600E).''; PubMed Europe PMC Scholia
  45. da Costa Martins P, van den Berk N, Ulfman LH, Koenderman L, Hordijk PL, Zwaginga JJ.; ''Platelet-monocyte complexes support monocyte adhesion to endothelium by enhancing secondary tethering and cluster formation.''; PubMed Europe PMC Scholia
  46. Wei SJ, Williams JG, Dang H, Darden TA, Betz BL, Humble MM, Chang FM, Trempus CS, Johnson K, Cannon RE, Tennant RW.; ''Identification of a specific motif of the DSS1 protein required for proteasome interaction and p53 protein degradation.''; PubMed Europe PMC Scholia
  47. Becker BF, Heindl B, Kupatt C, Zahler S.; ''Endothelial function and hemostasis.''; PubMed Europe PMC Scholia
  48. Müller J, Cacace AM, Lyons WE, McGill CB, Morrison DK.; ''Identification of B-KSR1, a novel brain-specific isoform of KSR1 that functions in neuronal signaling.''; PubMed Europe PMC Scholia
  49. Lorentzen A, Kinkhabwala A, Rocks O, Vartak N, Bastiaens PI.; ''Regulation of Ras localization by acylation enables a mode of intracellular signal propagation.''; PubMed Europe PMC Scholia
  50. Gauld SB, Cambier JC.; ''Src-family kinases in B-cell development and signaling.''; PubMed Europe PMC Scholia
  51. Deng Y, Shipps GW, Cooper A, English JM, Annis DA, Carr D, Nan Y, Wang T, Zhu HY, Chuang CC, Dayananth P, Hruza AW, Xiao L, Jin W, Kirschmeier P, Windsor WT, Samatar AA.; ''Discovery of novel, dual mechanism ERK inhibitors by affinity selection screening of an inactive kinase.''; PubMed Europe PMC Scholia
  52. Turjanski AG, Vaqué JP, Gutkind JS.; ''MAP kinases and the control of nuclear events.''; PubMed Europe PMC Scholia
  53. Harwood NE, Batista FD.; ''Early events in B cell activation.''; PubMed Europe PMC Scholia
  54. Sharma C, Vomastek T, Tarcsafalvi A, Catling AD, Schaeffer HJ, Eblen ST, Weber MJ.; ''MEK partner 1 (MP1): regulation of oligomerization in MAP kinase signaling.''; PubMed Europe PMC Scholia
  55. Plotnikov A, Zehorai E, Procaccia S, Seger R.; ''The MAPK cascades: signaling components, nuclear roles and mechanisms of nuclear translocation.''; PubMed Europe PMC Scholia
  56. Peterson SN, Trabalzini L, Brtva TR, Fischer T, Altschuler DL, Martelli P, Lapetina EG, Der CJ, White GC.; ''Identification of a novel RalGDS-related protein as a candidate effector for Ras and Rap1.''; PubMed Europe PMC Scholia
  57. Whitehurst AW, Robinson FL, Moore MS, Cobb MH.; ''The death effector domain protein PEA-15 prevents nuclear entry of ERK2 by inhibiting required interactions.''; PubMed Europe PMC Scholia
  58. Ferro E, Trabalzini L.; ''RalGDS family members couple Ras to Ral signalling and that's not all.''; PubMed Europe PMC Scholia
  59. Pylayeva-Gupta Y, Grabocka E, Bar-Sagi D.; ''RAS oncogenes: weaving a tumorigenic web.''; PubMed Europe PMC Scholia
  60. Khokhlatchev AV, Canagarajah B, Wilsbacher J, Robinson M, Atkinson M, Goldsmith E, Cobb MH.; ''Phosphorylation of the MAP kinase ERK2 promotes its homodimerization and nuclear translocation.''; PubMed Europe PMC Scholia
  61. Ishibe S, Joly D, Zhu X, Cantley LG.; ''Phosphorylation-dependent paxillin-ERK association mediates hepatocyte growth factor-stimulated epithelial morphogenesis.''; PubMed Europe PMC Scholia
  62. Lanier LL.; ''DAP10- and DAP12-associated receptors in innate immunity.''; PubMed Europe PMC Scholia
  63. Komuro A, Nagai M, Navin NE, Sudol M.; ''WW domain-containing protein YAP associates with ErbB-4 and acts as a co-transcriptional activator for the carboxyl-terminal fragment of ErbB-4 that translocates to the nucleus.''; PubMed Europe PMC Scholia
  64. Del Pino I, García-Frigola C, Dehorter N, Brotons-Mas JR, Alvarez-Salvado E, Martínez de Lagrán M, Ciceri G, Gabaldón MV, Moratal D, Dierssen M, Canals S, Marín O, Rico B.; ''Erbb4 deletion from fast-spiking interneurons causes schizophrenia-like phenotypes.''; PubMed Europe PMC Scholia
  65. McKay MM, Morrison DK.; ''Integrating signals from RTKs to ERK/MAPK.''; PubMed Europe PMC Scholia
  66. Hazan R, Margolis B, Dombalagian M, Ullrich A, Zilberstein A, Schlessinger J.; ''Identification of autophosphorylation sites of HER2/neu.''; PubMed Europe PMC Scholia
  67. Blalock WL, Weinstein-Oppenheimer C, Chang F, Hoyle PE, Wang XY, Algate PA, Franklin RA, Oberhaus SM, Steelman LS, McCubrey JA.; ''Signal transduction, cell cycle regulatory, and anti-apoptotic pathways regulated by IL-3 in hematopoietic cells: possible sites for intervention with anti-neoplastic drugs.''; PubMed Europe PMC Scholia
  68. Waterfall JJ, Arons E, Walker RL, Pineda M, Roth L, Killian JK, Abaan OD, Davis SR, Kreitman RJ, Meltzer PS.; ''High prevalence of MAP2K1 mutations in variant and IGHV4-34-expressing hairy-cell leukemias.''; PubMed Europe PMC Scholia
  69. McClatchey AI, Cichowski K.; ''SPRED proteins provide a NF-ty link to Ras suppression.''; PubMed Europe PMC Scholia
  70. Fischer A, Baljuls A, Reinders J, Nekhoroshkova E, Sibilski C, Metz R, Albert S, Rajalingam K, Hekman M, Rapp UR.; ''Regulation of RAF activity by 14-3-3 proteins: RAF kinases associate functionally with both homo- and heterodimeric forms of 14-3-3 proteins.''; PubMed Europe PMC Scholia
  71. Hollstein PE, Cichowski K.; ''Identifying the Ubiquitin Ligase complex that regulates the NF1 tumor suppressor and Ras.''; PubMed Europe PMC Scholia
  72. Chou FL, Hill JM, Hsieh JC, Pouyssegur J, Brunet A, Glading A, Uberall F, Ramos JW, Werner MH, Ginsberg MH.; ''PEA-15 binding to ERK1/2 MAPKs is required for its modulation of integrin activation.''; PubMed Europe PMC Scholia
  73. Maertens O, Cichowski K.; ''An expanding role for RAS GTPase activating proteins (RAS GAPs) in cancer.''; PubMed Europe PMC Scholia
  74. Wan PT, Garnett MJ, Roe SM, Lee S, Niculescu-Duvaz D, Good VM, Jones CM, Marshall CJ, Springer CJ, Barford D, Marais R, Cancer Genome Project.; ''Mechanism of activation of the RAF-ERK signaling pathway by oncogenic mutations of B-RAF.''; PubMed Europe PMC Scholia
  75. King AJ, Sun H, Diaz B, Barnard D, Miao W, Bagrodia S, Marshall MS.; ''The protein kinase Pak3 positively regulates Raf-1 activity through phosphorylation of serine 338.''; PubMed Europe PMC Scholia
  76. Spaargaren M, Bischoff JR.; ''Identification of the guanine nucleotide dissociation stimulator for Ral as a putative effector molecule of R-ras, H-ras, K-ras, and Rap.''; PubMed Europe PMC Scholia
  77. Vivanco I, Sawyers CL.; ''The phosphatidylinositol 3-Kinase AKT pathway in human cancer.''; PubMed Europe PMC Scholia
  78. Rodriguez-Viciana P, Warne PH, Dhand R, Vanhaesebroeck B, Gout I, Fry MJ, Waterfield MD, Downward J.; ''Phosphatidylinositol-3-OH kinase as a direct target of Ras.''; PubMed Europe PMC Scholia
  79. Rönnstrand L.; ''Signal transduction via the stem cell factor receptor/c-Kit.''; PubMed Europe PMC Scholia
  80. Brown MD, Sacks DB.; ''Protein scaffolds in MAP kinase signalling.''; PubMed Europe PMC Scholia
  81. Carpenter G.; ''Employment of the epidermal growth factor receptor in growth factor-independent signaling pathways.''; PubMed Europe PMC Scholia
  82. Martin GA, Viskochil D, Bollag G, McCabe PC, Crosier WJ, Haubruck H, Conroy L, Clark R, O'Connell P, Cawthon RM.; ''The GAP-related domain of the neurofibromatosis type 1 gene product interacts with ras p21.''; PubMed Europe PMC Scholia
  83. Castellano E, Downward J.; ''RAS Interaction with PI3K: More Than Just Another Effector Pathway.''; PubMed Europe PMC Scholia
  84. Brennan DF, Dar AC, Hertz NT, Chao WC, Burlingame AL, Shokat KM, Barford D.; ''A Raf-induced allosteric transition of KSR stimulates phosphorylation of MEK.''; PubMed Europe PMC Scholia
  85. Kubicek M, Pacher M, Abraham D, Podar K, Eulitz M, Baccarini M.; ''Dephosphorylation of Ser-259 regulates Raf-1 membrane association.''; PubMed Europe PMC Scholia
  86. Zehir A, Benayed R, Shah RH, Syed A, Middha S, Kim HR, Srinivasan P, Gao J, Chakravarty D, Devlin SM, Hellmann MD, Barron DA, Schram AM, Hameed M, Dogan S, Ross DS, Hechtman JF, DeLair DF, Yao J, Mandelker DL, Cheng DT, Chandramohan R, Mohanty AS, Ptashkin RN, Jayakumaran G, Prasad M, Syed MH, Rema AB, Liu ZY, Nafa K, Borsu L, Sadowska J, Casanova J, Bacares R, Kiecka IJ, Razumova A, Son JB, Stewart L, Baldi T, Mullaney KA, Al-Ahmadie H, Vakiani E, Abeshouse AA, Penson AV, Jonsson P, Camacho N, Chang MT, Won HH, Gross BE, Kundra R, Heins ZJ, Chen HW, Phillips S, Zhang H, Wang J, Ochoa A, Wills J, Eubank M, Thomas SB, Gardos SM, Reales DN, Galle J, Durany R, Cambria R, Abida W, Cercek A, Feldman DR, Gounder MM, Hakimi AA, Harding JJ, Iyer G, Janjigian YY, Jordan EJ, Kelly CM, Lowery MA, Morris LGT, Omuro AM, Raj N, Razavi P, Shoushtari AN, Shukla N, Soumerai TE, Varghese AM, Yaeger R, Coleman J, Bochner B, Riely GJ, Saltz LB, Scher HI, Sabbatini PJ, Robson ME, Klimstra DS, Taylor BS, Baselga J, Schultz N, Hyman DM, Arcila ME, Solit DB, Ladanyi M, Berger MF.; ''Mutational landscape of metastatic cancer revealed from prospective clinical sequencing of 10,000 patients.''; PubMed Europe PMC Scholia
  87. Edin ML, Juliano RL.; ''Raf-1 serine 338 phosphorylation plays a key role in adhesion-dependent activation of extracellular signal-regulated kinase by epidermal growth factor.''; PubMed Europe PMC Scholia
  88. Bollag G, Clapp DW, Shih S, Adler F, Zhang YY, Thompson P, Lange BJ, Freedman MH, McCormick F, Jacks T, Shannon K.; ''Loss of NF1 results in activation of the Ras signaling pathway and leads to aberrant growth in haematopoietic cells.''; PubMed Europe PMC Scholia
  89. Holzenberger M.; ''Igf-I signaling and effects on longevity.''; PubMed Europe PMC Scholia
  90. Papin C, Eychène A, Brunet A, Pagès G, Pouysségur J, Calothy G, Barnier JV.; ''B-Raf protein isoforms interact with and phosphorylate Mek-1 on serine residues 218 and 222.''; PubMed Europe PMC Scholia
  91. Sun Y, Alberta JA, Pilarz C, Calligaris D, Chadwick EJ, Ramkissoon SH, Ramkissoon LA, Garcia VM, Mazzola E, Goumnerova L, Kane M, Yao Z, Kieran MW, Ligon KL, Hahn WC, Garraway LA, Rosen N, Gray NS, Agar NY, Buhrlage SJ, Segal RA, Stiles CD.; ''A brain-penetrant RAF dimer antagonist for the noncanonical BRAF oncoprotein of pediatric low-grade astrocytomas.''; PubMed Europe PMC Scholia
  92. Kunkel HG.; ''Surface markers of human lymphocytes.''; PubMed Europe PMC Scholia
  93. Mitchell RM, Janssen MJ, Karavanova I, Vullhorst D, Furth K, Makusky A, Markey SP, Buonanno A.; ''ErbB4 reduces synaptic GABAA currents independent of its receptor tyrosine kinase activity.''; PubMed Europe PMC Scholia
  94. Sundaram M, Han M.; ''The C. elegans ksr-1 gene encodes a novel Raf-related kinase involved in Ras-mediated signal transduction.''; PubMed Europe PMC Scholia
  95. Higgins EM, Bos JM, Mason-Suares H, Tester DJ, Ackerman JP, MacRae CA, Sol-Church K, Gripp KW, Urrutia R, Ackerman MJ.; ''Elucidation of MRAS-mediated Noonan syndrome with cardiac hypertrophy.''; PubMed Europe PMC Scholia
  96. Kornfeld K, Hom DB, Horvitz HR.; ''The ksr-1 gene encodes a novel protein kinase involved in Ras-mediated signaling in C. elegans.''; PubMed Europe PMC Scholia
  97. Okaniwa M, Hirose M, Arita T, Yabuki M, Nakamura A, Takagi T, Kawamoto T, Uchiyama N, Sumita A, Tsutsumi S, Tottori T, Inui Y, Sang BC, Yano J, Aertgeerts K, Yoshida S, Ishikawa T.; ''Discovery of a selective kinase inhibitor (TAK-632) targeting pan-RAF inhibition: design, synthesis, and biological evaluation of C-7-substituted 1,3-benzothiazole derivatives.''; PubMed Europe PMC Scholia
  98. Jaumot M, Hancock JF.; ''Protein phosphatases 1 and 2A promote Raf-1 activation by regulating 14-3-3 interactions.''; PubMed Europe PMC Scholia
  99. von Kriegsheim A, Pitt A, Grindlay GJ, Kolch W, Dhillon AS.; ''Regulation of the Raf-MEK-ERK pathway by protein phosphatase 5.''; PubMed Europe PMC Scholia
  100. Alessi DR, Saito Y, Campbell DG, Cohen P, Sithanandam G, Rapp U, Ashworth A, Marshall CJ, Cowley S.; ''Identification of the sites in MAP kinase kinase-1 phosphorylated by p74raf-1.''; PubMed Europe PMC Scholia
  101. Rodriguez-Viciana P, Rauen KA.; ''Biochemical characterization of novel germline BRAF and MEK mutations in cardio-facio-cutaneous syndrome.''; PubMed Europe PMC Scholia
  102. Bezniakow N, Gos M, Obersztyn E.; ''The RASopathies as an example of RAS/MAPK pathway disturbances - clinical presentation and molecular pathogenesis of selected syndromes.''; PubMed Europe PMC Scholia
  103. Cargnello M, Roux PP.; ''Activation and function of the MAPKs and their substrates, the MAPK-activated protein kinases.''; PubMed Europe PMC Scholia
  104. Tang J, Qi X, Mercola D, Han J, Chen G.; ''Essential role of p38gamma in K-Ras transformation independent of phosphorylation.''; PubMed Europe PMC Scholia
  105. Vakana E, Pratt S, Blosser W, Dowless M, Simpson N, Yuan XJ, Jaken S, Manro J, Stephens J, Zhang Y, Huber L, Peng SB, Stancato LF.; ''LY3009120, a panRAF inhibitor, has significant anti-tumor activity in BRAF and KRAS mutant preclinical models of colorectal cancer.''; PubMed Europe PMC Scholia
  106. Vomastek T, Schaeffer HJ, Tarcsafalvi A, Smolkin ME, Bissonette EA, Weber MJ.; ''Modular construction of a signaling scaffold: MORG1 interacts with components of the ERK cascade and links ERK signaling to specific agonists.''; PubMed Europe PMC Scholia
  107. Rajalingam K, Wunder C, Brinkmann V, Churin Y, Hekman M, Sievers C, Rapp UR, Rudel T.; ''Prohibitin is required for Ras-induced Raf-MEK-ERK activation and epithelial cell migration.''; PubMed Europe PMC Scholia
  108. Simanshu DK, Nissley DV, McCormick F.; ''RAS Proteins and Their Regulators in Human Disease.''; PubMed Europe PMC Scholia
  109. Ray LB, Sturgill TW.; ''Insulin-stimulated microtubule-associated protein kinase is phosphorylated on tyrosine and threonine in vivo.''; PubMed Europe PMC Scholia
  110. Arasada RR, Carpenter G.; ''Secretase-dependent tyrosine phosphorylation of Mdm2 by the ErbB-4 intracellular domain fragment.''; PubMed Europe PMC Scholia
  111. Lito P, Rosen N, Solit DB.; ''Tumor adaptation and resistance to RAF inhibitors.''; PubMed Europe PMC Scholia
  112. Rowley RB, Burkhardt AL, Chao HG, Matsueda GR, Bolen JB.; ''Syk protein-tyrosine kinase is regulated by tyrosine-phosphorylated Ig alpha/Ig beta immunoreceptor tyrosine activation motif binding and autophosphorylation.''; PubMed Europe PMC Scholia
  113. Marshall CB, Meiri D, Smith MJ, Mazhab-Jafari MT, Gasmi-Seabrook GM, Rottapel R, Stambolic V, Ikura M.; ''Probing the GTPase cycle with real-time NMR: GAP and GEF activities in cell extracts.''; PubMed Europe PMC Scholia
  114. Zhang H, Koo CY, Stebbing J, Giamas G.; ''The dual function of KSR1: a pseudokinase and beyond.''; PubMed Europe PMC Scholia
  115. Garnett MJ, Rana S, Paterson H, Barford D, Marais R.; ''Wild-type and mutant B-RAF activate C-RAF through distinct mechanisms involving heterodimerization.''; PubMed Europe PMC Scholia
  116. Muraoka-Cook RS, Sandahl M, Hunter D, Miraglia L, Earp HS.; ''Prolactin and ErbB4/HER4 signaling interact via Janus kinase 2 to induce mammary epithelial cell gene expression differentiation.''; PubMed Europe PMC Scholia
  117. Rodriguez-Viciana P, Oses-Prieto J, Burlingame A, Fried M, McCormick F.; ''A phosphatase holoenzyme comprised of Shoc2/Sur8 and the catalytic subunit of PP1 functions as an M-Ras effector to modulate Raf activity.''; PubMed Europe PMC Scholia
  118. Cichowski K, Santiago S, Jardim M, Johnson BW, Jacks T.; ''Dynamic regulation of the Ras pathway via proteolysis of the NF1 tumor suppressor.''; PubMed Europe PMC Scholia
  119. Friedman WJ, Greene LA.; ''Neurotrophin signaling via Trks and p75.''; PubMed Europe PMC Scholia
  120. Luk PP, Yu B, Ng CC, Mercorella B, Selinger C, Lum T, Kao S, O'Toole SA, Cooper WA.; ''BRAF mutations in non-small cell lung cancer.''; PubMed Europe PMC Scholia
  121. Zheng CF, Guan KL.; ''Cytoplasmic localization of the mitogen-activated protein kinase activator MEK.''; PubMed Europe PMC Scholia
  122. Muik M, Frischauf I, Derler I, Fahrner M, Bergsmann J, Eder P, Schindl R, Hesch C, Polzinger B, Fritsch R, Kahr H, Madl J, Gruber H, Groschner K, Romanin C.; ''Dynamic coupling of the putative coiled-coil domain of ORAI1 with STIM1 mediates ORAI1 channel activation.''; PubMed Europe PMC Scholia
  123. Udell CM, Rajakulendran T, Sicheri F, Therrien M.; ''Mechanistic principles of RAF kinase signaling.''; PubMed Europe PMC Scholia
  124. Müller J, Ory S, Copeland T, Piwnica-Worms H, Morrison DK.; ''C-TAK1 regulates Ras signaling by phosphorylating the MAPK scaffold, KSR1.''; PubMed Europe PMC Scholia
  125. Anastasaki C, Estep AL, Marais R, Rauen KA, Patton EE.; ''Kinase-activating and kinase-impaired cardio-facio-cutaneous syndrome alleles have activity during zebrafish development and are sensitive to small molecule inhibitors.''; PubMed Europe PMC Scholia
  126. Ehrhardt GR, Korherr C, Wieler JS, Knaus M, Schrader JW.; ''A novel potential effector of M-Ras and p21 Ras negatively regulates p21 Ras-mediated gene induction and cell growth.''; PubMed Europe PMC Scholia
  127. Lanier LL, Corliss BC, Wu J, Leong C, Phillips JH.; ''Immunoreceptor DAP12 bearing a tyrosine-based activation motif is involved in activating NK cells.''; PubMed Europe PMC Scholia
  128. Wienands J, Freuler F, Baumann G.; ''Tyrosine-phosphorylated forms of Ig beta, CD22, TCR zeta and HOSS are major ligands for tandem SH2 domains of Syk.''; PubMed Europe PMC Scholia
  129. Nussinov R, Muratcioglu S, Tsai CJ, Jang H, Gursoy A, Keskin O.; ''The Key Role of Calmodulin in KRAS-Driven Adenocarcinomas.''; PubMed Europe PMC Scholia
  130. Casar B, Pinto A, Crespo P.; ''Essential role of ERK dimers in the activation of cytoplasmic but not nuclear substrates by ERK-scaffold complexes.''; PubMed Europe PMC Scholia
  131. Cordeddu V, Di Schiavi E, Pennacchio LA, Ma'ayan A, Sarkozy A, Fodale V, Cecchetti S, Cardinale A, Martin J, Schackwitz W, Lipzen A, Zampino G, Mazzanti L, Digilio MC, Martinelli S, Flex E, Lepri F, Bartholdi D, Kutsche K, Ferrero GB, Anichini C, Selicorni A, Rossi C, Tenconi R, Zenker M, Merlo D, Dallapiccola B, Iyengar R, Bazzicalupo P, Gelb BD, Tartaglia M.; ''Mutation of SHOC2 promotes aberrant protein N-myristoylation and causes Noonan-like syndrome with loose anagen hair.''; PubMed Europe PMC Scholia
  132. Ahmadian MR, Kiel C, Stege P, Scheffzek K.; ''Structural fingerprints of the Ras-GTPase activating proteins neurofibromin and p120GAP.''; PubMed Europe PMC Scholia
  133. Haskins JW, Zhang S, Means RE, Kelleher JK, Cline GW, Canfrán-Duque A, Suárez Y, Stern DF.; ''Neuregulin-activated ERBB4 induces the SREBP-2 cholesterol biosynthetic pathway and increases low-density lipoprotein uptake.''; PubMed Europe PMC Scholia
  134. Oh-hora M, Ogata M, Mori Y, Adachi M, Imai K, Kosugi A, Hamaoka T.; ''Direct suppression of TCR-mediated activation of extracellular signal-regulated kinase by leukocyte protein tyrosine phosphatase, a tyrosine-specific phosphatase.''; PubMed Europe PMC Scholia
  135. Wang X, Lupardus P, Laporte SL, Garcia KC.; ''Structural biology of shared cytokine receptors.''; PubMed Europe PMC Scholia
  136. Rajalingam K, Rudel T.; ''Ras-Raf signaling needs prohibitin.''; PubMed Europe PMC Scholia
  137. Bermudez O, Pagès G, Gimond C.; ''The dual-specificity MAP kinase phosphatases: critical roles in development and cancer.''; PubMed Europe PMC Scholia
  138. Ahmadian MR, Stege P, Scheffzek K, Wittinghofer A.; ''Confirmation of the arginine-finger hypothesis for the GAP-stimulated GTP-hydrolysis reaction of Ras.''; PubMed Europe PMC Scholia
  139. Saouaf SJ, Mahajan S, Rowley RB, Kut SA, Fargnoli J, Burkhardt AL, Tsukada S, Witte ON, Bolen JB.; ''Temporal differences in the activation of three classes of non-transmembrane protein tyrosine kinases following B-cell antigen receptor surface engagement.''; PubMed Europe PMC Scholia
  140. Jaiswal RK, Moodie SA, Wolfman A, Landreth GE.; ''The mitogen-activated protein kinase cascade is activated by B-Raf in response to nerve growth factor through interaction with p21ras.''; PubMed Europe PMC Scholia
  141. Clapéron A, Therrien M.; ''KSR and CNK: two scaffolds regulating RAS-mediated RAF activation.''; PubMed Europe PMC Scholia
  142. McKay MM, Ritt DA, Morrison DK.; ''Signaling dynamics of the KSR1 scaffold complex.''; PubMed Europe PMC Scholia
  143. Brummer T, Naegele H, Reth M, Misawa Y.; ''Identification of novel ERK-mediated feedback phosphorylation sites at the C-terminus of B-Raf.''; PubMed Europe PMC Scholia
  144. Formstecher E, Ramos JW, Fauquet M, Calderwood DA, Hsieh JC, Canton B, Nguyen XT, Barnier JV, Camonis J, Ginsberg MH, Chneiweiss H.; ''PEA-15 mediates cytoplasmic sequestration of ERK MAP kinase.''; PubMed Europe PMC Scholia
  145. Stone JC.; ''Regulation of Ras in lymphocytes: get a GRP.''; PubMed Europe PMC Scholia
  146. Wali VB, Haskins JW, Gilmore-Hebert M, Platt JT, Liu Z, Stern DF.; ''Convergent and divergent cellular responses by ErbB4 isoforms in mammary epithelial cells.''; PubMed Europe PMC Scholia
  147. Bélanger LF, Roy S, Tremblay M, Brott B, Steff AM, Mourad W, Hugo P, Erikson R, Charron J.; ''Mek2 is dispensable for mouse growth and development.''; PubMed Europe PMC Scholia
  148. Hou SW, Zhi HY, Pohl N, Loesch M, Qi XM, Li RS, Basir Z, Chen G.; ''PTPH1 dephosphorylates and cooperates with p38gamma MAPK to increase ras oncogenesis through PDZ-mediated interaction.''; PubMed Europe PMC Scholia
  149. Eswarakumar VP, Lax I, Schlessinger J.; ''Cellular signaling by fibroblast growth factor receptors.''; PubMed Europe PMC Scholia
  150. Beenken A, Mohammadi M.; ''The FGF family: biology, pathophysiology and therapy.''; PubMed Europe PMC Scholia
  151. Ritt DA, Monson DM, Specht SI, Morrison DK.; ''Impact of feedback phosphorylation and Raf heterodimerization on normal and mutant B-Raf signaling.''; PubMed Europe PMC Scholia
  152. Roskoski R.; ''Targeting ERK1/2 protein-serine/threonine kinases in human cancers.''; PubMed Europe PMC Scholia
  153. Irish JM, Czerwinski DK, Nolan GP, Levy R.; ''Kinetics of B cell receptor signaling in human B cell subsets mapped by phosphospecific flow cytometry.''; PubMed Europe PMC Scholia
  154. Omerovic J, Santangelo L, Puggioni EM, Marrocco J, Dall'Armi C, Palumbo C, Belleudi F, Di Marcotullio L, Frati L, Torrisi MR, Cesareni G, Gulino A, Alimandi M.; ''The E3 ligase Aip4/Itch ubiquitinates and targets ErbB-4 for degradation.''; PubMed Europe PMC Scholia
  155. Amin DN, Perkins AS, Stern DF.; ''Gene expression profiling of ErbB receptor and ligand-dependent transcription.''; PubMed Europe PMC Scholia
  156. 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
  157. Leicht DT, Balan V, Zhu J, Kaplun A, Bronisz A, Rana A, Tzivion G.; ''MEK-1 activates C-Raf through a Ras-independent mechanism.''; PubMed Europe PMC Scholia
  158. Reber L, Da Silva CA, Frossard N.; ''Stem cell factor and its receptor c-Kit as targets for inflammatory diseases.''; PubMed Europe PMC Scholia
  159. Xia K, Mukhopadhyay NK, Inhorn RC, Barber DL, Rose PE, Lee RS, Narsimhan RP, D'Andrea AD, Griffin JD, Roberts TM.; ''The cytokine-activated tyrosine kinase JAK2 activates Raf-1 in a p21ras-dependent manner.''; PubMed Europe PMC Scholia
  160. Ditlevsen DK, Povlsen GK, Berezin V, Bock E.; ''NCAM-induced intracellular signaling revisited.''; PubMed Europe PMC Scholia
  161. Orru G, Coghe F, Faa G, Pillai S, Manieli C, Montaldo C, Pilia F, Pichiri G, Piras V, Coni P.; ''Rapid multiplex real-time PCR by molecular beacons for different BRAF allele detection in papillary thyroid carcinoma.''; PubMed Europe PMC Scholia
  162. Hata D, Nakamura T, Kawakami T, Kawakami Y, Herren B, Mayumi M.; ''Tyrosine phosphorylation of MB-1, B29, and HS1 proteins in human B cells following receptor crosslinking.''; PubMed Europe PMC Scholia
  163. Germann UA, Furey BF, Markland W, Hoover RR, Aronov AM, Roix JJ, Hale M, Boucher DM, Sorrell DA, Martinez-Botella G, Fitzgibbon M, Shapiro P, Wick MJ, Samadani R, Meshaw K, Groover A, DeCrescenzo G, Namchuk M, Emery CM, Saha S, Welsch DJ.; ''Targeting the MAPK Signaling Pathway in Cancer: Promising Preclinical Activity with the Novel Selective ERK1/2 Inhibitor BVD-523 (Ulixertinib).''; PubMed Europe PMC Scholia
  164. Pettiford SM, Herbst R.; ''The MAP-kinase ERK2 is a specific substrate of the protein tyrosine phosphatase HePTP.''; PubMed Europe PMC Scholia
  165. King AJ, Patrick DR, Batorsky RS, Ho ML, Do HT, Zhang SY, Kumar R, Rusnak DW, Takle AK, Wilson DM, Hugger E, Wang L, Karreth F, Lougheed JC, Lee J, Chau D, Stout TJ, May EW, Rominger CM, Schaber MD, Luo L, Lakdawala AS, Adams JL, Contractor RG, Smalley KS, Herlyn M, Morrissey MM, Tuveson DA, Huang PS.; ''Demonstration of a genetic therapeutic index for tumors expressing oncogenic BRAF by the kinase inhibitor SB-590885.''; PubMed Europe PMC Scholia
  166. Williams CC, Allison JG, Vidal GA, Burow ME, Beckman BS, Marrero L, Jones FE.; ''The ERBB4/HER4 receptor tyrosine kinase regulates gene expression by functioning as a STAT5A nuclear chaperone.''; PubMed Europe PMC Scholia
  167. Gibbs JB, Sigal IS, Poe M, Scolnick EM.; ''Intrinsic GTPase activity distinguishes normal and oncogenic ras p21 molecules.''; PubMed Europe PMC Scholia
  168. Tran NH, Frost JA.; ''Phosphorylation of Raf-1 by p21-activated kinase 1 and Src regulates Raf-1 autoinhibition.''; PubMed Europe PMC Scholia
  169. Dailey L, Ambrosetti D, Mansukhani A, Basilico C.; ''Mechanisms underlying differential responses to FGF signaling.''; PubMed Europe PMC Scholia
  170. Roskoski R.; ''ERK1/2 MAP kinases: structure, function, and regulation.''; PubMed Europe PMC Scholia
  171. Hayes NV, Blackburn E, Smart LV, Boyle MM, Russell GA, Frost TM, Morgan BJ, Baines AJ, Gullick WJ.; ''Identification and characterization of novel spliced variants of neuregulin 4 in prostate cancer.''; PubMed Europe PMC Scholia
  172. Moschos SJ, Sullivan RJ, Hwu WJ, Ramanathan RK, Adjei AA, Fong PC, Shapira-Frommer R, Tawbi HA, Rubino J, Rush TS, Zhang D, Miselis NR, Samatar AA, Chun P, Rubin EH, Schiller J, Long BJ, Dayananth P, Carr D, Kirschmeier P, Bishop WR, Deng Y, Cooper A, Shipps GW, Moreno BH, Robert L, Ribas A, Flaherty KT.; ''Development of MK-8353, an orally administered ERK1/2 inhibitor, in patients with advanced solid tumors.''; PubMed Europe PMC Scholia
  173. Hatzivassiliou G, Haling JR, Chen H, Song K, Price S, Heald R, Hewitt JF, Zak M, Peck A, Orr C, Merchant M, Hoeflich KP, Chan J, Luoh SM, Anderson DJ, Ludlam MJ, Wiesmann C, Ultsch M, Friedman LS, Malek S, Belvin M.; ''Mechanism of MEK inhibition determines efficacy in mutant KRAS- versus BRAF-driven cancers.''; PubMed Europe PMC Scholia
  174. Ahrens S, Jaramillo S, Yu K, Ghosh S, Hwang GR, Paik R, Lai C, He M, Huang ZJ, Li B.; ''ErbB4 regulation of a thalamic reticular nucleus circuit for sensory selection.''; PubMed Europe PMC Scholia
  175. Lenormand P, Sardet C, Pagès G, L'Allemain G, Brunet A, Pouysségur J.; ''Growth factors induce nuclear translocation of MAP kinases (p42mapk and p44mapk) but not of their activator MAP kinase kinase (p45mapkk) in fibroblasts.''; PubMed Europe PMC Scholia
  176. Jackson SP, Mistry N, Yuan Y.; ''Platelets and the injured vessel wall-- "rolling into action": focus on glycoprotein Ib/V/IX and the platelet cytoskeleton.''; PubMed Europe PMC Scholia
  177. Carter RH, Park DJ, Rhee SG, Fearon DT.; ''Tyrosine phosphorylation of phospholipase C induced by membrane immunoglobulin in B lymphocytes.''; PubMed Europe PMC Scholia
  178. Schmid RS, Maness PF.; ''L1 and NCAM adhesion molecules as signaling coreceptors in neuronal migration and process outgrowth.''; PubMed Europe PMC Scholia
  179. Shinohara H, Kurosaki T.; ''Comprehending the complex connection between PKCbeta, TAK1, and IKK in BCR signaling.''; PubMed Europe PMC Scholia
  180. Wells A.; ''EGF receptor.''; PubMed Europe PMC Scholia
  181. Fu SM, Winchester RJ, Kunkel HG.; ''Similar idiotypic specificity for the membrane IgD and IgM of human B lymphocytes.''; PubMed Europe PMC Scholia
  182. Yamamori B, Kuroda S, Shimizu K, Fukui K, Ohtsuka T, Takai Y.; ''Purification of a Ras-dependent mitogen-activated protein kinase kinase kinase from bovine brain cytosol and its identification as a complex of B-Raf and 14-3-3 proteins.''; PubMed Europe PMC Scholia
  183. Marais R, Light Y, Paterson HF, Mason CS, Marshall CJ.; ''Differential regulation of Raf-1, A-Raf, and B-Raf by oncogenic ras and tyrosine kinases.''; PubMed Europe PMC Scholia
  184. Feng L, Xie X, Ding Q, Luo X, He J, Fan F, Liu W, Wang Z, Chen Y.; ''Spatial regulation of Raf kinase signaling by RKTG.''; PubMed Europe PMC Scholia
  185. Annunziata M, Granata R, Ghigo E.; ''The IGF system.''; PubMed Europe PMC Scholia
  186. Lu Y, Sun XD, Hou FQ, Bi LL, Yin DM, Liu F, Chen YJ, Bean JC, Jiao HF, Liu X, Li BM, Xiong WC, Gao TM, Mei L.; ''Maintenance of GABAergic activity by neuregulin 1-ErbB4 in amygdala for fear memory.''; PubMed Europe PMC Scholia
  187. Law CL, Sidorenko SP, Chandran KA, Draves KE, Chan AC, Weiss A, Edelhoff S, Disteche CM, Clark EA.; ''Molecular cloning of human Syk. A B cell protein-tyrosine kinase associated with the surface immunoglobulin M-B cell receptor complex.''; PubMed Europe PMC Scholia
  188. Tsai J, Lee JT, Wang W, Zhang J, Cho H, Mamo S, Bremer R, Gillette S, Kong J, Haass NK, Sproesser K, Li L, Smalley KS, Fong D, Zhu YL, Marimuthu A, Nguyen H, Lam B, Liu J, Cheung I, Rice J, Suzuki Y, Luu C, Settachatgul C, Shellooe R, Cantwell J, Kim SH, Schlessinger J, Zhang KY, West BL, Powell B, Habets G, Zhang C, Ibrahim PN, Hirth P, Artis DR, Herlyn M, Bollag G.; ''Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity.''; PubMed Europe PMC Scholia
  189. Rochman Y, Spolski R, Leonard WJ.; ''New insights into the regulation of T cells by gamma(c) family cytokines.''; PubMed Europe PMC Scholia
  190. Sun H, King AJ, Diaz HB, Marshall MS.; ''Regulation of the protein kinase Raf-1 by oncogenic Ras through phosphatidylinositol 3-kinase, Cdc42/Rac and Pak.''; PubMed Europe PMC Scholia
  191. Zhu J, Balan V, Bronisz A, Balan K, Sun H, Leicht DT, Luo Z, Qin J, Avruch J, Tzivion G.; ''Identification of Raf-1 S471 as a novel phosphorylation site critical for Raf-1 and B-Raf kinase activities and for MEK binding.''; PubMed Europe PMC Scholia
  192. Zhu Y, Sullivan LL, Nair SS, Williams CC, Pandey AK, Marrero L, Vadlamudi RK, Jones FE.; ''Coregulation of estrogen receptor by ERBB4/HER4 establishes a growth-promoting autocrine signal in breast tumor cells.''; PubMed Europe PMC Scholia
  193. Denley A, Kang S, Karst U, Vogt PK.; ''Oncogenic signaling of class I PI3K isoforms.''; PubMed Europe PMC Scholia
  194. Matheny SA, Chen C, Kortum RL, Razidlo GL, Lewis RE, White MA.; ''Ras regulates assembly of mitogenic signalling complexes through the effector protein IMP.''; PubMed Europe PMC Scholia
  195. Walmod PS, Kolkova K, Berezin V, Bock E.; ''Zippers make signals: NCAM-mediated molecular interactions and signal transduction.''; PubMed Europe PMC Scholia
  196. Povlsen GK, Ditlevsen DK, Berezin V, Bock E.; ''Intracellular signaling by the neural cell adhesion molecule.''; PubMed Europe PMC Scholia
  197. Marconcini R, Galli L, Antonuzzo A, Bursi S, Roncella C, Fontanini G, Sensi E, Falcone A.; ''Metastatic BRAF K601E-mutated melanoma reaches complete response to MEK inhibitor trametinib administered for over 36 months.''; PubMed Europe PMC Scholia
  198. Lloyd AC.; ''Distinct functions for ERKs?''; PubMed Europe PMC Scholia
  199. Chadee DN, Xu D, Hung G, Andalibi A, Lim DJ, Luo Z, Gutmann DH, Kyriakis JM.; ''Mixed-lineage kinase 3 regulates B-Raf through maintenance of the B-Raf/Raf-1 complex and inhibition by the NF2 tumor suppressor protein.''; PubMed Europe PMC Scholia
  200. Penington DJ, Bryant I, Riese DJ.; ''Constitutively active ErbB4 and ErbB2 mutants exhibit distinct biological activities.''; PubMed Europe PMC Scholia
  201. Brunet A, Roux D, Lenormand P, Dowd S, Keyse S, Pouysségur J.; ''Nuclear translocation of p42/p44 mitogen-activated protein kinase is required for growth factor-induced gene expression and cell cycle entry.''; PubMed Europe PMC Scholia
  202. Wagle N, Emery C, Berger MF, Davis MJ, Sawyer A, Pochanard P, Kehoe SM, Johannessen CM, Macconaill LE, Hahn WC, Meyerson M, Garraway LA.; ''Dissecting therapeutic resistance to RAF inhibition in melanoma by tumor genomic profiling.''; PubMed Europe PMC Scholia
  203. Chen KE, Lin SY, Wu MJ, Ho MR, Santhanam A, Chou CC, Meng TC, Wang AH.; ''Reciprocal allosteric regulation of p38γ and PTPN3 involves a PDZ domain-modulated complex formation.''; PubMed Europe PMC Scholia
  204. Casar B, Pinto A, Crespo P.; ''ERK dimers and scaffold proteins: unexpected partners for a forgotten (cytoplasmic) task.''; PubMed Europe PMC Scholia
  205. Sofroniew MV, Howe CL, Mobley WC.; ''Nerve growth factor signaling, neuroprotection, and neural repair.''; PubMed Europe PMC Scholia
  206. Wolthuis RM, Bauer B, van 't Veer LJ, de Vries-Smits AM, Cool RH, Spaargaren M, Wittinghofer A, Burgering BM, Bos JL.; ''RalGDS-like factor (Rlf) is a novel Ras and Rap 1A-associating protein.''; PubMed Europe PMC Scholia
  207. Omerovic J, Puggioni EM, Napoletano S, Visco V, Fraioli R, Frati L, Gulino A, Alimandi M.; ''Ligand-regulated association of ErbB-4 to the transcriptional co-activator YAP65 controls transcription at the nuclear level.''; PubMed Europe PMC Scholia
  208. White MF, Kahn CR.; ''The insulin signaling system.''; PubMed Europe PMC Scholia
  209. Blake JF, Burkard M, Chan J, Chen H, Chou KJ, Diaz D, Dudley DA, Gaudino JJ, Gould SE, Grina J, Hunsaker T, Liu L, Martinson M, Moreno D, Mueller L, Orr C, Pacheco P, Qin A, Rasor K, Ren L, Robarge K, Shahidi-Latham S, Stults J, Sullivan F, Wang W, Yin J, Zhou A, Belvin M, Merchant M, Moffat J, Schwarz JB.; ''Discovery of (S)-1-(1-(4-Chloro-3-fluorophenyl)-2-hydroxyethyl)-4-(2-((1-methyl-1H-pyrazol-5-yl)amino)pyrimidin-4-yl)pyridin-2(1H)-one (GDC-0994), an Extracellular Signal-Regulated Kinase 1/2 (ERK1/2) Inhibitor in Early Clinical Development.''; PubMed Europe PMC Scholia
  210. Yamanashi Y, Kakiuchi T, Mizuguchi J, Yamamoto T, Toyoshima K.; ''Association of B cell antigen receptor with protein tyrosine kinase Lyn.''; PubMed Europe PMC Scholia
  211. Fredriksson L, Li H, Eriksson U.; ''The PDGF family: four gene products form five dimeric isoforms.''; PubMed Europe PMC Scholia
  212. Therrien M, Michaud NR, Rubin GM, Morrison DK.; ''KSR modulates signal propagation within the MAPK cascade.''; PubMed Europe PMC Scholia
  213. Pavelić J, Matijević T, Knezević J.; ''Biological & physiological aspects of action of insulin-like growth factor peptide family.''; PubMed Europe PMC Scholia
  214. Martini M, De Santis MC, Braccini L, Gulluni F, Hirsch E.; ''PI3K/AKT signaling pathway and cancer: an updated review.''; PubMed Europe PMC Scholia
  215. Nazarian R, Shi H, Wang Q, Kong X, Koya RC, Lee H, Chen Z, Lee MK, Attar N, Sazegar H, Chodon T, Nelson SF, McArthur G, Sosman JA, Ribas A, Lo RS.; ''Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation.''; PubMed Europe PMC Scholia
  216. Karoulia Z, Gavathiotis E, Poulikakos PI.; ''New perspectives for targeting RAF kinase in human cancer.''; PubMed Europe PMC Scholia
  217. Carraway KL, Weber JL, Unger MJ, Ledesma J, Yu N, Gassmann M, Lai C.; ''Neuregulin-2, a new ligand of ErbB3/ErbB4-receptor tyrosine kinases.''; PubMed Europe PMC Scholia
  218. Marais R, Light Y, Paterson HF, Marshall CJ.; ''Ras recruits Raf-1 to the plasma membrane for activation by tyrosine phosphorylation.''; PubMed Europe PMC Scholia
  219. Seger R, Seger D, Reszka AA, Munar ES, Eldar-Finkelman H, Dobrowolska G, Jensen AM, Campbell JS, Fischer EH, Krebs EG.; ''Overexpression of mitogen-activated protein kinase kinase (MAPKK) and its mutants in NIH 3T3 cells. Evidence that MAPKK involvement in cellular proliferation is regulated by phosphorylation of serine residues in its kinase subdomains VII and VIII.''; PubMed Europe PMC Scholia
  220. Muñoz JJ, Tárrega C, Blanco-Aparicio C, Pulido R.; ''Differential interaction of the tyrosine phosphatases PTP-SL, STEP and HePTP with the mitogen-activated protein kinases ERK1/2 and p38alpha is determined by a kinase specificity sequence and influenced by reducing agents.''; PubMed Europe PMC Scholia
  221. Schlessinger J.; ''Common and distinct elements in cellular signaling via EGF and FGF receptors.''; PubMed Europe PMC Scholia
  222. Woo RS, Lee JH, Kim HS, Baek CH, Song DY, Suh YH, Baik TK.; ''Neuregulin-1 protects against neurotoxicities induced by Swedish amyloid precursor protein via the ErbB4 receptor.''; PubMed Europe PMC Scholia
  223. Mori Y, Wakamori M, Miyakawa T, Hermosura M, Hara Y, Nishida M, Hirose K, Mizushima A, Kurosaki M, Mori E, Gotoh K, Okada T, Fleig A, Penner R, Iino M, Kurosaki T.; ''Transient receptor potential 1 regulates capacitative Ca(2+) entry and Ca(2+) release from endoplasmic reticulum in B lymphocytes.''; PubMed Europe PMC Scholia
  224. Roberts PJ, Der CJ.; ''Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer.''; PubMed Europe PMC Scholia
  225. Ornitz DM, Marie PJ.; ''FGF signaling pathways in endochondral and intramembranous bone development and human genetic disease.''; PubMed Europe PMC Scholia
  226. Chapman PB, Hauschild A, Robert C, Haanen JB, Ascierto P, Larkin J, Dummer R, Garbe C, Testori A, Maio M, Hogg D, Lorigan P, Lebbe C, Jouary T, Schadendorf D, Ribas A, O'Day SJ, Sosman JA, Kirkwood JM, Eggermont AM, Dreno B, Nolop K, Li J, Nelson B, Hou J, Lee RJ, Flaherty KT, McArthur GA, BRIM-3 Study Group.; ''Improved survival with vemurafenib in melanoma with BRAF V600E mutation.''; PubMed Europe PMC Scholia
  227. Rath O, Park S, Tang HH, Banfield MJ, Brady RL, Lee YC, Dignam JD, Sedivy JM, Kolch W, Yeung KC.; ''The RKIP (Raf-1 Kinase Inhibitor Protein) conserved pocket binds to the phosphorylated N-region of Raf-1 and inhibits the Raf-1-mediated activated phosphorylation of MEK.''; PubMed Europe PMC Scholia
  228. Rommel C, Radziwill G, Lovrić J, Noeldeke J, Heinicke T, Jones D, Aitken A, Moelling K.; ''Activated Ras displaces 14-3-3 protein from the amino terminus of c-Raf-1.''; PubMed Europe PMC Scholia
  229. Kato R, Nonami A, Taketomi T, Wakioka T, Kuroiwa A, Matsuda Y, Yoshimura A.; ''Molecular cloning of mammalian Spred-3 which suppresses tyrosine kinase-mediated Erk activation.''; PubMed Europe PMC Scholia
  230. Guan YF, Wu CY, Fang YY, Zeng YN, Luo ZY, Li SJ, Li XW, Zhu XH, Mei L, Gao TM.; ''Neuregulin 1 protects against ischemic brain injury via ErbB4 receptors by increasing GABAergic transmission.''; PubMed Europe PMC Scholia
  231. Philips MR.; ''Sef: a MEK/ERK catcher on the Golgi.''; PubMed Europe PMC Scholia
  232. Ren JG, Li Z, Sacks DB.; ''IQGAP1 modulates activation of B-Raf.''; PubMed Europe PMC Scholia
  233. Kaplan DR, Miller FD.; ''Neurotrophin signal transduction in the nervous system.''; PubMed Europe PMC Scholia
  234. Chowdhury I, Thompson WE, Thomas K.; ''Prohibitins role in cellular survival through Ras-Raf-MEK-ERK pathway.''; PubMed Europe PMC Scholia
  235. Bollag G, McCormick F.; ''Ras regulation. NF is enough of GAP.''; PubMed Europe PMC Scholia
  236. Masson K, Rönnstrand L.; ''Oncogenic signaling from the hematopoietic growth factor receptors c-Kit and Flt3.''; PubMed Europe PMC Scholia
  237. Yang X, Lee WH, Sobott F, Papagrigoriou E, Robinson CV, Grossmann JG, Sundström M, Doyle DA, Elkins JM.; ''Structural basis for protein-protein interactions in the 14-3-3 protein family.''; PubMed Europe PMC Scholia
  238. Rauen KA, Tidyman WE, Estep AL, Sampath S, Peltier HM, Bale SJ, Lacassie Y.; ''Molecular and functional analysis of a novel MEK2 mutation in cardio-facio-cutaneous syndrome: transmission through four generations.''; PubMed Europe PMC Scholia
  239. Pacold ME, Suire S, Perisic O, Lara-Gonzalez S, Davis CT, Walker EH, Hawkins PT, Stephens L, Eccleston JF, Williams RL.; ''Crystal structure and functional analysis of Ras binding to its effector phosphoinositide 3-kinase gamma.''; PubMed Europe PMC Scholia
  240. Hofer F, Fields S, Schneider C, Martin GS.; ''Activated Ras interacts with the Ral guanine nucleotide dissociation stimulator.''; PubMed Europe PMC Scholia
  241. King JA, Straffon AF, D'Abaco GM, Poon CL, I ST, Smith CM, Buchert M, Corcoran NM, Hall NE, Callus BA, Sarcevic B, Martin D, Lock P, Hovens CM.; ''Distinct requirements for the Sprouty domain for functional activity of Spred proteins.''; PubMed Europe PMC Scholia
  242. Chen PC, Yin J, Yu HW, Yuan T, Fernandez M, Yung CK, Trinh QM, Peltekova VD, Reid JG, Tworog-Dube E, Morgan MB, Muzny DM, Stein L, McPherson JD, Roberts AE, Gibbs RA, Neel BG, Kucherlapati R.; ''Next-generation sequencing identifies rare variants associated with Noonan syndrome.''; PubMed Europe PMC Scholia
  243. Riese DJ, Komurasaki T, Plowman GD, Stern DF.; ''Activation of ErbB4 by the bifunctional epidermal growth factor family hormone epiregulin is regulated by ErbB2.''; PubMed Europe PMC Scholia
  244. Sanchez M, Misulovin Z, Burkhardt AL, Mahajan S, Costa T, Franke R, Bolen JB, Nussenzweig M.; ''Signal transduction by immunoglobulin is mediated through Ig alpha and Ig beta.''; PubMed Europe PMC Scholia
  245. Flaswinkel H, Reth M.; ''Dual role of the tyrosine activation motif of the Ig-alpha protein during signal transduction via the B cell antigen receptor.''; PubMed Europe PMC Scholia
  246. Fan F, Feng L, He J, Wang X, Jiang X, Zhang Y, Wang Z, Chen Y.; ''RKTG sequesters B-Raf to the Golgi apparatus and inhibits the proliferation and tumorigenicity of human malignant melanoma cells.''; PubMed Europe PMC Scholia
  247. Furie B, Furie BC.; ''The molecular basis of platelet and endothelial cell interaction with neutrophils and monocytes: role of P-selectin and the P-selectin ligand, PSGL-1.''; PubMed Europe PMC Scholia
  248. Matheny SA, White MA.; ''Signaling threshold regulation by the Ras effector IMP.''; PubMed Europe PMC Scholia
  249. Therrien M, Chang HC, Solomon NM, Karim FD, Wassarman DA, Rubin GM.; ''KSR, a novel protein kinase required for RAS signal transduction.''; PubMed Europe PMC Scholia
  250. Young LC, Hartig N, Muñoz-Alegre M, Oses-Prieto JA, Durdu S, Bender S, Vijayakumar V, Vietri Rudan M, Gewinner C, Henderson S, Jathoul AP, Ghatrora R, Lythgoe MF, Burlingame AL, Rodriguez-Viciana P.; ''An MRAS, SHOC2, and SCRIB complex coordinates ERK pathway activation with polarity and tumorigenic growth.''; PubMed Europe PMC Scholia
  251. Guthridge MA, Stomski FC, Thomas D, Woodcock JM, Bagley CJ, Berndt MC, Lopez AF.; ''Mechanism of activation of the GM-CSF, IL-3, and IL-5 family of receptors.''; PubMed Europe PMC Scholia
  252. Ory S, Zhou M, Conrads TP, Veenstra TD, Morrison DK.; ''Protein phosphatase 2A positively regulates Ras signaling by dephosphorylating KSR1 and Raf-1 on critical 14-3-3 binding sites.''; PubMed Europe PMC Scholia
  253. Xu Be, Wilsbacher JL, Collisson T, Cobb MH.; ''The N-terminal ERK-binding site of MEK1 is required for efficient feedback phosphorylation by ERK2 in vitro and ERK activation in vivo.''; PubMed Europe PMC Scholia
  254. Scheffzek K, Ahmadian MR, Kabsch W, Wiesmüller L, Lautwein A, Schmitz F, Wittinghofer A.; ''The Ras-RasGAP complex: structural basis for GTPase activation and its loss in oncogenic Ras mutants.''; PubMed Europe PMC Scholia
  255. Flaherty KT, Puzanov I, Kim KB, Ribas A, McArthur GA, Sosman JA, O'Dwyer PJ, Lee RJ, Grippo JF, Nolop K, Chapman PB.; ''Inhibition of mutated, activated BRAF in metastatic melanoma.''; PubMed Europe PMC Scholia
  256. Siddle K.; ''Molecular basis of signaling specificity of insulin and IGF receptors: neglected corners and recent advances.''; PubMed Europe PMC Scholia
  257. Mace PD, Wallez Y, Egger MF, Dobaczewska MK, Robinson H, Pasquale EB, Riedl SJ.; ''Structure of ERK2 bound to PEA-15 reveals a mechanism for rapid release of activated MAPK.''; PubMed Europe PMC Scholia
  258. Tomasello E, Vivier E.; ''KARAP/DAP12/TYROBP: three names and a multiplicity of biological functions.''; PubMed Europe PMC Scholia
  259. Eisenhardt AE, Olbrich H, Röring M, Janzarik W, Anh TN, Cin H, Remke M, Witt H, Korshunov A, Pfister SM, Omran H, Brummer T.; ''Functional characterization of a BRAF insertion mutant associated with pilocytic astrocytoma.''; PubMed Europe PMC Scholia
  260. Tran NH, Wu X, Frost JA.; ''B-Raf and Raf-1 are regulated by distinct autoregulatory mechanisms.''; PubMed Europe PMC Scholia
  261. Weber CK, Slupsky JR, Kalmes HA, Rapp UR.; ''Active Ras induces heterodimerization of cRaf and BRaf.''; PubMed Europe PMC Scholia
  262. Zhang M, Jang H, Nussinov R.; ''The structural basis for Ras activation of PI3Kα lipid kinase.''; PubMed Europe PMC Scholia
  263. Kikuchi A, Demo SD, Ye ZH, Chen YW, Williams LT.; ''ralGDS family members interact with the effector loop of ras p21.''; PubMed Europe PMC Scholia
  264. Van Allen EM, Wagle N, Sucker A, Treacy DJ, Johannessen CM, Goetz EM, Place CS, Taylor-Weiner A, Whittaker S, Kryukov GV, Hodis E, Rosenberg M, McKenna A, Cibulskis K, Farlow D, Zimmer L, Hillen U, Gutzmer R, Goldinger SM, Ugurel S, Gogas HJ, Egberts F, Berking C, Trefzer U, Loquai C, Weide B, Hassel JC, Gabriel SB, Carter SL, Getz G, Garraway LA, Schadendorf D, Dermatologic Cooperative Oncology Group of Germany (DeCOG).; ''The genetic landscape of clinical resistance to RAF inhibition in metastatic melanoma.''; PubMed Europe PMC Scholia
  265. Wakioka T, Sasaki A, Kato R, Shouda T, Matsumoto A, Miyoshi K, Tsuneoka M, Komiya S, Baron R, Yoshimura A.; ''Spred is a Sprouty-related suppressor of Ras signalling.''; PubMed Europe PMC Scholia
  266. Wu LC.; ''Immunoglobulin E receptor signaling and asthma.''; PubMed Europe PMC Scholia
  267. Hou XJ, Ni KM, Yang JM, Li XM.; ''Neuregulin 1/ErbB4 enhances synchronized oscillations of prefrontal cortex neurons via inhibitory synapses.''; PubMed Europe PMC Scholia
  268. Voges D, Zwickl P, Baumeister W.; ''The 26S proteasome: a molecular machine designed for controlled proteolysis.''; PubMed Europe PMC Scholia
  269. Torii S, Kusakabe M, Yamamoto T, Maekawa M, Nishida E.; ''Sef is a spatial regulator for Ras/MAP kinase signaling.''; PubMed Europe PMC Scholia
  270. Hayes SD, Liu H, MacDonald E, Sanderson CM, Coulson JM, Clague MJ, Urbé S.; ''Direct and indirect control of mitogen-activated protein kinase pathway-associated components, BRAP/IMP E3 ubiquitin ligase and CRAF/RAF1 kinase, by the deubiquitylating enzyme USP15.''; PubMed Europe PMC Scholia
  271. Lefloch R, Pouysségur J, Lenormand P.; ''Total ERK1/2 activity regulates cell proliferation.''; PubMed Europe PMC Scholia
  272. Gysin S, Salt M, Young A, McCormick F.; ''Therapeutic strategies for targeting ras proteins.''; PubMed Europe PMC Scholia
  273. Teis D, Taub N, Kurzbauer R, Hilber D, de Araujo ME, Erlacher M, Offterdinger M, Villunger A, Geley S, Bohn G, Klein C, Hess MW, Huber LA.; ''p14-MP1-MEK1 signaling regulates endosomal traffic and cellular proliferation during tissue homeostasis.''; PubMed Europe PMC Scholia
  274. Lennartsson J, Rönnstrand L.; ''The stem cell factor receptor/c-Kit as a drug target in cancer.''; PubMed Europe PMC Scholia
  275. Diaz B, Barnard D, Filson A, MacDonald S, King A, Marshall M.; ''Phosphorylation of Raf-1 serine 338-serine 339 is an essential regulatory event for Ras-dependent activation and biological signaling.''; PubMed Europe PMC Scholia
  276. Elenius K, Paul S, Allison G, Sun J, Klagsbrun M.; ''Activation of HER4 by heparin-binding EGF-like growth factor stimulates chemotaxis but not proliferation.''; PubMed Europe PMC Scholia
  277. Li Z, Mei Y, Liu X, Zhou M.; ''Neuregulin-1 only induces trans-phosphorylation between ErbB receptor heterodimer partners.''; PubMed Europe PMC Scholia
  278. Luik RM, Wang B, Prakriya M, Wu MM, Lewis RS.; ''Oligomerization of STIM1 couples ER calcium depletion to CRAC channel activation.''; PubMed Europe PMC Scholia
  279. Kiryushko D, Berezin V, Bock E.; ''Regulators of neurite outgrowth: role of cell adhesion molecules.''; PubMed Europe PMC Scholia
  280. Ni CY, Murphy MP, Golde TE, Carpenter G.; ''gamma -Secretase cleavage and nuclear localization of ErbB-4 receptor tyrosine kinase.''; PubMed Europe PMC Scholia
  281. Nikolaev SI, Rimoldi D, Iseli C, Valsesia A, Robyr D, Gehrig C, Harshman K, Guipponi M, Bukach O, Zoete V, Michielin O, Muehlethaler K, Speiser D, Beckmann JS, Xenarios I, Halazonetis TD, Jongeneel CV, Stevenson BJ, Antonarakis SE.; ''Exome sequencing identifies recurrent somatic MAP2K1 and MAP2K2 mutations in melanoma.''; PubMed Europe PMC Scholia
  282. Krauthammer M, Kong Y, Bacchiocchi A, Evans P, Pornputtapong N, Wu C, McCusker JP, Ma S, Cheng E, Straub R, Serin M, Bosenberg M, Ariyan S, Narayan D, Sznol M, Kluger HM, Mane S, Schlessinger J, Lifton RP, Halaban R.; ''Exome sequencing identifies recurrent mutations in NF1 and RASopathy genes in sun-exposed melanomas.''; PubMed Europe PMC Scholia
  283. Fu SM, Winchester RJ, Kunkel HG.; ''Occurrence of surface IgM, IgD, and free light chains of human lymphocytes.''; PubMed Europe PMC Scholia
  284. Roskoski R.; ''RAF protein-serine/threonine kinases: structure and regulation.''; PubMed Europe PMC Scholia
  285. 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
  286. Saouaf SJ, Kut SA, Fargnoli J, Rowley RB, Bolen JB, Mahajan S.; ''Reconstitution of the B cell antigen receptor signaling components in COS cells.''; PubMed Europe PMC Scholia
  287. Turnbull IR, Colonna M.; ''Activating and inhibitory functions of DAP12.''; PubMed Europe PMC Scholia
  288. Panicker AK, Buhusi M, Thelen K, Maness PF.; ''Cellular signalling mechanisms of neural cell adhesion molecules.''; PubMed Europe PMC Scholia
  289. Naresh A, Long W, Vidal GA, Wimley WC, Marrero L, Sartor CI, Tovey S, Cooke TG, Bartlett JM, Jones FE.; ''The ERBB4/HER4 intracellular domain 4ICD is a BH3-only protein promoting apoptosis of breast cancer cells.''; PubMed Europe PMC Scholia
  290. Tzahar E, Levkowitz G, Karunagaran D, Yi L, Peles E, Lavi S, Chang D, Liu N, Yayon A, Wen D.; ''ErbB-3 and ErbB-4 function as the respective low and high affinity receptors of all Neu differentiation factor/heregulin isoforms.''; PubMed Europe PMC Scholia
  291. Nel AE, Landreth GE, Goldschmidt-Clermont PJ, Tung HE, Galbraith RM.; ''Enhanced tyrosine phosphorylation in B lymphocytes upon complexing of membrane immunoglobulin.''; PubMed Europe PMC Scholia
  292. Yeung K, Janosch P, McFerran B, Rose DW, Mischak H, Sedivy JM, Kolch W.; ''Mechanism of suppression of the Raf/MEK/extracellular signal-regulated kinase pathway by the raf kinase inhibitor protein.''; PubMed Europe PMC Scholia
  293. Zhang BH, Guan KL.; ''Activation of B-Raf kinase requires phosphorylation of the conserved residues Thr598 and Ser601.''; PubMed Europe PMC Scholia
  294. Pei D, Chen K, Liao H.; ''Targeting Ras with Macromolecules.''; PubMed Europe PMC Scholia
  295. Adachi M, Fukuda M, Nishida E.; ''Two co-existing mechanisms for nuclear import of MAP kinase: passive diffusion of a monomer and active transport of a dimer.''; PubMed Europe PMC Scholia
  296. Rio C, Buxbaum JD, Peschon JJ, Corfas G.; ''Tumor necrosis factor-alpha-converting enzyme is required for cleavage of erbB4/HER4.''; PubMed Europe PMC Scholia
  297. Kim YJ, Sekiya F, Poulin B, Bae YS, Rhee SG.; ''Mechanism of B-cell receptor-induced phosphorylation and activation of phospholipase C-gamma2.''; PubMed Europe PMC Scholia
  298. Sim GC, Radvanyi L.; ''The IL-2 cytokine family in cancer immunotherapy.''; PubMed Europe PMC Scholia
  299. Henry JR, Kaufman MD, Peng SB, Ahn YM, Caldwell TM, Vogeti L, Telikepalli H, Lu WP, Hood MM, Rutkoski TJ, Smith BD, Vogeti S, Miller D, Wise SC, Chun L, Zhang X, Zhang Y, Kays L, Hipskind PA, Wrobleski AD, Lobb KL, Clay JM, Cohen JD, Walgren JL, McCann D, Patel P, Clawson DK, Guo S, Manglicmot D, Groshong C, Logan C, Starling JJ, Flynn DL.; ''Discovery of 1-(3,3-dimethylbutyl)-3-(2-fluoro-4-methyl-5-(7-methyl-2-(methylamino)pyrido[2,3-d]pyrimidin-6-yl)phenyl)urea (LY3009120) as a pan-RAF inhibitor with minimal paradoxical activation and activity against BRAF or RAS mutant tumor cells.''; PubMed Europe PMC Scholia
  300. Genau HM, Huber J, Baschieri F, Akutsu M, Dötsch V, Farhan H, Rogov V, Behrends C.; ''CUL3-KBTBD6/KBTBD7 ubiquitin ligase cooperates with GABARAP proteins to spatially restrict TIAM1-RAC1 signaling.''; PubMed Europe PMC Scholia
  301. Hou S, Suresh PS, Qi X, Lepp A, Mirza SP, Chen G.; ''p38γ Mitogen-activated protein kinase signals through phosphorylating its phosphatase PTPH1 in regulating ras protein oncogenesis and stress response.''; PubMed Europe PMC Scholia
  302. Lorenz K, Schmid E, Deiss K.; ''RKIP: a governor of intracellular signaling.''; PubMed Europe PMC Scholia
  303. Roy M, Li Z, Sacks DB.; ''IQGAP1 binds ERK2 and modulates its activity.''; PubMed Europe PMC Scholia
  304. Sosman JA, Kim KB, Schuchter L, Gonzalez R, Pavlick AC, Weber JS, McArthur GA, Hutson TE, Moschos SJ, Flaherty KT, Hersey P, Kefford R, Lawrence D, Puzanov I, Lewis KD, Amaravadi RK, Chmielowski B, Lawrence HJ, Shyr Y, Ye F, Li J, Nolop KB, Lee RJ, Joe AK, Ribas A.; ''Survival in BRAF V600-mutant advanced melanoma treated with vemurafenib.''; PubMed Europe PMC Scholia
  305. Kaushansky A, Gordus A, Budnik BA, Lane WS, Rush J, MacBeath G.; ''System-wide investigation of ErbB4 reveals 19 sites of Tyr phosphorylation that are unusually selective in their recruitment properties.''; PubMed Europe PMC Scholia
  306. Kyriakis JM, Avruch J.; ''Mammalian MAPK signal transduction pathways activated by stress and inflammation: a 10-year update.''; PubMed Europe PMC Scholia
  307. Ishii N, Harada N, Joseph EW, Ohara K, Miura T, Sakamoto H, Matsuda Y, Tomii Y, Tachibana-Kondo Y, Iikura H, Aoki T, Shimma N, Arisawa M, Sowa Y, Poulikakos PI, Rosen N, Aoki Y, Sakai T.; ''Enhanced inhibition of ERK signaling by a novel allosteric MEK inhibitor, CH5126766, that suppresses feedback reactivation of RAF activity.''; PubMed Europe PMC Scholia
  308. Ahearn I, Zhou M, Philips MR.; ''Posttranslational Modifications of RAS Proteins.''; PubMed Europe PMC Scholia
  309. Edling CE, Hallberg B.; ''c-Kit--a hematopoietic cell essential receptor tyrosine kinase.''; PubMed Europe PMC Scholia
  310. 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
  311. Goetz CA, O'Neil JJ, Farrar MA.; ''Membrane localization, oligomerization, and phosphorylation are required for optimal raf activation.''; PubMed Europe PMC Scholia
  312. Burack WR, Shaw AS.; ''Live Cell Imaging of ERK and MEK: simple binding equilibrium explains the regulated nucleocytoplasmic distribution of ERK.''; PubMed Europe PMC Scholia
  313. Aqeilan RI, Donati V, Palamarchuk A, Trapasso F, Kaou M, Pekarsky Y, Sudol M, Croce CM.; ''WW domain-containing proteins, WWOX and YAP, compete for interaction with ErbB-4 and modulate its transcriptional function.''; PubMed Europe PMC Scholia
  314. Brezski RJ, Monroe JG.; ''B-cell receptor.''; PubMed Europe PMC Scholia
  315. Hennig A, Markwart R, Esparza-Franco MA, Ladds G, Rubio I.; ''Ras activation revisited: role of GEF and GAP systems.''; PubMed Europe PMC Scholia
  316. Huang EJ, Reichardt LF.; ''Trk receptors: roles in neuronal signal transduction.''; PubMed Europe PMC Scholia
  317. Parrella E, Longo VD.; ''Insulin/IGF-I and related signaling pathways regulate aging in nondividing cells: from yeast to the mammalian brain.''; PubMed Europe PMC Scholia
  318. Akdis M, Burgler S, Crameri R, Eiwegger T, Fujita H, Gomez E, Klunker S, Meyer N, O'Mahony L, Palomares O, Rhyner C, Ouaked N, Schaffartzik A, Van De Veen W, Zeller S, Zimmermann M, Akdis CA.; ''Interleukins, from 1 to 37, and interferon-γ: receptors, functions, and roles in diseases.''; PubMed Europe PMC Scholia
  319. Tessarz AS, Cerwenka A.; ''The TREM-1/DAP12 pathway.''; PubMed Europe PMC Scholia
  320. Tsang M, Friesel R, Kudoh T, Dawid IB.; ''Identification of Sef, a novel modulator of FGF signalling.''; PubMed Europe PMC Scholia
  321. Estep AL, Palmer C, McCormick F, Rauen KA.; ''Mutation analysis of BRAF, MEK1 and MEK2 in 15 ovarian cancer cell lines: implications for therapy.''; PubMed Europe PMC Scholia
  322. Fürthauer M, Lin W, Ang SL, Thisse B, Thisse C.; ''Sef is a feedback-induced antagonist of Ras/MAPK-mediated FGF signalling.''; PubMed Europe PMC Scholia
  323. Dougherty MK, Müller J, Ritt DA, Zhou M, Zhou XZ, Copeland TD, Conrads TP, Veenstra TD, Lu KP, Morrison DK.; ''Regulation of Raf-1 by direct feedback phosphorylation.''; PubMed Europe PMC Scholia
  324. Kondoh K, Nishida E.; ''Regulation of MAP kinases by MAP kinase phosphatases.''; PubMed Europe PMC Scholia
  325. Raabe T, Rapp UR.; ''Ras signaling: PP2A puts Ksr and Raf in the right place.''; PubMed Europe PMC Scholia
  326. Riese DJ, van Raaij TM, Plowman GD, Andrews GC, Stern DF.; ''The cellular response to neuregulins is governed by complex interactions of the erbB receptor family.''; PubMed Europe PMC Scholia
  327. Samatar AA, Poulikakos PI.; ''Targeting RAS-ERK signalling in cancer: promises and challenges.''; PubMed Europe PMC Scholia
  328. Matallanas D, Birtwistle M, Romano D, Zebisch A, Rauch J, von Kriegsheim A, Kolch W.; ''Raf family kinases: old dogs have learned new tricks.''; PubMed Europe PMC Scholia
  329. Roy M, Li Z, Sacks DB.; ''IQGAP1 is a scaffold for mitogen-activated protein kinase signaling.''; PubMed Europe PMC Scholia
  330. Catalanotti F, Reyes G, Jesenberger V, Galabova-Kovacs G, de Matos Simoes R, Carugo O, Baccarini M.; ''A Mek1-Mek2 heterodimer determines the strength and duration of the Erk signal.''; PubMed Europe PMC Scholia
  331. Geng F, Zhang J, Wu JL, Zou WJ, Liang ZP, Bi LL, Liu JH, Kong Y, Huang CQ, Li XW, Yang JM, Gao TM.; ''Neuregulin 1-ErbB4 signaling in the bed nucleus of the stria terminalis regulates anxiety-like behavior.''; PubMed Europe PMC Scholia
  332. Sweeney C, Lai C, Riese DJ, Diamonti AJ, Cantley LC, Carraway KL.; ''Ligand discrimination in signaling through an ErbB4 receptor homodimer.''; PubMed Europe PMC Scholia
  333. Rajakulendran T, Sahmi M, Lefrançois M, Sicheri F, Therrien M.; ''A dimerization-dependent mechanism drives RAF catalytic activation.''; PubMed Europe PMC Scholia
  334. Terai K, Matsuda M.; ''Ras binding opens c-Raf to expose the docking site for mitogen-activated protein kinase kinase.''; PubMed Europe PMC Scholia
  335. Marks JL, Gong Y, Chitale D, Golas B, McLellan MD, Kasai Y, Ding L, Mardis ER, Wilson RK, Solit D, Levine R, Michel K, Thomas RK, Rusch VW, Ladanyi M, Pao W.; ''Novel MEK1 mutation identified by mutational analysis of epidermal growth factor receptor signaling pathway genes in lung adenocarcinoma.''; PubMed Europe PMC Scholia
  336. Rodriguez-Viciana P, Warne PH, Vanhaesebroeck B, Waterfield MD, Downward J.; ''Activation of phosphoinositide 3-kinase by interaction with Ras and by point mutation.''; PubMed Europe PMC Scholia
  337. Gentry LR, Martin TD, Reiner DJ, Der CJ.; ''Ral small GTPase signaling and oncogenesis: More than just 15minutes of fame.''; PubMed Europe PMC Scholia
  338. Nakamura A, Arita T, Tsuchiya S, Donelan J, Chouitar J, Carideo E, Galvin K, Okaniwa M, Ishikawa T, Yoshida S.; ''Antitumor activity of the selective pan-RAF inhibitor TAK-632 in BRAF inhibitor-resistant melanoma.''; PubMed Europe PMC Scholia
  339. Lito P, Saborowski A, Yue J, Solomon M, Joseph E, Gadal S, Saborowski M, Kastenhuber E, Fellmann C, Ohara K, Morikami K, Miura T, Lukacs C, Ishii N, Lowe S, Rosen N.; ''Disruption of CRAF-mediated MEK activation is required for effective MEK inhibition in KRAS mutant tumors.''; PubMed Europe PMC Scholia
  340. Stowe IB, Mercado EL, Stowe TR, Bell EL, Oses-Prieto JA, Hernández H, Burlingame AL, McCormick F.; ''A shared molecular mechanism underlies the human rasopathies Legius syndrome and Neurofibromatosis-1.''; PubMed Europe PMC Scholia
  341. Hu J, Stites EC, Yu H, Yu H, Germino EA, Meharena HS, Stork PJS, Kornev AP, Taylor SS, Shaw AS.; ''Allosteric activation of functionally asymmetric RAF kinase dimers.''; PubMed Europe PMC Scholia
  342. Kainulainen V, Sundvall M, Määttä JA, Santiestevan E, Klagsbrun M, Elenius K.; ''A natural ErbB4 isoform that does not activate phosphoinositide 3-kinase mediates proliferation but not survival or chemotaxis.''; PubMed Europe PMC Scholia
  343. Chaikuad A, Tacconi EM, Zimmer J, Liang Y, Gray NS, Tarsounas M, Knapp S.; ''A unique inhibitor binding site in ERK1/2 is associated with slow binding kinetics.''; PubMed Europe PMC Scholia
  344. Cheng QC, Tikhomirov O, Zhou W, Carpenter G.; ''Ectodomain cleavage of ErbB-4: characterization of the cleavage site and m80 fragment.''; PubMed Europe PMC Scholia
  345. Bourne HR, Sanders DA, McCormick F.; ''The GTPase superfamily: conserved structure and molecular mechanism.''; PubMed Europe PMC Scholia
  346. Tohgo A, Choy EW, Gesty-Palmer D, Pierce KL, Laporte S, Oakley RH, Caron MG, Lefkowitz RJ, Luttrell LM.; ''The stability of the G protein-coupled receptor-beta-arrestin interaction determines the mechanism and functional consequence of ERK activation.''; PubMed Europe PMC Scholia
  347. Coggeshall KM, McHugh JC, Altman A.; ''Predominant expression and activation-induced tyrosine phosphorylation of phospholipase C-gamma 2 in B lymphocytes.''; PubMed Europe PMC Scholia
  348. Vigil D, Cherfils J, Rossman KL, Der CJ.; ''Ras superfamily GEFs and GAPs: validated and tractable targets for cancer therapy?''; PubMed Europe PMC Scholia
  349. Catling AD, Schaeffer HJ, Reuter CW, Reddy GR, Weber MJ.; ''A proline-rich sequence unique to MEK1 and MEK2 is required for raf binding and regulates MEK function.''; PubMed Europe PMC Scholia
  350. Chitnis MM, Yuen JS, Protheroe AS, Pollak M, Macaulay VM.; ''The type 1 insulin-like growth factor receptor pathway.''; PubMed Europe PMC Scholia
  351. Suire S, Hawkins P, Stephens L.; ''Activation of phosphoinositide 3-kinase gamma by Ras.''; PubMed Europe PMC Scholia
  352. Chen C, Lewis RE, White MA.; ''IMP modulates KSR1-dependent multivalent complex formation to specify ERK1/2 pathway activation and response thresholds.''; PubMed Europe PMC Scholia
  353. Bradshaw JM.; ''The Src, Syk, and Tec family kinases: distinct types of molecular switches.''; PubMed Europe PMC Scholia
  354. Gripp KW, Aldinger KA, Bennett JT, Baker L, Tusi J, Powell-Hamilton N, Stabley D, Sol-Church K, Timms AE, Dobyns WB.; ''A novel rasopathy caused by recurrent de novo missense mutations in PPP1CB closely resembles Noonan syndrome with loose anagen hair.''; PubMed Europe PMC Scholia
  355. Hu J, Yu H, Yu H, Kornev AP, Zhao J, Filbert EL, Taylor SS, Shaw AS.; ''Mutation that blocks ATP binding creates a pseudokinase stabilizing the scaffolding function of kinase suppressor of Ras, CRAF and BRAF.''; PubMed Europe PMC Scholia
  356. Lavoie H, Therrien M.; ''Regulation of RAF protein kinases in ERK signalling.''; PubMed Europe PMC Scholia
  357. Morris EJ, Jha S, Restaino CR, Dayananth P, Zhu H, Cooper A, Carr D, Deng Y, Jin W, Black S, Long B, Liu J, Dinunzio E, Windsor W, Zhang R, Zhao S, Angagaw MH, Pinheiro EM, Desai J, Xiao L, Shipps G, Hruza A, Wang J, Kelly J, Paliwal S, Gao X, Babu BS, Zhu L, Daublain P, Zhang L, Lutterbach BA, Pelletier MR, Philippar U, Siliphaivanh P, Witter D, Kirschmeier P, Bishop WR, Hicklin D, Gilliland DG, Jayaraman L, Zawel L, Fawell S, Samatar AA.; ''Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors.''; PubMed Europe PMC Scholia
  358. Shin SY, Rath O, Choo SM, Fee F, McFerran B, Kolch W, Cho KH.; ''Positive- and negative-feedback regulations coordinate the dynamic behavior of the Ras-Raf-MEK-ERK signal transduction pathway.''; PubMed Europe PMC Scholia
  359. Montagut C, Sharma SV, Shioda T, McDermott U, Ulman M, Ulkus LE, Dias-Santagata D, Stubbs H, Lee DY, Singh A, Drew L, Haber DA, Settleman J.; ''Elevated CRAF as a potential mechanism of acquired resistance to BRAF inhibition in melanoma.''; PubMed Europe PMC Scholia
  360. Teis D, Wunderlich W, Huber LA.; ''Localization of the MP1-MAPK scaffold complex to endosomes is mediated by p14 and required for signal transduction.''; PubMed Europe PMC Scholia
  361. Feng SM, Muraoka-Cook RS, Hunter D, Sandahl MA, Caskey LS, Miyazawa K, Atfi A, Earp HS.; ''The E3 ubiquitin ligase WWP1 selectively targets HER4 and its proteolytically derived signaling isoforms for degradation.''; PubMed Europe PMC Scholia
  362. Cseh B, Doma E, Baccarini M.; ''"RAF" neighborhood: protein-protein interaction in the Raf/Mek/Erk pathway.''; PubMed Europe PMC Scholia
  363. Ohren JF, Chen H, Pavlovsky A, Whitehead C, Zhang E, Kuffa P, Yan C, McConnell P, Spessard C, Banotai C, Mueller WT, Delaney A, Omer C, Sebolt-Leopold J, Dudley DT, Leung IK, Flamme C, Warmus J, Kaufman M, Barrett S, Tecle H, Hasemann CA.; ''Structures of human MAP kinase kinase 1 (MEK1) and MEK2 describe novel noncompetitive kinase inhibition.''; PubMed Europe PMC Scholia
  364. Heldin CH, Westermark B.; ''Mechanism of action and in vivo role of platelet-derived growth factor.''; PubMed Europe PMC Scholia
  365. Salzano M, Rusciano MR, Russo E, Bifulco M, Postiglione L, Vitale M.; ''Calcium/calmodulin-dependent protein kinase II (CaMKII) phosphorylates Raf-1 at serine 338 and mediates Ras-stimulated Raf-1 activation.''; PubMed Europe PMC Scholia
  366. Boga SB, Deng Y, Zhu L, Nan Y, Cooper AB, Shipps GW, Doll R, Shih NY, Zhu H, Sun R, Wang T, Paliwal S, Tsui HC, Gao X, Yao X, Desai J, Wang J, Alhassan AB, Kelly J, Patel M, Muppalla K, Gudipati S, Zhang LK, Buevich A, Hesk D, Carr D, Dayananth P, Black S, Mei H, Cox K, Sherborne B, Hruza AW, Xiao L, Jin W, Long B, Liu G, Taylor SA, Kirschmeier P, Windsor WT, Bishop R, Samatar AA.; ''MK-8353: Discovery of an Orally Bioavailable Dual Mechanism ERK Inhibitor for Oncology.''; PubMed Europe PMC Scholia
  367. Luo X, Feng L, Jiang X, Xiao F, Wang Z, Feng GS, Chen Y.; ''Characterization of the topology and functional domains of RKTG.''; PubMed Europe PMC Scholia
  368. Prior IA, Lewis PD, Mattos C.; ''A comprehensive survey of Ras mutations in cancer.''; PubMed Europe PMC Scholia
  369. Baldock D, Graham B, Akhlaq M, Graff P, Jones CE, Menear K.; ''Purification and characterization of human Syk produced using a baculovirus expression system.''; PubMed Europe PMC Scholia
  370. Herbst RS.; ''Review of epidermal growth factor receptor biology.''; PubMed Europe PMC Scholia
  371. Roskoski R.; ''MEK1/2 dual-specificity protein kinases: structure and regulation.''; PubMed Europe PMC Scholia
  372. Brunet A, Pagès G, Pouysségur J.; ''Growth factor-stimulated MAP kinase induces rapid retrophosphorylation and inhibition of MAP kinase kinase (MEK1).''; PubMed Europe PMC Scholia
  373. Crosier PS, Ricciardi ST, Hall LR, Vitas MR, Clark SC, Crosier KE.; ''Expression of isoforms of the human receptor tyrosine kinase c-kit in leukemic cell lines and acute myeloid leukemia.''; PubMed Europe PMC Scholia
  374. Chen RH, Sarnecki C, Blenis J.; ''Nuclear localization and regulation of erk- and rsk-encoded protein kinases.''; PubMed Europe PMC Scholia
  375. Muraoka-Cook RS, Caskey LS, Sandahl MA, Hunter DM, Husted C, Strunk KE, Sartor CI, Rearick WA, McCall W, Sgagias MK, Cowan KH, Earp HS.; ''Heregulin-dependent delay in mitotic progression requires HER4 and BRCA1.''; PubMed Europe PMC Scholia
  376. Zeng F, Xu J, Harris RC.; ''Nedd4 mediates ErbB4 JM-a/CYT-1 ICD ubiquitination and degradation in MDCK II cells.''; PubMed Europe PMC Scholia
  377. Lemmon MA, Schlessinger J.; ''Cell signaling by receptor tyrosine kinases.''; PubMed Europe PMC Scholia
  378. Relou IA, Bax LA, van Rijn HJ, Akkerman JW.; ''Site-specific phosphorylation of platelet focal adhesion kinase by low-density lipoprotein.''; PubMed Europe PMC Scholia
  379. Whittaker SR, Theurillat JP, Van Allen E, Wagle N, Hsiao J, Cowley GS, Schadendorf D, Root DE, Garraway LA.; ''A genome-scale RNA interference screen implicates NF1 loss in resistance to RAF inhibition.''; PubMed Europe PMC Scholia
  380. Schulze WX, Deng L, Mann M.; ''Phosphotyrosine interactome of the ErbB-receptor kinase family.''; PubMed Europe PMC Scholia
  381. Schlessinger J.; ''Ligand-induced, receptor-mediated dimerization and activation of EGF receptor.''; PubMed Europe PMC Scholia
  382. Matsubayashi Y, Fukuda M, Nishida E.; ''Evidence for existence of a nuclear pore complex-mediated, cytosol-independent pathway of nuclear translocation of ERK MAP kinase in permeabilized cells.''; PubMed Europe PMC Scholia
  383. Blank U, Rivera J.; ''The ins and outs of IgE-dependent mast-cell exocytosis.''; PubMed Europe PMC Scholia
  384. Cherfils J, Zeghouf M.; ''Regulation of small GTPases by GEFs, GAPs, and GDIs.''; PubMed Europe PMC Scholia
  385. McGillicuddy LT, Fromm JA, Hollstein PE, Kubek S, Beroukhim R, De Raedt T, Johnson BW, Williams SM, Nghiemphu P, Liau LM, Cloughesy TF, Mischel PS, Parret A, Seiler J, Moldenhauer G, Scheffzek K, Stemmer-Rachamimov AO, Sawyers CL, Brennan C, Messiaen L, Mellinghoff IK, Cichowski K.; ''Proteasomal and genetic inactivation of the NF1 tumor suppressor in gliomagenesis.''; PubMed Europe PMC Scholia
  386. Chadee DN, Kyriakis JM.; ''A novel role for mixed lineage kinase 3 (MLK3) in B-Raf activation and cell proliferation.''; PubMed Europe PMC Scholia
  387. Paatero I, Jokilammi A, Heikkinen PT, Iljin K, Kallioniemi OP, Jones FE, Jaakkola PM, Elenius K.; ''Interaction with ErbB4 promotes hypoxia-inducible factor-1α signaling.''; PubMed Europe PMC Scholia
  388. King PD, Lubeck BA, Lapinski PE.; ''Nonredundant functions for Ras GTPase-activating proteins in tissue homeostasis.''; PubMed Europe PMC Scholia
  389. Cantwell-Dorris ER, O'Leary JJ, Sheils OM.; ''BRAFV600E: implications for carcinogenesis and molecular therapy.''; PubMed Europe PMC Scholia

History

View all...
CompareRevisionActionTimeUserComment
114761view16:25, 25 January 2021ReactomeTeamReactome version 75
113205view11:27, 2 November 2020ReactomeTeamReactome version 74
112429view15:37, 9 October 2020ReactomeTeamReactome version 73
101333view11:22, 1 November 2018ReactomeTeamreactome version 66
100871view20:55, 31 October 2018ReactomeTeamreactome version 65
100412view19:28, 31 October 2018ReactomeTeamreactome version 64
99961view16:13, 31 October 2018ReactomeTeamreactome version 63
99516view14:46, 31 October 2018ReactomeTeamreactome version 62 (2nd attempt)
99158view12:41, 31 October 2018ReactomeTeamreactome version 62
93933view13:46, 16 August 2017ReactomeTeamreactome version 61
93520view11:25, 9 August 2017ReactomeTeamreactome version 61
88127view10:15, 26 July 2016RyanmillerOntology Term : 'kinase mediated signaling pathway' added !
88126view10:15, 26 July 2016RyanmillerOntology Term : 'signaling pathway' added !
86618view09:22, 11 July 2016ReactomeTeamreactome version 56
83142view10:09, 18 November 2015ReactomeTeamVersion54
81486view13:01, 21 August 2015ReactomeTeamVersion53
76963view08:24, 17 July 2014ReactomeTeamFixed remaining interactions
76668view12:03, 16 July 2014ReactomeTeamFixed remaining interactions
75998view10:05, 11 June 2014ReactomeTeamRe-fixing comment source
75701view11:04, 10 June 2014ReactomeTeamReactome 48 Update
75057view13:56, 8 May 2014AnweshaFixing comment source for displaying WikiPathways description
74701view08:46, 30 April 2014ReactomeTeamNew pathway

External references

DataNodes

View all...
NameTypeDatabase referenceComment
'activator' RAF:YWHAB dimerComplexR-HSA-5672710 (Reactome)
26S proteasomeComplexR-HSA-68819 (Reactome)
ACTN2 ProteinP35609 (Uniprot-TrEMBL)
ADPMetaboliteCHEBI:16761 (ChEBI)
AKAP9 ProteinQ99996 (Uniprot-TrEMBL)
ANGPT1 ProteinQ15389 (Uniprot-TrEMBL)
APBB1IP ProteinQ7Z5R6 (Uniprot-TrEMBL)
ARRB1 ProteinP49407 (Uniprot-TrEMBL)
ARRB2 ProteinP32121 (Uniprot-TrEMBL)
ARTN ProteinQ5T4W7 (Uniprot-TrEMBL)
ATPMetaboliteCHEBI:15422 (ChEBI)
Activated FGFR2b homodimer bound to FGF R-HSA-192606 (Reactome)
Activated FGFR2c homodimer bound to FGF R-HSA-192616 (Reactome)
BRAP ProteinQ7Z569 (Uniprot-TrEMBL)
BRAP:KSR1:MARK3ComplexR-HSA-5674011 (Reactome)
BRAPProteinQ7Z569 (Uniprot-TrEMBL)
CALM1 ProteinP62158 (Uniprot-TrEMBL)
CNKSR1 ProteinQ969H4 (Uniprot-TrEMBL)
CNKSR2 ProteinQ8WXI2 (Uniprot-TrEMBL)
CSK ProteinP41240 (Uniprot-TrEMBL)
CUL3 ProteinQ13618 (Uniprot-TrEMBL)
Ca2+ MetaboliteCHEBI:29108 (ChEBI)
Cell surface

interactions at the

vascular wall
PathwayR-HSA-202733 (Reactome) Leukocyte extravasation is a rigorously controlled process that guides white cell movement from the vascular lumen to sites of tissue inflammation. The powerful adhesive interactions that are required for leukocytes to withstand local flow at the vessel wall is a multistep process mediated by different adhesion molecules. Platelets adhered to injured vessel walls form strong adhesive substrates for leukocytes. For instance, the initial tethering and rolling of leukocytes over the site of injury are mediated by reversible binding of selectins to their cognate cell-surface glycoconjugates.

Endothelial cells are tightly connected through various proteins, which regulate the organization of the junctional complex and bind to cytoskeletal proteins or cytoplasmic interaction partners that allow the transfer of intracellular signals. An important role for these junctional proteins in governing the transendothelial migration of leukocytes under normal or inflammatory conditions has been established.

This pathway describes some of the key interactions that assist in the process of platelet and leukocyte interaction with the endothelium, in response to injury.

DAB2IP ProteinQ5VWQ8 (Uniprot-TrEMBL)
DAG MetaboliteCHEBI:17815 (ChEBI)
DAP12 interactionsPathwayR-HSA-2172127 (Reactome) DNAX activation protein of 12kDa (DAP12) is an immunoreceptor tyrosine-based activation motif (ITAM)-bearing adapter molecule that transduces activating signals in natural killer (NK) and myeloid cells. It mediates signalling for multiple cell-surface receptors expressed by these cells, associating with receptor chains through complementary charged transmembrane amino acids that form a salt-bridge in the context of the hydrophobic lipid bilayer (Lanier et al. 1998). DAP12 homodimers associate with a variety of receptors expressed by macrophages, monocytes and myeloid cells including TREM2, Siglec H and SIRP-beta, as well as activating KIR, LY49 and the NKG2C proteins expressed by NK cells. DAP12 is expressed at the cell surface, with most of the protein lying on the cytoplasmic side of the membrane (Turnbull & Colonna 2007, Tessarz & Cerwenka 2008).
DUSP1 ProteinP28562 (Uniprot-TrEMBL)
DUSP10 ProteinQ9Y6W6 (Uniprot-TrEMBL)
DUSP16 ProteinQ9BY84 (Uniprot-TrEMBL)
DUSP2 ProteinQ05923 (Uniprot-TrEMBL)
DUSP4 ProteinQ13115 (Uniprot-TrEMBL)
DUSP5 ProteinQ16690 (Uniprot-TrEMBL)
DUSP6 ProteinQ16828 (Uniprot-TrEMBL)
DUSP7 ProteinQ16829 (Uniprot-TrEMBL)
DUSP8 ProteinQ13202 (Uniprot-TrEMBL)
DUSP9 ProteinQ99956 (Uniprot-TrEMBL)
EGF ProteinP01133 (Uniprot-TrEMBL)
EGF-like ligands R-HSA-1233230 (Reactome)
F-actin R-HSA-196015 (Reactome)
FGA ProteinP02671 (Uniprot-TrEMBL)
FGB ProteinP02675 (Uniprot-TrEMBL)
FGF1 ProteinP05230 (Uniprot-TrEMBL)
FGF10 ProteinO15520 (Uniprot-TrEMBL)
FGF16 ProteinO43320 (Uniprot-TrEMBL)
FGF17-1 ProteinO60258-1 (Uniprot-TrEMBL)
FGF18 ProteinO76093 (Uniprot-TrEMBL)
FGF19 ProteinO95750 (Uniprot-TrEMBL)
FGF2(10-155) ProteinP09038 (Uniprot-TrEMBL)
FGF20 ProteinQ9NP95 (Uniprot-TrEMBL)
FGF22 ProteinQ9HCT0 (Uniprot-TrEMBL)
FGF23(25-251) ProteinQ9GZV9 (Uniprot-TrEMBL)
FGF3 ProteinP11487 (Uniprot-TrEMBL)
FGF4 ProteinP08620 (Uniprot-TrEMBL)
FGF5-1 ProteinP12034-1 (Uniprot-TrEMBL)
FGF6 ProteinP10767 (Uniprot-TrEMBL)
FGF8-1 ProteinP55075-1 (Uniprot-TrEMBL)
FGF9 ProteinP31371 (Uniprot-TrEMBL)
FGG ProteinP02679 (Uniprot-TrEMBL)
FN1(32-2386) ProteinP02751 (Uniprot-TrEMBL)
Fc epsilon receptor (FCERI) signalingPathwayR-HSA-2454202 (Reactome) Mast cells (MC) are distributed in tissues throughout the human body and have long been recognized as key cells of type I hypersensitivity reactions. They also play important roles in inflammatory and immediate allergic reactions. Activation through FCERI-bound antigen-specific IgE causes release of potent inflammatory mediators, such as histamine, proteases, chemotactic factors, cytokines and metabolites of arachidonic acid that act on the vasculature, smooth muscle, connective tissue, mucous glands and inflammatory cells (Borish & Joseph 1992, Amin 2012, Metcalfe et al. 1993). FCERI is a multimeric cell-surface receptor that binds the Fc fragment of IgE with high affinity. On mast cells and basophils FCERI exists as a tetrameric complex consisting of one alpha-chain, one beta-chain, and two disulfide-bonded gamma-chains, and on dendritic cells, Langerhans cells, macrophages, and eosinophils it exists as a trimeric complex with one alpha-chain and two disulfide-bonded gamma-chains (Wu 2011, Kraft & Kinet 2007). FCERI signaling in mast cells includes a network of signaling molecules and adaptor proteins. These molecules coordinate ultimately leading to effects on degranulation, eicosanoid production, and cytokine and chemokine production and cell migration and adhesion, growth and survival.
The first step in FCERI signaling is the phosphorylation of the tyrosine residues in the ITAM of both the beta and the gamma subunits of the FCERI by LYN, which is bound to the FCERI beta-chain. The phosphorylated ITAM then recruits the protein tyrosine kinase SYK (spleen tyrosine kinase) which then phosphorylates the adaptor protein LAT. Phosphorylated LAT (linker for activation of T cells) acts as a scaffolding protein and recruits other cytosolic adaptor molecules GRB2 (growth-factor-receptor-bound protein 2), GADS (GRB2-related adaptor protein), SHC (SRC homology 2 (SH2)-domain-containing transforming protein C) and SLP76 (SH2-domain-containing leukocyte protein of 76 kDa), as well as the exchange factors and adaptor molecules VAV and SOS (son of sevenless homologue), and the signalling enzyme phospholipase C gamma1 (PLC-gamma1). Tyrosoine phosphorylation of enzymes and adaptors, including VAV, SHC GRB2 and SOS stimulate small GTPases such as RAC, RAS and RAF. These pathways lead to activation of the ERK, JNK and p38 MAP kinases, histamine release and cytokine production. FCERI activation also triggers the phosphorylation of PLC-gamma which upon membrane localisation hydrolyse PIP2 to form IP3 and 1,2-diacylglycerol (DAG) - second messengers that release Ca2+ from internal stores and activate PKC, respectively. Degranulation or histamine release follows the activation of PLC-gamma and protein kinase C (PKC) and the increased mobilization of calcium (Ca2+). Receptor aggregation also results in the phosphorylation of adaptor protein NTAL/LAT2 which then recruits GAB2. PI3K associates with phosphorylated GAB2 and catalyses the formation of PIP3 in the membrane, which attracts many PH domain proteins like BTK, PLC-gamma, AKT and PDK. PI3K mediated activation of AKT then regulate the mast cell proliferation, development and survival (Gu et al. 2001).
GDNF ProteinP39905 (Uniprot-TrEMBL)
GDP MetaboliteCHEBI:17552 (ChEBI)
GDPMetaboliteCHEBI:17552 (ChEBI)
GFRA1 ProteinP56159 (Uniprot-TrEMBL)
GFRA2 ProteinO00451 (Uniprot-TrEMBL)
GFRA3 ProteinO60609 (Uniprot-TrEMBL)
GFRA4 ProteinQ9GZZ7 (Uniprot-TrEMBL)
GRB2-1 ProteinP62993-1 (Uniprot-TrEMBL)
GRIN1 ProteinQ05586 (Uniprot-TrEMBL)
GRIN2A ProteinQ12879 (Uniprot-TrEMBL)
GRIN2B ProteinQ13224 (Uniprot-TrEMBL)
GRIN2C ProteinQ14957 (Uniprot-TrEMBL)
GRIN2D ProteinO15399 (Uniprot-TrEMBL)
GTP MetaboliteCHEBI:15996 (ChEBI)
GTPMetaboliteCHEBI:15996 (ChEBI)
GalNAc-T178-FGF23(25-251) ProteinQ9GZV9 (Uniprot-TrEMBL)
Gly MetaboliteCHEBI:15428 (ChEBI)
H2OMetaboliteCHEBI:15377 (ChEBI)
HBEGF(63-148) ProteinQ99075 (Uniprot-TrEMBL)
HRAS ProteinP01112 (Uniprot-TrEMBL)
HS MetaboliteCHEBI:28815 (ChEBI)
High affinity binding complex dimers of cytokine receptors using Bc, inactive JAK2, p-(Y593,628)-Bc:p(427,349,350)-SHC1 R-HSA-913439 (Reactome)
IL17RD ProteinQ8NFM7 (Uniprot-TrEMBL)
IL17RD:p-2S MAP2Ks:MAPKsComplexR-HSA-5674360 (Reactome)
IL17RD:p-2S MAP2Ks:p-T,Y MAPKsComplexR-HSA-5674362 (Reactome)
IL17RDProteinQ8NFM7 (Uniprot-TrEMBL)
IL2 ProteinP60568 (Uniprot-TrEMBL)
IL2RA ProteinP01589 (Uniprot-TrEMBL)
IL2RG ProteinP31785 (Uniprot-TrEMBL)
IQGAP1 ProteinP46940 (Uniprot-TrEMBL)
ITGA2B(32-1039) ProteinP08514 (Uniprot-TrEMBL)
ITGB3 ProteinP05106 (Uniprot-TrEMBL)
Interleukin-2 signalingPathwayR-HSA-451927 (Reactome) Interleukin-2 (IL-2) is a cytokine that is produced by T cells in response to antigen stimulation. Originally, IL-2 was discovered because of its potent growth factor activity on activated T cells in vitro and was therefore named 'T cell growth factor' (TCGF). However, the generation of IL-2- and IL-2 receptor-deficient mice revealed that IL-2 also plays a regulatory role in the immune system by suppressing autoimmune responses. Two main mechanisms have been identified that explain this suppressive function: (1) IL-2 sensitizes activated T cells for activation-induced cell death (AICD) and (2) IL-2 is critical for the survival and function of regulatory T cells (Tregs), which possess potent immunosuppressive properties.

IL-2 signaling occurs when IL-2 binds to the heterotrimeric high-affinity IL-2 receptor (IL-2R), which consists of alpha, beta and gamma chains. The IL-2R was identified in 1981 via radiolabeled ligand binding (Robb et al. 1981). The IL-2R alpha chain was identified in 1982 (Leonard et al.), the beta chain in 1986/7 (Sharon et al. 1986, Teshigawara et al. 1987) and the IL-2R gamma chain in 1992 (Takeshita et al.). The high affinity of IL-2 binding to the IL-2R is created by a very rapid association rate to the IL-2R alpha chain, combined with a much slower dissociation rate contributed by the combination of the IL-2R beta and gamma chains (Wang & Smith 1987). After antigen stimulation, T cells upregulate the high-affinity IL-2R alpha chain; IL-2R alpha captures IL-2 and this complex then associates with the constitutively expressed IL-2R beta and gamma chains. The IL-2R gamma chain is shared by several other members of the cytokine receptor superfamily including IL-4, IL-7, IL-9, IL-15 and IL-21 receptors, and consequently is often referred to as the Common gamma chain (Gamma-c). The tyrosine kinases Jak1 and Jak3, which are constitutively associated with IL-2R beta and Gamma-c respectively, are activated resulting in phosphorylation of three critical tyrosine residues in the IL-2R beta cytoplasmic tail. These phosphorylated residues enable recruitment of the adaptor molecule Shc, activating the MAPK and PI3K pathways, and the transcription factor STAT5. After phosphorylation, STAT5 forms dimers that translocate to the nucleus and initiate gene expression. While STAT5 activation is critical for IL-2 function in most cell types, the contribution of the PI3K/Akt pathway differs between distinct T cell subsets. In Tregs for example, PI3K/Akt is not involved in IL-2 signaling and this may explain some of the different functional outcomes of IL-2 signaling in Tregs vs. effector T cells.
Interleukin-3, 5 and GM-CSF signalingPathwayR-HSA-512988 (Reactome) The Interleukin-3 (IL-3), IL-5 and Granulocyte-macrophage colony stimulating factor (GM-CSF) receptors form a family of heterodimeric receptors that have specific alpha chains but share a common beta subunit, often referred to as the common beta (Bc). Both subunits contain extracellular conserved motifs typical of the cytokine receptor superfamily. The cytoplasmic domains have limited similarity with other cytokine receptors and lack detectable catalytic domains such as tyrosine kinase domains.

IL-3 is a 20-26 kDa product of CD4+ T cells that acts on the most immature marrow progenitors. IL-3 is capable of inducing the growth and differentiation of multi-potential hematopoietic stem cells, neutrophils, eosinophils, megakaryocytes, macrophages, lymphoid and erythroid cells. IL-3 has been used to support the proliferation of murine cell lines with properties of multi-potential progenitors, immature myeloid as well as T and pre-B lymphoid cells (Miyajima et al. 1992). IL-5 is a hematopoietic growth factor responsible for the maturation and differentiation of eosinophils. It was originally defined as a T-cell-derived cytokine that triggers activated B cells for terminal differentiation into antibody-secreting plasma cells. It also promotes the generation of cytotoxic T-cells from thymocytes. IL-5 induces the expression of IL-2 receptors (Kouro & Takatsu 2009). GM-CSF is produced by cells (T-lymphocytes, tissue macrophages, endothelial cells, mast cells) found at sites of inflammatory responses. It stimulates the growth and development of progenitors of granulocytes and macrophages, and the production and maturation of dendritic cells. It stimulates myeloblast and monoblast differentiation, synergises with Epo in the proliferation of erythroid and megakaryocytic progenitor cells, acts as an autocrine mediator of growth for some types of acute myeloid leukemia, is a strong chemoattractant for neutrophils and eosinophils. It enhances the activity of neutrophils and macrophages. Under steady-state conditions GM-CSF is not essential for the production of myeloid cells, but it is required for the proper development of alveolar macrophages, otherwise, pulmonary alvelolar proteinosis (PAP) develops. A growing body of evidence suggests that GM-CSF plays a key role in emergency hematopoiesis (predominantly myelopoiesis) in response to infection, including the production of granulocytes and macrophages in the bone marrow and their maintenance, survival, and functional activation at sites of injury or insult (Hercus et al. 2009).

All three receptors have alpha chains that bind their specific ligands with low affinity (de Groot et al. 1998). Bc then associates with the alpha chain forming a high affinity receptor (Geijsen et al. 2001), though the in vivo receptor is likely be a higher order multimer as recently demonstrated for the GM-CSF receptor (Hansen et al. 2008).

The receptor chains lack intrinsic kinase activity, instead they interact with and activate signaling kinases, notably Janus Kinase 2 (JAK2). These phosphorylate the common beta subunit, allowing recruitment of signaling molecules such as Shc, the phosphatidylinositol 3-kinases (PI3Ks), and the Signal Transducers and Activators of Transcription (STATs). The cytoplasmic domain of Bc has two distinct functional domains: the membrane proximal region mediates the induction of proliferation-associated genes such as c-myc, pim-1 and oncostatin M. This region binds multiple signal-transducing proteins including JAK2 (Quelle et al. 1994), STATs, c-Src and PI3 kinase (Rao and Mufson, 1995). The membrane distal domain is required for cytokine-induced growth inhibition and is necessary for the viability of hematopoietic cells (Inhorn et al. 1995). This region interacts with signal-transducing proteins such as Shc (Inhorn et al. 1995) and SHP and mediates the transcriptional activation of c-fos, c-jun, c-Raf and p70S6K (Reddy et al. 2000).



Figure reproduced by permission from Macmillan Publishers Ltd: Leukemia, WL Blalock et al. 13:1109-1166, copyright 1999. Note that residue numbering in this diagram refers to the mature Common beta chain with signal peptide removed.
JAK2 ProteinO60674 (Uniprot-TrEMBL)
JAK3 ProteinP52333 (Uniprot-TrEMBL)
KBTBD7 ProteinQ8WVZ9 (Uniprot-TrEMBL)
KBTBD7:CUL3:RBX1ComplexR-HSA-5658416 (Reactome)
KITLG-1(26-190) ProteinP21583-1 (Uniprot-TrEMBL)
KL-1 ProteinQ9UEF7-1 (Uniprot-TrEMBL)
KL-2 ProteinQ9UEF7-2 (Uniprot-TrEMBL)
KLB ProteinQ86Z14 (Uniprot-TrEMBL)
KRAS ProteinP01116 (Uniprot-TrEMBL)
KSR1 ProteinQ8IVT5 (Uniprot-TrEMBL)
KSR1:MARK3ComplexR-HSA-5672689 (Reactome)
KSR2 ProteinQ6VAB6 (Uniprot-TrEMBL)
L-Glu MetaboliteCHEBI:16015 (ChEBI)
LAMTOR2 ProteinQ9Y2Q5 (Uniprot-TrEMBL)
LAMTOR3 ProteinQ9UHA4 (Uniprot-TrEMBL)
MAP2K homo/heterodimersComplexR-HSA-5672716 (Reactome)
MAP2K1 ProteinQ02750 (Uniprot-TrEMBL)
MAP2K2 ProteinP36507 (Uniprot-TrEMBL)
MAP3K11ProteinQ16584 (Uniprot-TrEMBL)
MAPK monomers and dimersComplexR-HSA-5675361 (Reactome)
MAPK monomers and dimersComplexR-HSA-5675363 (Reactome)
MAPK1 ProteinP28482 (Uniprot-TrEMBL)
MAPK3 ProteinP27361 (Uniprot-TrEMBL)
MAPKs R-HSA-169291 (Reactome)
MAPKsComplexR-HSA-169291 (Reactome)
MARK3 ProteinP27448 (Uniprot-TrEMBL)
Mg2+ MetaboliteCHEBI:18420 (ChEBI)
Mn2+ MetaboliteCHEBI:29035 (ChEBI)
NCAM signaling for neurite out-growthPathwayR-HSA-375165 (Reactome) The neural cell adhesion molecule, NCAM, is a member of the immunoglobulin (Ig) superfamily and is involved in a variety of cellular processes of importance for the formation and maintenance of the nervous system. The role of NCAM in neural differentiation and synaptic plasticity is presumed to depend on the modulation of intracellular signal transduction cascades. NCAM based signaling complexes can initiate downstream intracellular signals by at least two mechanisms: (1) activation of FGFR and (2) formation of intracellular signaling complexes by direct interaction with cytoplasmic interaction partners such as Fyn and FAK. Tyrosine kinases Fyn and FAK interact with NCAM and undergo phosphorylation and this transiently activates the MAPK, ERK 1 and 2, cAMP response element binding protein (CREB) and transcription factors ELK and NFkB. CREB activates transcription of genes which are important for axonal growth, survival, and synaptic plasticity in neurons.

NCAM1 mediated intracellular signal transduction is represented in the figure below. The Ig domains in NCAM1 are represented in orange ovals and Fn domains in green squares. The tyrosine residues susceptible to phosphorylation are represented in red circles and their positions are numbered. Phosphorylation is represented by red arrows and dephosphorylation by yellow. Ig, Immunoglobulin domain; Fn, Fibronectin domain; Fyn, Proto-oncogene tyrosine-protein kinase Fyn; FAK, focal adhesion kinase; RPTPalpha, Receptor-type tyrosine-protein phosphatase; Grb2, Growth factor receptor-bound protein 2; SOS, Son of sevenless homolog; Raf, RAF proto-oncogene serine/threonine-protein kinase; MEK, MAPK and ERK kinase; ERK, Extracellular signal-regulated kinase; MSK1, Mitogen and stress activated protein kinase 1; CREB, Cyclic AMP-responsive element-binding protein; CRE, cAMP response elements.
NCAM1 ProteinP13591 (Uniprot-TrEMBL)
NEFL ProteinP07196 (Uniprot-TrEMBL)
NF1(2-2839) ProteinP21359 (Uniprot-TrEMBL)
NF1(2-2839)ProteinP21359 (Uniprot-TrEMBL)
NGF signalling via

TRKA from the

plasma membrane
PathwayR-HSA-187037 (Reactome) Trk receptors signal from the plasma membrane and from intracellular membranes, particularly from early endosomes. Signalling from the plasma membrane is fast but transient; signalling from endosomes is slower but long lasting. Signalling from the plasma membrane is annotated here. TRK signalling leads to proliferation in some cell types and neuronal differentiation in others. Proliferation is the likely outcome of short term signalling, as observed following stimulation of EGFR (EGF receptor). Long term signalling via TRK receptors, instead, was clearly shown to be required for neuronal differentiation in response to neurotrophins.
NRAS ProteinP01111 (Uniprot-TrEMBL)
NRTN ProteinQ99748 (Uniprot-TrEMBL)
Neuregulins R-HSA-1227957 (Reactome)
Neurotransmitter

Receptor Binding And Downstream Transmission In The

Postsynaptic Cell
PathwayR-HSA-112314 (Reactome) The neurotransmitter in the synaptic cleft released by the pre-synaptic neuron binds specific receptors located on the post-synaptic terminal. These receptors are either ion channels or G protein coupled receptors that function to transmit the signals from the post-synaptic membrane to the cell body.
PAQR3 ProteinQ6TCH7 (Uniprot-TrEMBL)
PAQR3:inactive RAFsComplexR-HSA-5674139 (Reactome)
PAQR3ProteinQ6TCH7 (Uniprot-TrEMBL)
PDGFA-1 ProteinP04085-1 (Uniprot-TrEMBL)
PDGFA-2 ProteinP04085-2 (Uniprot-TrEMBL)
PDGFB ProteinP01127 (Uniprot-TrEMBL)
PDGFB(82-241) ProteinP01127 (Uniprot-TrEMBL)
PEA15 ProteinQ15121 (Uniprot-TrEMBL)
PEA15ProteinQ15121 (Uniprot-TrEMBL)
PEBP1 ProteinP30086 (Uniprot-TrEMBL)
PEBP1ProteinP30086 (Uniprot-TrEMBL)
PHBProteinP35232 (Uniprot-TrEMBL)
PI(4,5)P2 MetaboliteCHEBI:18348 (ChEBI)
PP2AComplexR-HSA-196206 (Reactome)
PPP2CA ProteinP67775 (Uniprot-TrEMBL)
PPP2CB ProteinP62714 (Uniprot-TrEMBL)
PPP2R1A ProteinP30153 (Uniprot-TrEMBL)
PPP2R1B ProteinP30154 (Uniprot-TrEMBL)
PPP2R5A ProteinQ15172 (Uniprot-TrEMBL)
PPP2R5B ProteinQ15173 (Uniprot-TrEMBL)
PPP2R5C ProteinQ13362 (Uniprot-TrEMBL)
PPP2R5D ProteinQ14738 (Uniprot-TrEMBL)
PPP2R5E ProteinQ16537 (Uniprot-TrEMBL)
PPP5CProteinP53041 (Uniprot-TrEMBL)
PSD-95 ProteinP78352 (Uniprot-TrEMBL)
PSMA1 ProteinP25786 (Uniprot-TrEMBL)
PSMA2 ProteinP25787 (Uniprot-TrEMBL)
PSMA3 ProteinP25788 (Uniprot-TrEMBL)
PSMA4 ProteinP25789 (Uniprot-TrEMBL)
PSMA5 ProteinP28066 (Uniprot-TrEMBL)
PSMA6 ProteinP60900 (Uniprot-TrEMBL)
PSMA7 ProteinO14818 (Uniprot-TrEMBL)
PSMA8 ProteinQ8TAA3 (Uniprot-TrEMBL)
PSMB1 ProteinP20618 (Uniprot-TrEMBL)
PSMB10 ProteinP40306 (Uniprot-TrEMBL)
PSMB11 ProteinA5LHX3 (Uniprot-TrEMBL)
PSMB2 ProteinP49721 (Uniprot-TrEMBL)
PSMB3 ProteinP49720 (Uniprot-TrEMBL)
PSMB4 ProteinP28070 (Uniprot-TrEMBL)
PSMB5 ProteinP28074 (Uniprot-TrEMBL)
PSMB6 ProteinP28072 (Uniprot-TrEMBL)
PSMB7 ProteinQ99436 (Uniprot-TrEMBL)
PSMB8 ProteinP28062 (Uniprot-TrEMBL)
PSMB9 ProteinP28065 (Uniprot-TrEMBL)
PSMC1 ProteinP62191 (Uniprot-TrEMBL)
PSMC2 ProteinP35998 (Uniprot-TrEMBL)
PSMC3 ProteinP17980 (Uniprot-TrEMBL)
PSMC4 ProteinP43686 (Uniprot-TrEMBL)
PSMC5 ProteinP62195 (Uniprot-TrEMBL)
PSMC6 ProteinP62333 (Uniprot-TrEMBL)
PSMD1 ProteinQ99460 (Uniprot-TrEMBL)
PSMD10 ProteinO75832 (Uniprot-TrEMBL)
PSMD11 ProteinO00231 (Uniprot-TrEMBL)
PSMD12 ProteinO00232 (Uniprot-TrEMBL)
PSMD13 ProteinQ9UNM6 (Uniprot-TrEMBL)
PSMD14 ProteinO00487 (Uniprot-TrEMBL)
PSMD2 ProteinQ13200 (Uniprot-TrEMBL)
PSMD3 ProteinO43242 (Uniprot-TrEMBL)
PSMD4 ProteinP55036 (Uniprot-TrEMBL)
PSMD5 ProteinQ16401 (Uniprot-TrEMBL)
PSMD6 ProteinQ15008 (Uniprot-TrEMBL)
PSMD7 ProteinP51665 (Uniprot-TrEMBL)
PSMD8 ProteinP48556 (Uniprot-TrEMBL)
PSMD9 ProteinO00233 (Uniprot-TrEMBL)
PSME1 ProteinQ06323 (Uniprot-TrEMBL)
PSME2 ProteinQ9UL46 (Uniprot-TrEMBL)
PSME3 ProteinP61289 (Uniprot-TrEMBL)
PSME4 ProteinQ14997 (Uniprot-TrEMBL)
PSMF1 ProteinQ92530 (Uniprot-TrEMBL)
PSPN ProteinO60542 (Uniprot-TrEMBL)
PTPRA ProteinP18433 (Uniprot-TrEMBL)
Phosphorylated ERBB2:ERBB4cyt1 heterodimers R-HSA-1963568 (Reactome)
Phosphorylated ERBB2:ERBB4cyt2 heterodimers R-HSA-1963591 (Reactome)
Phosphorylated p-6Y-ERBB2 heterodimers R-HSA-1963585 (Reactome)
Phosphorylated p-Y877-ERBB2 heterodimers R-HSA-1963580 (Reactome)
PiMetaboliteCHEBI:18367 (ChEBI)
RAF activating kinasesComplexR-HSA-5672734 (Reactome)
RAF/MAPK scaffoldsComplexR-HSA-5672717 (Reactome)
RAP1A ProteinP62834 (Uniprot-TrEMBL)
RAP1B ProteinP61224 (Uniprot-TrEMBL)
RAPGEF2 ProteinQ9Y4G8 (Uniprot-TrEMBL)
RAS GAPsComplexR-HSA-5658216 (Reactome)
RAS GEFsComplexR-HSA-5672601 (Reactome)
RASA1 ProteinP20936 (Uniprot-TrEMBL)
RASA2 ProteinQ15283 (Uniprot-TrEMBL)
RASA3 ProteinQ14644 (Uniprot-TrEMBL)
RASA4 ProteinO43374 (Uniprot-TrEMBL)
RASAL1 ProteinO95294 (Uniprot-TrEMBL)
RASAL2 ProteinQ9UJF2 (Uniprot-TrEMBL)
RASAL3 ProteinQ86YV0 (Uniprot-TrEMBL)
RASGEF1A ProteinQ8N9B8 (Uniprot-TrEMBL)
RASGRF1 ProteinQ13972 (Uniprot-TrEMBL)
RASGRF2 ProteinO14827 (Uniprot-TrEMBL)
RASGRP1 ProteinO95267 (Uniprot-TrEMBL)
RASGRP4 ProteinQ8TDF6 (Uniprot-TrEMBL)
RBX1 ProteinP62877 (Uniprot-TrEMBL)
RPS27A(1-76) ProteinP62979 (Uniprot-TrEMBL)
SOS1 ProteinQ07889 (Uniprot-TrEMBL)
SPRED dimer:NF1ComplexR-HSA-5658232 (Reactome)
SPRED dimer:ub-NF1ComplexR-HSA-5658420 (Reactome)
SPRED dimerComplexR-HSA-5658219 (Reactome)
SPRED1 ProteinQ7Z699 (Uniprot-TrEMBL)
SPRED2 ProteinQ7Z698 (Uniprot-TrEMBL)
SPRED3 ProteinQ2MJR0 (Uniprot-TrEMBL)
SPTA1 ProteinP02549 (Uniprot-TrEMBL)
SPTAN1 ProteinQ13813 (Uniprot-TrEMBL)
SPTB ProteinP11277 (Uniprot-TrEMBL)
SPTBN1 ProteinQ01082 (Uniprot-TrEMBL)
SPTBN2 ProteinO15020 (Uniprot-TrEMBL)
SPTBN4 ProteinQ9H254 (Uniprot-TrEMBL)
SPTBN5 ProteinQ9NRC6 (Uniprot-TrEMBL)
SYNGAP1 ProteinQ96PV0 (Uniprot-TrEMBL)
Signaling by EGFRPathwayR-HSA-177929 (Reactome) The epidermal growth factor receptor (EGFR) is one member of the ERBB family of transmembrane glycoprotein tyrosine receptor kinases (RTK). Binding of EGFR to its ligands induces conformational change that unmasks the dimerization interface in the extracellular domain of EGFR, leading to receptor homo- or heterodimerization at the cell surface. Dimerization of the extracellular regions of EGFR triggers additional conformational change of the cytoplasmic EGFR regions, enabling the kinase domains of two EGFR molecules to achieve the catalytically active conformation. Ligand activated EGFR dimers trans-autophosphorylate on tyrosine residues in the cytoplasmic tail of the receptor. Phosphorylated tyrosines serve as binding sites for the recruitment of signal transducers and activators of intracellular substrates, which then stimulate intracellular signal transduction cascades that are involved in regulating cellular proliferation, differentiation, and survival. Recruitment of complexes containing GRB2 and SOS1 to phosphorylated EGFR dimers either directly, through phosphotyrosine residues that serve as GRB2 docking sites, or indirectly, through SHC1 recruitment, promotes GDP to GTP exchange on RAS, resulting in the activation of RAF/MAP kinase cascade. Binding of complexes of GRB2 and GAB1 to phosphorylated EGFR dimers leads to formation of the active PI3K complex, conversion of PIP2 into PIP3, and activation of AKT signaling. Phospholipase C-gamma1 (PLCG1) can also be recruited directly, through EGFR phosphotyrosine residues that serve as PLCG1 docking sites, which leads to PLCG1 phosphorylation by EGFR and activation of DAG and IP3 signaling. EGFR signaling is downregulated by the action of ubiquitin ligase CBL. CBL binds directly to the phosphorylated EGFR dimer through the phosphotyrosine Y1045 in the C-tail of EGFR, and after CBL is phosphorylated by EGFR, it becomes active and ubiquitinates phosphorylated EGFR dimers, targeting them for degradation. For a recent review of EGFR signaling, please refer to Avraham and Yarden, 2011.
Signaling by ERBB2PathwayR-HSA-1227986 (Reactome) ERBB2, also known as HER2 or NEU, is a receptor tyrosine kinase (RTK) belonging to the EGFR family. ERBB2 possesses an extracellular domain that does not bind any known ligand, contrary to other EGFR family members, a single transmembrane domain, and an intracellular domain consisting of an active kinase and a C-tail with multiple tyrosine phosphorylation sites. Inactive ERBB2 is associated with a chaperone heat shock protein 90 (HSP90) and its co-chaperone CDC37 (Xu et al. 2001, Citri et al. 2004, Xu et al. 2005). In addition, ERBB2 is associated with ERBB2IP (also known as ERBIN or LAP2), a protein responsible for proper localization of ERBB2. In epithelial cells, ERBB2IP restricts expression of ERBB2 to basolateral plasma membrane regions (Borg et al. 2000).

ERBB2 becomes activated by forming a heterodimer with another ligand-activated EGFR family member, either EGFR, ERBB3 or ERBB4, which is accompanied by dissociation of chaperoning proteins HSP90 and CDC37 (Citri et al. 2004), as well as ERBB2IP (Borg et al. 2000) from ERBB2. ERBB2 heterodimers function to promote cell proliferation, cell survival and differentiation, depending on the cellular context. ERBB2 can also be activated by homodimerization when it is overexpressed, in cancer for example.

In cells expressing both ERBB2 and EGFR, EGF stimulation of EGFR leads to formation of both ERBB2:EGFR heterodimers (Wada et al. 1990, Karunagaran et al. 1996) and EGFR homodimers. Heterodimers of ERBB2 and EGFR trans-autophosphorylate on twelve tyrosine residues, six in the C-tail of EGFR and six in the C-tail of ERBB2 - Y1023, Y1139, Y1196, Y1221, Y1222 and Y1248 (Margolis et al. 1989, Hazan et al. 1990,Walton et al. 1990, Helin et al. 1991, Ricci et al. 1995, Pinkas-Kramarski 1996). Phosphorylated tyrosine residues in the C-tail of EGFR and ERBB2 serve as docking sites for downstream signaling molecules. Three key signaling pathways activated by ERBB2:EGFR heterodimers are RAF/MAP kinase cascade, PI3K-induced AKT signaling, and signaling by phospholipase C gamma (PLCG1). Downregulation of EGFR signaling is mediated by ubiquitin ligase CBL, and is shown under Signaling by EGFR.

In cells expressing ERBB2 and ERBB3, ERBB3 activated by neuregulin NRG1 or NRG2 binding (Tzahar et al. 1994) forms a heterodimer with ERBB2 (Pinkas-Kramarski et al. 1996, Citri et al. 2004). ERBB3 is the only EGFR family member with no kinase activity, and can only function in heterodimers, with ERBB2 being its preferred heterodimerization partner. After heterodimerization, ERBB2 phosphorylates ten tyrosine residues in the C-tail of ERBB3, Y1054, Y1197, Y1199, Y1222, Y1224, Y1260, Y1262, Y1276, Y1289 and Y1328 (Prigent et al. 1994, Pinkas-Kramarski et al. 1996, Vijapurkar et al. 2003, Li et al. 2007) that subsequently serve as docking sites for downstream signaling molecules, resulting in activation of PI3K-induced AKT signaling and RAF/MAP kinase cascade. Signaling by ERBB3 is downregulated by the action of RNF41 ubiquitin ligase, also known as NRDP1.

In cells expressing ERBB2 and ERBB4, ligand stimulated ERBB4 can either homodimerize or form heterodimers with ERBB2 (Li et al. 2007), resulting in trans-autophosphorylation of ERBB2 and ERBB4 on C-tail tyrosine residues that will subsequently serve as docking sites for downstream signaling molecules, leading to activation of RAF/MAP kinase cascade and, in the case of ERBB4 CYT1 isoforms, PI3K-induced AKT signaling (Hazan et al. 1990, Cohen et al. 1996, Li et al. 2007, Kaushansky et al. 2008). Signaling by ERBB4 is downregulated by the action of WWP1 and ITCH ubiquitin ligases, and is shown under Signaling by ERBB4.
Signaling by ERBB4PathwayR-HSA-1236394 (Reactome) ERBB4, also known as HER4, belongs to the ERBB family of receptors, which also includes ERBB1 (EGFR i.e. HER1), ERBB2 (HER2 i.e. NEU) and ERBB3 (HER3). Similar to EGFR, ERBB4 has an extracellular ligand binding domain, a single transmembrane domain and a cytoplasmic domain which contains an active tyrosine kinase and a C-tail with multiple phosphorylation sites. At least three and possibly four splicing isoforms of ERBB4 exist that differ in their C-tail and/or the extracellular juxtamembrane regions: ERBB4 JM-A CYT1, ERBB4 JM-A CYT2 and ERBB4 JM-B CYT1 (the existence of ERBB4 JM-B CYT2 has not been confirmed).

ERBB4 becomes activated by binding one of its seven ligands, three of which, HB-EGF, epiregulin EPR and betacellulin BTC, are EGF-like (Elenius et al. 1997, Riese et al. 1998), while four, NRG1, NRG2, NRG3 and NRG4, belong to the neuregulin family (Tzahar et al. 1994, Carraway et al. 1997, Zhang et al. 1997, Hayes et al. 2007). Upon ligand binding, ERBB4 forms homodimers (Sweeney et al. 2000) or it heterodimerizes with ERBB2 (Li et al. 2007). Dimers of ERBB4 undergo trans-autophosphorylation on tyrosine residues in the C-tail (Cohen et al. 1996, Kaushansky et al. 2008, Hazan et al. 1990, Li et al. 2007), triggering downstream signaling cascades. The pathway Signaling by ERBB4 only shows signaling by ERBB4 homodimers. Signaling by heterodimers of ERBB4 and ERBB2 is shown in the pathway Signaling by ERBB2. Ligand-stimulated ERBB4 is also able to form heterodimers with ligand-stimulated EGFR (Cohen et al. 1996) and ligand-stimulated ERBB3 (Riese et al. 1995). Dimers of ERBB4 with EGFR and dimers of ERBB4 with ERBB3 were demonstrated in mouse cell lines in which human ERBB4 and EGFR or ERBB3 were exogenously expressed. These heterodimers undergo trans-autophosphorylation, but their downstream signaling and physiological significance have not been studied.


All splicing isoforms of ERBB4 possess two tyrosine residues in the C-tail that serve as docking sites for SHC1 (Kaushansky et al. 2008, Pinkas-Kramarski et al. 1996, Cohen et al. 1996). Once bound to ERBB4, SHC1 becomes phosphorylated on tyrosine residues by the tyrosine kinase activity of ERBB4, which enables it to recruit the complex of GRB2 and SOS1, resulting in the guanyl-nucleotide exchange on RAS and activation of RAF and MAP kinase cascade (Kainulainen et al. 2000).

The CYT1 isoforms of ERBB4 also possess a C-tail tyrosine residue that, upon trans-autophosphorylation, serves as a docking site for the p85 alpha subunit of PI3K (Kaushansky et al. 2008, Cohen et al. 1996), leading to assembly of an active PI3K complex that converts PIP2 to PIP3 and activates AKT signaling (Kainulainen et al. 2000).

Besides signaling as a transmembrane receptor, ligand activated homodimers of ERBB4 JM-A isoforms (ERBB4 JM-A CYT1 and ERBB4 JM-A CYT2) undergo proteolytic cleavage by ADAM17 (TACE) in the juxtamembrane region, resulting in shedding of the extracellular domain and formation of an 80 kDa membrane bound ERBB4 fragment known as ERBB4 m80 (Rio et al. 2000, Cheng et al. 2003). ERBB4 m80 undergoes further proteolytic cleavage, mediated by the gamma-secretase complex, which releases the soluble 80 kDa ERBB4 intracellular domain, known as ERBB4 s80 or E4ICD, into the cytosol (Ni et al. 2001). ERBB4 s80 is able to translocate to the nucleus, promote nuclear translocation of various transcription factors, and act as a transcription co-factor. In neuronal precursors, ERBB4 s80 binds the complex of TAB and NCOR1, helps to move the complex into the nucleus, and is a co-factor of TAB:NCOR1-mediated inhibition of expression of astrocyte differentiation genes GFAP and S100B (Sardi et al. 2006). In mammary cells, ERBB4 s80 recruits STAT5A transcription factor in the cytosol, shuttles it to the nucleus, and acts as the STAT5A co-factor in binding to and promoting transcription from the beta-casein (CSN2) promoter, and may be involved in the regulation of other lactation-related genes (Williams et al. 2004, Muraoka-Cook et al. 2008). ERBB4 s80 was also shown to bind activated estrogen receptor in the nucleus and act as its transcriptional co-factor in promoting transcription of some estrogen-regulated genes, such as progesterone receptor gene NR3C3 and CXCL12 i.e. SDF1 (Zhu et al. 2006).

The C-tail of ERBB4 possesses several WW-domain binding motifs (three in CYT1 isoform and two in CYT2 isoform), which enable interaction of ERBB4 with WW-domain containing proteins. ERBB4 s80, through WW-domain binding motifs, interacts with YAP1 transcription factor, a known proto-oncogene, and may be a co-regulator of YAP1-mediated transcription (Komuro et al. 2003, Omerovic et al. 2004). The tumor suppressor WWOX, another WW-domain containing protein, competes with YAP1 in binding to ERBB4 s80 and prevents translocation of ERBB4 s80 to the nucleus (Aqeilan et al. 2005). ERBB4 s80 is also able to translocate to the mitochondrial matrix, presumably when its nuclear translocation is inhibited. Once in the mitochondrion, the BH3 domain of ERBB4, characteristic of BCL2 family members, may enable it to act as a pro-apoptotic factor (Naresh et al. 2006). Activation of ERBB4 in breast cancer cell lines leads to JNK-dependent increase in BRCA1 mRNA level and mitotic cell cycle delay, but the exact mechanism has not been elucidated (Muraoka-Cook et al. 2006).

WW-domain binding motifs in the C-tail of ERBB4 play an important role in the downregulation of ERBB4 receptor signaling, enabling the interaction of intact ERBB4, ERBB4 m80 and ERBB4 s80 with NEDD4 family of E3 ubiquitin ligases WWP1 and ITCH. The interaction of WWP1 and ITCH with intact ERBB4 is independent of receptor activation and autophosphorylation. Binding of WWP1 and ITCH ubiquitin ligases leads to ubiquitination of ERBB4 and its cleavage products, and subsequent degradation through both proteasomal and lysosomal routes (Omerovic et al. 2007, Feng et al. 2009). In addition, the s80 cleavage product of ERBB4 JM-A CYT-1 isoform is the target of NEDD4 ubiquitin ligase. NEDD4 binds ERBB4 JM-A CYT-1 s80 (ERBB4jmAcyt1s80) through its PIK3R1 interaction site and mediates ERBB4jmAcyt1s80 ubiquitination, thereby decreasing the amount of ERBB4jmAcyt1s80 that reaches the nucleus (Zeng et al. 2009).
Signaling by FGFR1PathwayR-HSA-5654736 (Reactome) 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.
Signaling by FGFR2PathwayR-HSA-5654738 (Reactome) 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.
Signaling by FGFR3PathwayR-HSA-5654741 (Reactome) 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.
Signaling by FGFR4PathwayR-HSA-5654743 (Reactome) 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.
Signaling by Insulin receptorPathwayR-HSA-74752 (Reactome) Insulin binding to its receptor results in receptor autophosphorylation on tyrosine residues and the tyrosine phosphorylation of insulin receptor substrates (e.g. IRS and Shc) by the insulin receptor tyrosine kinase. This allows association of IRSs with downstream effectors such as PI-3K via its Src homology 2 (SH2) domains leading to end point events such as Glut4 (Slc2a4) translocation. Shc when tyrosine phosphorylated associates with Grb2 and can thus activate the Ras/MAPK pathway independent of the IRSs.

Signal transduction by the insulin receptor is not limited to its activation at the cell surface. The activated ligand-receptor complex initially at the cell surface, is internalised into endosomes itself a process which is dependent on tyrosine autophosphorylation. Endocytosis of activated receptors has the dual effect of concentrating receptors within endosomes and allows the insulin receptor tyrosine kinase to phosphorylate substrates that are spatially distinct from those accessible at the plasma membrane. Acidification of the endosomal lumen, due to the presence of proton pumps, results in dissociation of insulin from its receptor. (The endosome constitutes the major site of insulin degradation). This loss of the ligand-receptor complex attenuates any further insulin-driven receptor re-phosphorylation events and leads to receptor dephosphorylation by extra-lumenal endosomally-associated protein tyrosine phosphatases (PTPs). The identity of these PTPs is not clearly established yet. A discussion of candidates will be added in the near future.

Signaling by PDGFPathwayR-HSA-186797 (Reactome) Platelet-derived Growth Factor (PDGF) is a potent stimulator of growth and motility of connective tissue cells such as fibroblasts and smooth muscle cells as well as other cells such as capillary endothelial cells and neurons.The PDGF family of growth factors is composed of four different polypeptide chains encoded by four different genes. The classical PDGF chains, PDGF-A and PDGF-B, and more recently discovered PDGF-C and PDGF-D. The four PDGF chains assemble into disulphide-bonded dimers via homo- or heterodimerization, and five different dimeric isoforms have been described so far; PDGF-AA, PDGF-AB, PDGF-BB, PDGF-CC and PDGF-DD. It is notable that no heterodimers involving PDGF-C and PDGF-D chains have been described. PDGF exerts its effects by binding to, and activating, two protein tyrosine kinase (PTK) receptors, alpha and beta. These receptors dimerize and undergo autophosphorylation. The phosphorylation sites then attract downstream effectors to transduct the signal into the cell.
Signaling by SCF-KITPathwayR-HSA-1433557 (Reactome) Stem cell factor (SCF) is a growth factor with membrane bound and soluble forms. It is expressed by fibroblasts and endothelial cells throughout the body, promoting proliferation, migration, survival and differentiation of hematopoetic progenitors, melanocytes and germ cells.(Linnekin 1999, Ronnstrand 2004, Lennartsson and Ronnstrand 2006). The receptor for SCF is KIT, a tyrosine kinase receptor (RTK) closely related to the receptors for platelet derived growth factor receptor, colony stimulating factor 1 (Linnekin 1999) and Flt3 (Rosnet et al. 1991). Four isoforms of c-Kit have been identified in humans. Alternative splicing results in isoforms of KIT differing in the presence or absence of four residues (GNNK) in the extracellular region. This occurs due to the use of an alternate 5' splice donor site. These GNNK+ and GNNK- variants are co-expressed in most tissues; the GNNK- form predominates and was more strongly tyrosine-phosphorylated and more rapidly internalized (Ronnstrand 2004). There are also splice variants that arise from alternative usage of splice acceptor site resulting in the presence or absence of a serine residue (Crosier et al., 1993). Finally, there is an alternative shorter transcript of KIT expressed in postmeiotic germ cells in the testis which encodes a truncated KIT consisting only of the second part of the kinase domain and thus lackig the extracellular and transmembrane domains as well as the first part of the kinase domain (Rossi et al. 1991). Binding of SCF homodimers to KIT results in KIT homodimerization followed by activation of its intrinsic tyrosine kinase activity. KIT stimulation activates a wide array of signalling pathways including MAPK, PI3K and JAK/STAT (Reber et al. 2006, Ronnstrand 2004). Defects of KIT in humans are associated with different genetic diseases and also in several types of cancers like mast cell leukaemia, germ cell tumours, certain subtypes of malignant melanoma and gastrointestinal tumours.
Signaling by Type 1

Insulin-like Growth Factor 1 Receptor

(IGF1R)
PathwayR-HSA-2404192 (Reactome) Binding of IGF1 (IGF-I) or IGF2 (IGF-II) to the extracellular alpha peptides of the type 1 insulin-like growth factor receptor (IGF1R) triggers the activation of two major signaling pathways: the SOS-RAS-RAF-MAPK (ERK) pathway and the PI3K-PKB (AKT) pathway (recently reviewed in Pavelic et al. 2007, Chitnis et al. 2008, Maki et al. 2010, Parella et al. 2010, Annunziata et al. 2011, Siddle et al. 2012, Holzenberger 2012).
Signaling by the B Cell Receptor (BCR)PathwayR-HSA-983705 (Reactome) Mature B cells express IgM and IgD immunoglobulins which are complexed at the plasma membrane with Ig-alpha (CD79A, MB-1) and Ig-beta (CD79B, B29) to form the B cell receptor (BCR) (Fu et al. 1974, Fu et al. 1975, Kunkel et al. 1975, Van Noesel et al. 1992, Sanchez et al. 1993, reviewed in Brezski and Monroe 2008). Binding of antigen to the immunoglobulin activates phosphorylation of immunoreceptor tyrosine-based activation motifs (ITAMs) in the cytoplasmic tails of Ig-alpha and Ig-beta by Src family tyrosine kinases, including LYN, FYN, and BLK (Nel et al. 1984, Yamanashi et al. 1991, Flaswinkel and Reth 1994, Saouaf et al. 1994, Hata et al. 1994, Saouaf et al. 1995, reviewed in Gauld and Cambier 2004, reviewed in Harwood and Batista 2010).
The protein kinase SYK binds the phosphorylated immunoreceptor tyrosine-activated motifs (ITAMs) on the cytoplasmic tails of Ig-alpha (CD79A, MB-1) and Ig-beta (CD79B, B29) (Wienands et al. 1995, Rowley et al. 1995, Tsang et al. 2008). The binding causes the activation and autophosphorylation of SYK (Law et al. 1994, Baldock et al. 2000, Irish et al. 2006, Tsang et al. 2008, reviewed in Bradshaw 2010).
Activated SYK and other kinases phosphorylate BLNK (SLP-65), BCAP, and CD19 which serve as scaffolds for the assembly of large complexes, the signalosomes, by recruiting phosphoinositol 3-kinase (PI3K), phospholipase C gamma (predominantly PLC-gamma2 in B cells, Coggeshall et al. 1992), NCK, BAM32, BTK, VAV1, and SHC. The effectors are phosphorylated by SYK and other kinases.
PLC-gamma associated with BLNK hydrolyzes phosphatidylinositol-4,5-bisphosphate to yield inositol-1,4,5-trisphosphate (IP3) and diacylglycerol (Carter et al. 1991, Kim et al. 2004). IP3 binds receptors on the endoplasmic reticulum and causes release of calcium ions from the ER into the cytosol. The depletion of calcium from the ER in turn activates STIM1 to interact with ORAI and TRPC1 channels in the plasma membrane, resulting in an influx of extracellular calcium ions (Muik et al. 2008, Luik et al. 2008, Park et al. 2009, Mori et al. 2002). PI3K associated with BCAP and CD19 phosphorylates phosphatidylinositol 4,5-bisphosphate to yield phosphatidyinositol 3,4,5-trisphosphate.
Second messengers (calcium, diacylglycerol, inositol 1,4,5-trisphosphate, and phosphatidylinositol 3,4,5-trisphosphate) trigger signaling pathways: NF-kappaB is activated via protein kinase C beta, RAS is activated via RasGRP proteins, NF-AT is activated via calcineurin, and AKT (PKB) is activated via PDK1 (reviewed in Shinohara and Kurosaki 2009, Stone 2006).
TLN1 ProteinQ9Y490 (Uniprot-TrEMBL)
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)
UbComplexR-HSA-113595 (Reactome)
VCL ProteinP18206 (Uniprot-TrEMBL)
VWF(23-763) ProteinP04275 (Uniprot-TrEMBL)
WDR83 ProteinQ9BRX9 (Uniprot-TrEMBL)
WDR83:LAMTOR2:LAMTOR3:activated RAF:MAP2K:MAPK complexComplexR-HSA-5674138 (Reactome)
WDR83:LAMTOR2:LAMTOR3:activated RAF:p-2S MAP2K:p-T,Y MAPK complexComplexR-HSA-5674131 (Reactome)
WDR83:LAMTOR2:LAMTOR3ComplexR-HSA-5674133 (Reactome)
YWHAB ProteinP31946 (Uniprot-TrEMBL)
YWHAB dimerComplexR-HSA-2028645 (Reactome)
activated RAF:scaffold:MAP2K:MAPK complexComplexR-HSA-5672720 (Reactome)
activated

RAF:scaffold:p-2S

MAP2K:MAPK complex
ComplexR-HSA-5672723 (Reactome)
activated

RAF:scaffold:p-2S MAP2K:p-2T MAPK

complex
ComplexR-HSA-5672724 (Reactome)
activated RAF homo/heterodimerComplexR-HSA-5674136 (Reactome)
cytosolic MAPK DUSPsComplexR-HSA-5675365 (Reactome)
dephosphorylated "receiver" RAF/KSR1ComplexR-HSA-5672731 (Reactome)
dephosphorylated

inactive RAFS:YWHAB

dimer
ComplexR-HSA-5672728 (Reactome)
dephosphorylated inactive RAFsComplexR-HSA-5672726 (Reactome)
hyperphosphorylated BRAFProteinP15056 (Uniprot-TrEMBL)
hyperphosphorylated RAF1ProteinP04049 (Uniprot-TrEMBL)
inactive RAFs:YWHAB dimerComplexR-HSA-5672707 (Reactome)
inactive RAFsComplexR-HSA-5672708 (Reactome)
nuclear MAPK DUSPsComplexR-HSA-5675368 (Reactome)
p-11Y-PDGFRA ProteinP16234 (Uniprot-TrEMBL)
p-12Y-PDGFRB ProteinP09619 (Uniprot-TrEMBL)
p-2S MAP2K homo/heterodimersComplexR-HSA-5672721 (Reactome)
p-2S MAP2K homo/heterodimers R-HSA-5672721 (Reactome)
p-2S MAP2K1:p-2S MAPK2K2 heterodimerComplexR-HSA-5672705 (Reactome)
p-2S,T MAP2K1:p-2S MAPK2K2 heterodimerComplexR-HSA-5674489 (Reactome)
p-5Y,S1119-TEK ProteinQ02763 (Uniprot-TrEMBL)
p-5Y-FGFR4 ProteinP22455 (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-5Y-LAT-2 ProteinO43561-2 (Uniprot-TrEMBL)
p-5Y-RET ProteinP07949 (Uniprot-TrEMBL)
p-6Y-EGFR ProteinP00533 (Uniprot-TrEMBL)
p-6Y-ERBB2 ProteinP04626 (Uniprot-TrEMBL)
p-6Y-FGFR3b ProteinP22607-2 (Uniprot-TrEMBL)
p-6Y-FGFR3c ProteinP22607-1 (Uniprot-TrEMBL)
p-6Y-FRS2 ProteinQ8WU20 (Uniprot-TrEMBL)
p-6Y-PTK2 ProteinQ05397 (Uniprot-TrEMBL)
p-7Y-ERBB2 ProteinP04626 (Uniprot-TrEMBL)
p-7Y-KIT ProteinP10721 (Uniprot-TrEMBL)
p-8Y-FGFR1b ProteinP11362-19 (Uniprot-TrEMBL) While the existence of a "b" isoform of fibroblast growth factor receptor 1 is well established and its biochemical and functional properties have been extensively characterized (e.g., Mohammadi et al. 2005; Zhang et al. 2006), its amino acid sequence is not represented in reference protein sequence databases, except as the 47-residue polypeptide (deposited in GenBank as accession AAB19502) first used by Johnson et al. (1991) to distinguish the "b" and "c" isoforms of the receptor.
p-8Y-FGFR1c ProteinP11362-1 (Uniprot-TrEMBL)
p-ERBB4cyt1 homodimers R-HSA-1250351 (Reactome)
p-S,218, S222,T292 MAP2K1 ProteinQ02750 (Uniprot-TrEMBL)
p-S214,S576 ARAF ProteinP10398 (Uniprot-TrEMBL)
p-S218,S222-MAP2K1 ProteinQ02750 (Uniprot-TrEMBL)
p-S222,S226-MAP2K2 ProteinP36507 (Uniprot-TrEMBL)
p-S259,S621-RAF1 ProteinP04049 (Uniprot-TrEMBL)
p-S299,S302,T452,T455,S576 ARAF ProteinP10398 (Uniprot-TrEMBL)
p-S311

KSR1:MARK3:YWHAB

dimer
ComplexR-HSA-5672692 (Reactome)
p-S311 KSR1 ProteinQ8IVT5 (Uniprot-TrEMBL)
p-S311 KSR1:MARK3ComplexR-HSA-5672691 (Reactome)
p-S311,S406

KSR1:MARK3:YWHAB

dimer
ComplexR-HSA-5672696 (Reactome)
p-S311,S406 KSR1:MARK3ComplexR-HSA-5672695 (Reactome)
p-S311,S406 KSR1 ProteinQ8IVT5 (Uniprot-TrEMBL)
p-S311,Y552 KSR1 ProteinQ8IVT5 (Uniprot-TrEMBL)
p-S338,Y341,T491,S494,S621 RAF1 ProteinP04049 (Uniprot-TrEMBL)
p-S338,Y341,T491,S494,S621 RAF1ProteinP04049 (Uniprot-TrEMBL)
p-S365,S445,S729 BRAF ProteinP15056 (Uniprot-TrEMBL)
p-S445,S729-BRAF ProteinP15056 (Uniprot-TrEMBL)
p-S445,T599,S602,S729 BRAF ProteinP15056 (Uniprot-TrEMBL)
p-S445,T599,S602,S729 BRAFProteinP15056 (Uniprot-TrEMBL)
p-S576 ARAF ProteinP10398 (Uniprot-TrEMBL)
p-S621-RAF1 ProteinP04049 (Uniprot-TrEMBL)
p-T,Y MAPK dimers R-HSA-1268261 (Reactome)
p-T,Y MAPK dimers R-HSA-198701 (Reactome)
p-T,Y MAPK dimersComplexR-HSA-1268261 (Reactome)
p-T,Y MAPK monomers and dimers:PEA15ComplexR-HSA-5675205 (Reactome)
p-T,Y MAPK monomers and dimersComplexR-HSA-5674340 (Reactome)
p-T,Y MAPK monomers and dimersComplexR-HSA-5674341 (Reactome)
p-T,Y MAPKs R-HSA-169289 (Reactome)
p-T,Y MAPKs R-HSA-5674338 (Reactome)
p-T,Y MAPKsComplexR-HSA-169289 (Reactome)
p-T133-RASGRP3 ProteinQ8IV61 (Uniprot-TrEMBL)
p-T184-RASGRP1 ProteinO95267 (Uniprot-TrEMBL)
p-T185,Y187-MAPK1 ProteinP28482 (Uniprot-TrEMBL)
p-T202,Y204-MAPK3 ProteinP27361 (Uniprot-TrEMBL)
p-T286-CAMK2A ProteinQ9UQM7 (Uniprot-TrEMBL)
p-T287-CAMK2B ProteinQ13554 (Uniprot-TrEMBL)
p-T287-CAMK2D ProteinQ13557 (Uniprot-TrEMBL)
p-T287-CAMK2G ProteinQ13555 (Uniprot-TrEMBL)
p-Y-IRS1 ProteinP35568 (Uniprot-TrEMBL)
p-Y-IRS2 ProteinQ9Y4H2 (Uniprot-TrEMBL)
p-Y-JAK1 ProteinP23458 (Uniprot-TrEMBL)
p-Y-SHC1 ProteinP29353 (Uniprot-TrEMBL)
p-Y-SHC2 ProteinP98077 (Uniprot-TrEMBL)
p-Y-SHC3 ProteinQ92529 (Uniprot-TrEMBL)
p-Y1172,Y1226-ERBB4 JM-A CYT-2 isoform ProteinQ15303-3 (Uniprot-TrEMBL)
p-Y194,Y195,Y272-SHC1-1(156-583) ProteinP29353-3 (Uniprot-TrEMBL)
p-Y239,Y240,Y317-SHC1-2 ProteinP29353-2 (Uniprot-TrEMBL)
p-Y341,T491,S494,S621 RAF1 ProteinP04049 (Uniprot-TrEMBL)
p-Y349,Y350,Y427-SHC1 ProteinP29353 (Uniprot-TrEMBL)
p-Y349,Y350-SHC1 ProteinP29353 (Uniprot-TrEMBL)
p-Y364,Y418,Y536-IL2RB ProteinP14784 (Uniprot-TrEMBL)
p-Y419-SRC ProteinP12931-1 (Uniprot-TrEMBL)
p-Y420-FYN ProteinP06241 (Uniprot-TrEMBL)
p-Y427-SHC1 ProteinP29353 (Uniprot-TrEMBL)
p-Y530-SRC ProteinP12931-1 (Uniprot-TrEMBL)
p21

RAS:GTP:'activator'

RAF:YWHAB dimer
ComplexR-HSA-5672729 (Reactome)
p21

RAS:GTP:activated RAF homo/heterodimer

complexes
ComplexR-HSA-5672718 (Reactome)
p21

RAS:GTP:activated RAF1

homo/heterodimer:PEBP1
ComplexR-HSA-5675413 (Reactome)
p21 RAS:GTP:homo/heterodimerized RAF complexComplexR-HSA-5672733 (Reactome)
p21 RAS:GDPComplexR-HSA-109796 (Reactome)
p21 RAS:GTP:BRAPComplexR-HSA-5674003 (Reactome)
p21 RAS:GTP:RAS GAPsComplexR-HSA-5658227 (Reactome)
p21 RAS:GTP:ub-BRAPComplexR-HSA-5674005 (Reactome)
p21 RAS:GTPComplexR-HSA-109783 (Reactome)
p21RAS:GTP:activated

RAF

homo/heterodimers
ComplexR-HSA-5672712 (Reactome)
p21RAS:GTP:activated

RAF1

homo/heterodimer
ComplexR-HSA-5675414 (Reactome)
p21RAS:GTP:dephosphorylated RAF1 homo/heterodimerComplexR-HSA-5675430 (Reactome)
p21RAS:GTP:p-S621

RAF1

homo/heterodimer
ComplexR-HSA-5675412 (Reactome)
ub-BRAP ProteinQ7Z569 (Uniprot-TrEMBL)
ub-NF1(2-2839) ProteinP21359 (Uniprot-TrEMBL)

Annotated Interactions

View all...
SourceTargetTypeDatabase referenceComment
'activator' RAF:YWHAB dimerR-HSA-5672950 (Reactome)
26S proteasomemim-catalysisR-HSA-5658430 (Reactome)
ADPArrowR-HSA-5672948 (Reactome)
ADPArrowR-HSA-5672969 (Reactome)
ADPArrowR-HSA-5672973 (Reactome)
ADPArrowR-HSA-5672978 (Reactome)
ADPArrowR-HSA-5674130 (Reactome)
ADPArrowR-HSA-5674373 (Reactome)
ADPArrowR-HSA-5674496 (Reactome)
ADPArrowR-HSA-5675194 (Reactome)
ADPArrowR-HSA-5675198 (Reactome)
ATPR-HSA-5672948 (Reactome)
ATPR-HSA-5672969 (Reactome)
ATPR-HSA-5672973 (Reactome)
ATPR-HSA-5672978 (Reactome)
ATPR-HSA-5674130 (Reactome)
ATPR-HSA-5674373 (Reactome)
ATPR-HSA-5674496 (Reactome)
ATPR-HSA-5675194 (Reactome)
ATPR-HSA-5675198 (Reactome)
BRAP:KSR1:MARK3ArrowR-HSA-5674019 (Reactome)
BRAP:KSR1:MARK3TBarR-HSA-5672966 (Reactome)
BRAPR-HSA-5674018 (Reactome)
BRAPR-HSA-5674019 (Reactome)
GDPArrowR-HSA-5672965 (Reactome)
GTPR-HSA-5672965 (Reactome)
H2OR-HSA-5672957 (Reactome)
H2OR-HSA-5672961 (Reactome)
H2OR-HSA-5675373 (Reactome)
H2OR-HSA-5675376 (Reactome)
H2OR-HSA-5675431 (Reactome)
H2OR-HSA-5675433 (Reactome)
IL17RD:p-2S MAP2Ks:MAPKsArrowR-HSA-5674366 (Reactome)
IL17RD:p-2S MAP2Ks:MAPKsR-HSA-5674373 (Reactome)
IL17RD:p-2S MAP2Ks:MAPKsmim-catalysisR-HSA-5674373 (Reactome)
IL17RD:p-2S MAP2Ks:p-T,Y MAPKsArrowR-HSA-5674373 (Reactome)
IL17RDR-HSA-5674366 (Reactome)
KBTBD7:CUL3:RBX1mim-catalysisR-HSA-5658424 (Reactome)
KSR1:MARK3R-HSA-5672948 (Reactome)
KSR1:MARK3R-HSA-5674019 (Reactome)
KSR1:MARK3mim-catalysisR-HSA-5672948 (Reactome)
MAP2K homo/heterodimersR-HSA-5672972 (Reactome)
MAP2K homo/heterodimersR-HSA-5674132 (Reactome)
MAP3K11ArrowR-HSA-5672966 (Reactome)
MAPK monomers and dimersArrowR-HSA-5675373 (Reactome)
MAPK monomers and dimersArrowR-HSA-5675376 (Reactome)
MAPKsR-HSA-5672972 (Reactome)
MAPKsR-HSA-5674132 (Reactome)
MAPKsR-HSA-5674366 (Reactome)
NF1(2-2839)R-HSA-5658438 (Reactome)
PAQR3:inactive RAFsArrowR-HSA-5674140 (Reactome)
PAQR3R-HSA-5674140 (Reactome)
PEA15R-HSA-5675206 (Reactome)
PEBP1R-HSA-5675417 (Reactome)
PHBArrowR-HSA-5672950 (Reactome)
PP2Amim-catalysisR-HSA-5672957 (Reactome)
PP2Amim-catalysisR-HSA-5672961 (Reactome)
PP2Amim-catalysisR-HSA-5675431 (Reactome)
PPP5Cmim-catalysisR-HSA-5675433 (Reactome)
PiArrowR-HSA-5672957 (Reactome)
PiArrowR-HSA-5672961 (Reactome)
PiArrowR-HSA-5675373 (Reactome)
PiArrowR-HSA-5675376 (Reactome)
PiArrowR-HSA-5675431 (Reactome)
PiArrowR-HSA-5675433 (Reactome)
R-HSA-5658231 (Reactome) The intrinsic GTPase activity of RAS proteins is stimulated by the GAP proteins, of which there are at least 10 in the human genome (reviewed in King et al, 2013).
R-HSA-5658424 (Reactome) NF1 levels are controlled by proteasomal degradation in response to stimulation by some growth factors (Cichowski et al, 2003). Ubiquitination is mediated by the CUL3:RBX1 RING E3 ligase complex in conjunction with the BTB adaptor protein KBTBD7 (Hollstein et al, 2013). After its initial rapid degradation, NF1 protein levels are re-established shortly after growth factor treatment, allowing appropriate termination of RAS MAPK signaling (Cichowski et al, 2003). Aberrant destabilization of NF1 by CUL3:KBTBD7-mediated proteasomal degradation has been identified in cases of glioblastoma and depends on activation of PKC alpha (Cichowski et al, 2003; McGillicuddy et al, 2009; Hollstein et al, 2013).
R-HSA-5658430 (Reactome) After ubiquitination by the CUL3:KBTBD7 E3 RING ligase complex, NF1 is degraded by the proteasome (Cichowski et al, 2003; McGillicuddy et al, 2009; Hollstein et al, 2013).
R-HSA-5658435 (Reactome) The human genome encodes at least 10 proteins that bind RAS and activate its intrinsic GTPase activity, resulting in the formation of inactive RAS:GDP and attenuating RAS signaling (reviewed in King et al, 2013). These identified RAS GAP proteins are RASA1 (also known as p120 GAP), NF1, the GAP1 family (RASA2, RASA3, RASA4 and RASAL1) and the SYNGAP family (SYNGAP1, DAB2IP, RASAL2 and RASAL3). GAP proteins stimulate RAS GTPase activity by inserting a conserved arginine residue into the RAS active site, promoting a conformational change in the active site to allow GTP hydrolysis (Ahamdian et al, 2003; Scheffzek et al, 1997; Ahamdian et al, 1997). In addition to the GAP domain, most RAS GAP proteins also contain membrane targeting domains that facilitate interaction with the plasma membrane where RAS is tethered. In some cases, such as RASA3, membrane localization is constitutive, whereas in others, the GAP proteins are targeted to the membrane in response to cellular signaling. In addition to binding RAS, a number of GAP proteins also mediate other protein-protein interactions and act as scaffolds to integrate signaling; some GAPs are also known to bind and activate other small GTPases such as RAP (reviewed in King et al, 2013). Loss-of-functions mutations in RAS GAP proteins have been identified in a number of cancers (reviewed in Maertens and Cichowski, 2014).
R-HSA-5658438 (Reactome) Sprouty-related proteins (SPRED) 1, 2 and 3 are negative regulators of the MAPK pathway that act at least in part by recruiting the RAS GAP protein neurofibromin 1 (NF1) to the plasma membrane (Kato et al, 2003; King et al, 2006; Stowe et al, 2012). NF1, a negative regulator of RAS is a tumor suppressor that is mutated in the familial cancer syndrome neurofibromatosis I as well as in sporadic cases of glioblastoma, non-small cell lung cancers, neuroblastoma and melanoma (Martin et al, 1990; Bollag et al, 1996; reviewed in Bollag and McCormick, 1992; Maertens and Cichowski, 2014).

Plasma membrane-association of the SPRED proteins themselves depends on the C-terminal SPR domain. Mutations in this region abrogate membrane localization of the protein (King et al, 2005; Stowe et al, 2012). Membrane association may also be promoted by interaction of the SPRED proteins with RAS (Wakioka et al, 2001). Interaction with NF1 is mediated by the SPRED EVH1 domain, and mutations in this region affect both NF1 recruitment and the ability of SPRED and NF1 proteins to negatively regulate RAS pathway activity (Stowe et al, 2012; reviewed in McClatchey and Cichowski, 2012).
R-HSA-5672948 (Reactome) KSR1 (Kinase suppressor of RAS 1) was originally identified in Drosophila and C. elegans as a suppressor of activated RAS, and is one of a number of scaffolding proteins that bring RAS-RAF-MAPK members together to promote pathway activation (Therrien et al, 1995; Kornfeld et al, 1995; Sundaram et al, 1995; reviewed in Zhang et al, 2013). Consistent with its role as a scaffolding protein, KSR1 interacts with all of the kinases of the MAPK pathway. Interaction with the MEK proteins MAP2K1 and MAP2K2 is constitutive, while pathway activation promotes heterodimerization with activated RAF proteins and subsequent interaction with ERK/MAPK proteins (Rajakulendran et al, 2009; Therrien et al, 1996; McKay et al, 2009; Hu et al, 2011; Hu et al, 2013; Brennan et al, 2011; note, however, that for simplicity MAP2K/MEK proteins are not depicted as part of this reaction).
Like the RAF proteins, KSR1 is maintained in an inactive state in quiescent cells by interaction with 14-3-3 dimers; this interaction is promoted by phosphorylation of KSR1 residues S311 and S406 by the MAP/microtubule affinity-regulating kinase 3 (MARK3), which is constitutively bound to KSR1 (Muller et al, 2001).



R-HSA-5672950 (Reactome) Activation of RAS downstream of extracellular signals allows RAS:GTP to recruit BRAF to the plasma membrane, disrupting the inactivating interaction between BRAF and the 14-3-3 protein YWHAB (Marais et al, 1997; Yamamori et al, 1995; reviewed in Cseh et al, 2014). BRAF, unique of the three mammalian RAF isoforms, is constitutively phosphorylated on the conserved serine residue (445 in BRAF) in the N-terminal acidic motif (NtA). Constitutive negative charge in this region is critical for BRAF to function as an activator of other RAF molecules, allowing signal amplification (Marais et al, 1997; Mason et al, 1999; Wan et al, 2004; Garnett et al, 2005; Hu et al, 2013; reviewed in Cseh et al, 2014). RAS:GTP-bound BRAF heterodimerizes with additional RAF monomers, allowing cis-autophosphorylation in the activation loop of the second RAF protein. Once activated by BRAF in this manner, the 'receiver' kinase monomer is competent to dimerize with and transactivate other monomers in turn (Weber et al, 2001; Garnett et al, 2005; Hu et al, 2013; reviewed in Cseh et al, 2014). Intruigingly, the scaffold protein KSR1 is activated by BRAF in a manner analogous other RAF monomers and can similarly act as a RAF activator once it is itself activated (Brennan et al, 2011; Ory et al, 2003; reviewed in Raabe and Rapp, 2003; Cseh et al, 2014).

Although this pathway shows PP2A-mediated dephosphorylation of RAF and transient displacement of 14-3-3 proteins as preceding RAS and plasma membrane binding of RAF proteins, the order and dependency of these events is not clear. Both membrane binding and 14-3-3 displacement also appear to be facilitated by an interaction between RAF and the cell cycle protein Prohibitin (PHB; Rajalingam et al, 2005; reviewed in Rajalingam and Rudel, 2005; Chowdhury et al, 2014).
R-HSA-5672951 (Reactome) In quiescent cells, RAF is maintained in a closed state in which the N-terminal regulatory region sterically blocks the catalytic region (Cutler et al, 1998; Tran et al, 2003; Terai et al, 2005; Tran et al, 2005; reviewed in Udell et al, 2011). This closed state is mediated in part by the intramolecular binding of YWHAB/14-3-3 dimers to two phosphorylated serine residues (S259 and S621 in RAF1, S214 and S582 in ARAF and S365 and S729 in BRAF) (Ory et al, 2003; Jaumot et al, 2001; Fischer et al, 2009; reviewed in Udell et al, 2011).


R-HSA-5672954 (Reactome) MARK3-mediated phosphorylation of S311 and particularly S406 promotes the binding of 14-3-3 dimers, sequestering KSR1 in the cytosol in quiescent cells (Cacace et al, 1999; Muller et al, 2000; Muller et al, 2001; reviewed in Raabe and Raap, 2003). Mutation of S406 abrogates 14-3-3 binding and results in constitutive plasma membrane localization of KSR1 (Muller et al, 2001).
R-HSA-5672957 (Reactome) Upon growth factor stimulation, KSR1 is dephosphorylated at S406 by PP2A, disrupting 14-3-3 binding and promoting membrane translocation of KSR1 (Ory et al, 2003; Muller et al, 2001; reviewed in Raabe and Raap, 2003).
R-HSA-5672958 (Reactome) Dephosphorylation of KSR1 S406 by PP2A promotes the dissociation of 14-3-3 from this site, exposing both the CR1 region that is required for membrane localization of KSR1 and the MAPK-binding FxFP motif (Ory et al, 2003; Muller et al, 2001; reviewed in Raabe and Raap, 2003).
R-HSA-5672960 (Reactome) Dephosphorylation of S259 (S365/S214) by PP2A promotes the transient dissociation of 14-3-3 dimers from this site (Ory et al, 2003; Jaumot et al, 2001; Rommel et al, 1996; reviewed in Raabe and Raap, 2003; Matallanas et al, 2011). In the case of RAF1, displacement of 14-3-3 has also been shown to be promoted by a direct interaction between RAF1 and the cell cycle protein prohibitin (PHB; Rajalingam et al, 2005; reviewed in Rajalingam and Rudel, 2005; Chowdhury et al, 2014).

R-HSA-5672961 (Reactome) Upon growth factor stimulation, RAF1 S259 (or S365 and S214 in BRAF and ARAF respectively) is dephosphorylated by PP2A and/or PP1, abrogating one of the YWHAB/14-3-3 binding sites (Terai et al, 2005; Ory et al, 2003; Kubicek et al, 2002; Jaumot et al, 2001; Rommel et al, 1996; reviewed in Roskoski, 2010; Matallanas et al, 2011). Release of 14-3-3 binding from the N-terminal site promotes a conformational change that exposes the membrane and RAS interacting RBD and CRD and facilitates recruitment of RAF to the plasma membrane where it binds RAS:GTP (Kubicek et al, 2002; Goetz et al, 2003; Ory et al, 2003). Dephosphorylation of S259/S365/S214 may also promote dimerization of RAF monomers by replacing the intramolecular 14-3-3 binding interaction with an intermolecular one (Rushworth et al, 2006; Weber et al, 2001; Ritt et al, 2010; reviewed in Matallanas et al, 2011). Although the PP2A-mediated dephosphorylation is shown as occurring before both YWHAB displacement and recruitment of RAF to the plasma membrane, the order of and relationship between these events is not completely clear. In addition, the displacement of 14-3-3 and recruitment of RAF1 to the membrane is also promoted by a direct interaction with cell cycle protein Prohibitin (PHB; Rajalingam et al, 2005; reviewed in Rajalingam and Rudel, 2005; Chowdhury et al, 2014).
R-HSA-5672965 (Reactome) The human genome is predicted to encode 27 RAS guanine nucleotide exchange factors (GEFs) that promote the exchange of GDP for GTP on membrane-associated RAS in response to RAS-MAPK pathway activation by growth factors, hormones, cytokines and other stimuli (reviewed in Cherfils and Zeghouf, 2013; Cargnello and Roux, 2011). Nucleotide exchange stimulates a conformational change in RAS to facilitate its interaction with RAF, ultimately promoting the phosphorylation of downstream effectors MAPK3 and MAPK1 (also known as ERK1 and ERK2) (reviewed in Cseh et al, 2014; Vigil et al, 2010).
R-HSA-5672966 (Reactome) RAF activation depends on the formation of a side-by-side asymmetric homo- or heterodimer (formed from either 2 RAF monomers or a RAF monomer and KSR1) (Weber et al, 2001; Garnett et al, 2005; Rushworth et al, 2006; Rajakulendran et al, 2009; Hu et al, 2013). Dimerization is mediated by cluster of basic residues in the kinase domain, and mutation of these critical residues abrogates RAF activation (Rajakulendran et al, 2009). Dimerization is required for the 'activator' monomer to induce an allosteric change in the 'receiver' monomer that, in conjunction with activation loop phosphorylation, activates the kinase activity of the receiver (Hu et al, 2013). BRAF, by virtue of its constitutive negative charge in the NtA region, is uniquely able to function as an activator RAF without further modification, while RAF1, ARAF and KSR1 can function as activators after being phosphorylated in the NtA region downstream of RAF pathway activation (Hu et al, 2013; Leicht et al, 2013; reviewed in Cseh et al, 2014). Homo and heterodimerization of RAF monomers may be promoted by association with MAP3K11, which interacts with BRAF and RAF1 in vitro and in vivo and which is required for RAF activation (Chadee et al, 2004a, Chadee et al, 2004b; Chadee et al, 2006).
R-HSA-5672969 (Reactome) Downstream of RAF dimerization and allosteric activation, RAF monomers and KSR1 undergo a series of activating phosphorylations in both the activation loop (AL) and, in the case of ARAF, RAF1 and KSR1, in the NtA . Phosphorylation of the activation loop residues (T491, S494 in RAF1, T452, T455 in ARAF and T599, S602 in BRAF) contributes to full kinase activity, although this may be less critical for ARAF, and RAFs in general, than for other kinases due to their regulation by 14-3-3 binding (Zhu et al, 2005; Zhang et al, 2000; Baljuls et al, 2008, reviewed in Matallanas et al, 2011; Udell et al, 2011). AL phosphorylation may occur through cis-autophosphorylation within the RAF dimer, although phosphorylation by other kinases is also possible (Hu et al, 2013; reviewed in Matallanas et al, 2011).

Phosphorylation in the RAF NtA region is required for full kinase activity, for interaction with MAP2K substrates and for the ability of activated RAF to act as an allosteric activator of other RAF monomers (Marais et al, 1995; Diaz et al, 1997; Xiang et al, 2002; Edin et al, 2005; Hu et al, 2013; Mason et al, 1999). Phosphorylation of these residues (S338 and Y441 in RAF1, S299 and Y302 in ARAF and Y602 in KSR1) may be mediated by a kinase of the SRC, JAK or PAK family kinases, by MAP2K kinases or through autophosphorylation by RAF itself (Marais et al, 1995; Xia et al, 1996; King et al, 1998; Sun et al, 2000; Tran et al, 2003; Tran et al, 2005; Hu et al, 2013; reviewed in Matallanas et al, 2011). The phosphorylated NtA of RAF1 is also the binding site for the negative regulator PEPB1, also known as RKIP. PEBP1 binding to RAF1 prevents phosphorylation of the MAP2K substrates (Park et al, 2006; Rath et al, 2008; reviewed in Shin et al, 2009).
R-HSA-5672972 (Reactome) RAF kinases have restricted substrate specificity and have as their primary substrates the two MAP2K proteins MAP2K1 and MAP2K2 (also known as MEK1 and 2). MAP2K1 knockout is embryonic lethal in mice, while MAP2K2 knockouts have no apparent abnormalities, suggesting that MAP2K1 can compensate for MAP2K2 in vivo (Giroux et al, 1999; Belanger et al, 2003). MAP2K proteins exist as stable homo- and heterodimers independent of growth factor stimulation and are generally recruited to activated RAF proteins in conjunction with a scaffolding protein and the MAP2K substrates, MAPK1 and 3 (also known as ERK1 and 2) (Ohren et al, 2004; Catalanotti et al, 2009; Catling et al, 1995; reviewed in Matallanas et al, 2011; Roskoski et al, 2012a; Roskoski et al, 2012b).

Scaffolding proteins promote signaling by providing a docking platform that colocalizes components of the signaling cascade, and provide specificity by controlling the spatial and temporal regulation of the pathway (reviewed in Brown and Sacks, 2009; Matallanas et al, 2011). KSR1 and 2, CNKSR1 and 2, IQGAP1 and the beta arrestins are among the known MAPK scaffold proteins that act at the plasma membrane upon MAPK pathway activation; in addition, paxillin localizes MAPK pathway components to focal adhesion sites in the plasma membrane (Roy et al, 2005; Ren et al, 2007; DeFea et al, 2000; Togho et al, 2003; Ishibe et al, 2003; reviewed in Claperon and Therrien, 2007; Brown and Sacks, 2009; Matallanas et al, 2011). Although this reaction depicts these scaffolding proteins acting equivalently, the details of how they promote pathway activation vary. For instance, KSR1 and 2 are constitutively bound to MAP2K dimers but recruit MAPKs only upon pathway stimulation, while IQGAP1 associates constitutively with both MAP2K and MAPK proteins in unstimulated cells and shows increased interaction with MAP2K1 upon pathway activation by EGF (Stewart et al, 1999; Cacace et al, 2000; Muller et al, 2000; Roy et al, 2004; Roy et al, 2005; reviewed in Brown and Sacks, 2009). Scaffolding complexes may be particularly important for the phosphorylation of cytosolic MAPK targets (reviewed in Casar et al, 2009).
R-HSA-5672973 (Reactome) Activated MAP2K phosphorylates MAPK on threonine and tyrosine residues in the activation loop (residues T202 and Y204 in MAPK3, residues T185 and Y187 in MAPK1) (Ray et al, 1988; reviewed in Roskoski, 2012b). MAPK3 and MAPK1 are 84% identical and appear to be stimulated in parallel by all known activators of the MAPK pathway (Lefloch et al, 2009; reviewed in Lloyd, 2006).
R-HSA-5672978 (Reactome) Activated RAF phosphorylates the MEK kinases MAP2K1 and MAP2K2 on 2 serine residues in the MAP2K activation loop (S218 and S222 in MAP2K1 and S222 and S226 in MAP2K2 (Zheng and Guan, 1994; Alessi et al, 1994; Catling et al, 1995; Papin et al, 1995; Seger et al, 1994; reviewed in Roskoski, 2012a). Although all three RAF kinases can phosphorylate MAP2K1 and MAP2K2, BRAF appears to be the primary activator in vivo (Marais et al, 1997; Jaiswal et al, 1994; Pritchard et al, 1995; reviewed in Welbrock et al, 2004)
R-HSA-5672980 (Reactome) The mechanisms governing the dissociation or trafficking of activated MAPK signaling complexes at the plasma membrane is not fully worked out. Some active complexes may be endocytosed and targeted to other cellular locations, for example the Golgi complex (Lorentzen et al, 2010). Activated RAF monomers may dissociate and homo- or heterodimerize with additional inactive RAF monomers and in this way amplify the signal (reviewed in Matallanas et al, 2011; Cseh et al, 2014).
Ultimately, active RAF complexes are subject to PP5- and PP2A-mediated dephosphorylation, which promotes a return to the inactive state. Hydrolysis of RAS-bound GTP by the intrinsic GTPase activity, stimulated by association with RAS GAP proteins, ultimately promotes dissociation of RAS from RAF allowing a return to the quiescent state (reviewed in Wellbrock et al, 2004; Matallanas et al, 2011).
R-HSA-5674018 (Reactome) BRAP is a negative regulator of the RAF/MAPK cascade that inhibits the homo- and heterodimerization of KSR1 and RAF and preventing downstream signal propagation (Matheny et al, 2004; Chen et al, 2008; reviewed in Matheny et al, 2009). Upon RAS stimulation, BRAP binds to RAS:GTP. This stimulates BRAP's E3 HECT ubiquitin ligase activity, promoting its autoubiquitination and thereby relieving the inhibition of KSR1 activity (Matheny et al, 2004; reviewed in Matheny et al, 2009). USP15 is a deubiquitinase that stabilizes BRAP protein levels and thus acts to dampen MAPK signaling (Hayes et al, 2013).
R-HSA-5674019 (Reactome) BRAP is a negative regulator of MAPK signaling that binds KSR1 as assessed by coimmunoprecipitation. This interaction abrogates KSR1 homodimer and KSR1:RAF heterodimer formation, and disrupts the recruitment of MAP2K kinases to RAF (Methany et al, 2004; Chen et al, 2008; reviewed in Methany et al, 2009). BRAP inhibition of KSR1 is relieved in an unknown manner by autoubiquitination after RAS pathway activation (reviewed in Methany et al, 2009).
R-HSA-5674022 (Reactome) Binding to activated RAS stimulates the ubiquitinase activity of BRAP, promoting autoubiquitination and relieving the inhibition of KSR1 (Methany et al, 2004; Chen et al, 2008; Methany et al, 2009).
R-HSA-5674130 (Reactome) Interaction of MAPK pathway components with LAMTOR2, 3 and MORG1 facilitates pathway activation in response to varied stimuli, resulting in the phosphorylation of MAP2K and MAPK proteins at conserved sites in their activation loops (Schaeffer et al, 1998; Teis et al, 2002; Teis et al, 2006; Vomastek et al, 2004; Sharma et al, 2005; reviewed in Matallanas et al, 2011).
R-HSA-5674132 (Reactome) LAMTOR3 (also known as MEK partner 1, MP1) exists in an obligatory complex with LAMTOR2 (p14) at the endosomal membrane where they act as a scaffold and promote MAPK activation (Schaeffer et al, 1998; Teis et al, 2002; Teis et al, 2006; Sharma et al, 2005). The LAMTOR2/LAMTOR3 complex may also be part of a larger molecular weight complex at the endosome that includes the MAPK organizer protein MORG1 (Vomastek et al, 2004; Sharma et al, 2005; reviewed in Matallanas et al, 2011).
R-HSA-5674140 (Reactome) PAQR3, also known as RKTG (Raf kinase trapping to Golgi) is a multi-pass transmembrane protein that binds to RAF1 and BRAF and sequesters them in the Golgi. This inhibits the interaction of RAF with activated RAS and the plasma membrane and inhibits RAF signaling (Feng et al, 2007; Fan et al, 2008; Luo et al, 2008).
R-HSA-5674366 (Reactome) IL17RD, also known as SEF (similar expression to FGF), was identified as a negative regulator of nuclear MAPK signaling (Tsang et al, 2002; Furthauer et al, 2002). IL17RD is a spatial regulator of MAPK signaling that binds activated MAP2K dimers at the Golgi membrane and prevents the dissociation and translocation of phosphorylated MAPK into the nucleus. In this way, IL17RD restricts activation of nuclear MAPK targets while not affecting activation of cytosolic ones (Torii et al, 2004; reviewed in Phillips, 2004; Matallanas et al, 2011).
R-HSA-5674373 (Reactome) Activated MAP2Ks in complex with IL17RD phosphorylate MAPKs at the Golgi membrane. IL17RD prevents the dissociation of phosphorylated MAPK from the complex at the Golgi as assessed by coimmunoprecipitation, preventing MAPK nuclear translocation and activation of nuclear targets (Torii et al, 2004; reviewed in Philips, 2004; Brown and Sacks, 2009).
R-HSA-5674385 (Reactome) Phosphorylated MAPK monomers can dimerize - generally into MAPK1 and MAPK3 homodimers, as the heterodimer is unstable- but the physiological significance of dimerization is unclear (Khokhlatchev et al, 1998; reviewed Rosokoski, 2012b). MAPKs have both cytosolic and nuclear targets and dimerization may be particularly important for MAPK-dependent phosphorylation of cytosolic targets. Phosphorylation of cytosolic MAPK targets appears to happen predominantly in the context of larger scaffolding complexes, and since the scaffolds and cytosolic MAPK substrates contact the same hydrophobic surface of MAPK, dimerization is necessary to allow assembly of a functional complex (Casar et al, 2008; Lidke et al, 2010; reviewed in Casar et al, 2009). Consistent with this, disrupting either MAPK dimerization or the MAPK interaction with the scaffolding protein abrogated proliferation and transformation (Casar et al, 2008). Note that, for simplicity in this diagram, dimerization is shown as happening between free cytosolic monomers of activated MAPK rather than in the context of the scaffolding complex.
Although predominantly cytoplasmic in resting cells, a proportion of activated MAPK translocates to the nucleus upon stimulation where it activates nuclear targets. Despite early studies to the suggesting that dimerization was required for nuclear translocation, a few recent papers have challenged this notion (Lenormand et al, 1993; Chen et al, 1992; Khokhlatchev et al, 1998; Casar et al, 2008; Lidke et al, 2010; Burack and Shaw, 2005; reviewed in Roskoski, 2012b).
R-HSA-5674387 (Reactome) After phosphorylation by MAP2Ks, a proportion of activated MAPK translocates into the nucleus where it activates nuclear targets (reviewed in Roskoski, 2012b). MAPKs, which lack a nuclear localization signal (NLS), may 'piggyback' into the nucleus in complex with other nuclear-targeted proteins or may translocate by virtue of interaction with components of the nuclear pore complex (Brunet et al, 1999; Adachi et al, 1999; Matsubayashi et al, 2001; Whitehurst et al, 2002; Khokhlatchev et al, 1998; reviewed in Roskoski, 2012b). Although dimerization of MAPKs was thought to be critical for nuclear translocation, a number of studies have now challenged the physiological relevance of MAPK dimerization and this remains an area of uncertainty (Lenormand et al, 1993; Chen et al, 1992; Casar et al, 2008; Lidke et al, 2010; Burack and Shaw, 2005; reviewed in Casar et al, 2009; Roskoski, 2012b)
R-HSA-5674496 (Reactome) Activated MAPK proteins negatively regulate MAP2K1:MAP2K2 heterodimers by phosphorylating MAP2K1 at T292, a residue that is not present in MAP2K2. Phosphorylation of this site in MAP2K1 promotes the dephosphorylation of the MAP2K phosphorylated activation loop (AL) by an unknown mechanism, establishing a negative feedback loop that limits MAPK signaling (Catalanotti et al, 2009; Brunet et al, 1994; Xu et al, 1999). Deletion of MAP2K1 or mutation of this site prolongs MAP2K2 AL phosphorylation and MAPK activation (Catalanotti et al, 2009).
R-HSA-5675194 (Reactome) RAF1 is phosphorylated by activated MAPK at 6 serine residues (S29, S43, S289, S296, S301 and S642). MAPK-dependent hyperphosphorylation of RAF1 abrogates the ability of activated RAF1 to interact with RAS and is coincident with inactivation of RAF1. RAF1 proteins containing mutation of these phosphorylation sites persist at the plasma membrane, show sustained S338 phosphorylation and persistent activation relative to WT RAF1 protein. In wild type cells, PP2A and the prolyl-isomerase PIN1 contribute to the dephosphorylation of hyperphosphorylated RAF1, allowing subsequent cycles of activation to occur (Dougherty et al, 2005; reviewed in Roskoski, 2010)
R-HSA-5675198 (Reactome) BRAF is subject to MAPK-dependent phosphorylation that limits its activity. Phosphorylation of S151 inhibits binding of BRAF to activated RAS, while phosphorylation of T401, S750 and S753 abrogates heterodimerization with RAF1 (Ritt et al, 2010; Rushworth et al, 2006; Brummer et al, 2003).
R-HSA-5675206 (Reactome) PEA15 is a cytoplasmic anchor that binds directly to activated MAPKs prevents their translocation into the nucleus (Formstecher et al, 2001; Whitehurst et al, 2004; Hill et al, 2002; Chou et al, 2003). PEA15 also protects phosphorylated MAPKs in the cytoplasm from inactivating dephosphorylation (Mace et al, 2013). In this way, binding of PEA15 promotes phosphorylation of cytoplasmic MAPK targets at the expense of nuclear ones.
R-HSA-5675373 (Reactome) MAPKs are inactivated by dephosphorylation of the activation loop T and Y residues by dual-specificity MAPK phosphatases (DUSPs) (reviewed in Roskoski, 2012b). Class 1 DUSPs, including DUSP 1, 2, 4 and 5 are nuclear and are generally activated by the same extracellular stimuli that promote MAPK signaling, establishing a negative feedback loop. DUSP5 is specific for MAPK3 and 1, while the other class 1 enzymes have broad specificity. Nuclear MAPKs may also be inactivated by nuclear forms of class III DUSPs, including DUSP8, 10 and 16, although the preferred substrate of these enzymes are the p38 and JNK MAP kinases (reviewed in Bermudez et al, 2010; Kondoh and Nishida, 2007).
R-HSA-5675376 (Reactome) MAPKs are inactivated by dephosphorylation of the activation loop T and Y residues by dual-specificity MAPK phosphatases (DUSPs) (reviewed in Roskoski, 2012b). Cytosolic MAPKs are dephosphorylated by the MAPK-specific class II DUSPs 6,7 and 9, but may also be dephosphorylated by cytosolic forms of class III DUSPs 8, 10 and 16, which preferentially dephosphorylate p38 and JNK MAP kinases (reviewed in Bermudez et al, 2010; Kandoh and Nishida, 2007).
R-HSA-5675417 (Reactome) PEBP1, also known as RKIP (Raf kinase inhibitor protein), is a negative regulator of RAF1 that binds to the phosphorylated NtA region and prevents activation of MAP2K substrates (Yeung et al, 1999; Yeung et al, 2000; Rath et al, 2008; Park et al, 2006; reviewed in Lorenz et al, 2014). Relief of this PEBP1 repression of RAF1 activity is stimulated by phosphorylation of PEBP1, which promotes it dissociation from RAF1. Candidate kinases for phosphorylation of PEBP1 include PKC and the MAPKs themselves, which would establish a positive feedback loop stimulating MAPK pathway activity (Corbit et al, 2003; Cho et al, 2003; Shin et al, 2009).
R-HSA-5675431 (Reactome) Along with PPP5C-mediated dephophosphorylation of the NtA region, PP2A contributes to the inactivation of RAF1 by mediating the dephosphorylation of AL loop residues. PP2A-mediated dephosphorylation of RAF1 may be stimulated by the prior hyperphosphorylation of RAF1 by MAPKs (Dougherty et al, 2005; reviewed in Matallanas et al, 2011).
R-HSA-5675433 (Reactome) PPP5C dephosphorylates S338 in the NtA region of RAF1, which reduces the catalytic activity of RAF1 towards MAP2K proteins (von Kriegsheim et al, 2006; reviewed in Matallanas et al, 2011).
RAF activating kinasesmim-catalysisR-HSA-5672969 (Reactome)
RAF/MAPK scaffoldsArrowR-HSA-5672980 (Reactome)
RAF/MAPK scaffoldsR-HSA-5672972 (Reactome)
RAS GAPsArrowR-HSA-5658231 (Reactome)
RAS GAPsR-HSA-5658435 (Reactome)
RAS GEFsmim-catalysisR-HSA-5672965 (Reactome)
SPRED dimer:NF1ArrowR-HSA-5658231 (Reactome)
SPRED dimer:NF1ArrowR-HSA-5658438 (Reactome)
SPRED dimer:NF1R-HSA-5658424 (Reactome)
SPRED dimer:ub-NF1ArrowR-HSA-5658424 (Reactome)
SPRED dimer:ub-NF1R-HSA-5658430 (Reactome)
SPRED dimerArrowR-HSA-5658430 (Reactome)
SPRED dimerR-HSA-5658438 (Reactome)
UbR-HSA-5658424 (Reactome)
UbR-HSA-5674022 (Reactome)
WDR83:LAMTOR2:LAMTOR3:activated RAF:MAP2K:MAPK complexArrowR-HSA-5674132 (Reactome)
WDR83:LAMTOR2:LAMTOR3:activated RAF:MAP2K:MAPK complexR-HSA-5674130 (Reactome)
WDR83:LAMTOR2:LAMTOR3:activated RAF:p-2S MAP2K:p-T,Y MAPK complexArrowR-HSA-5674130 (Reactome)
WDR83:LAMTOR2:LAMTOR3R-HSA-5674132 (Reactome)
YWHAB dimerArrowR-HSA-5672958 (Reactome)
YWHAB dimerArrowR-HSA-5672960 (Reactome)
YWHAB dimerR-HSA-5672951 (Reactome)
YWHAB dimerR-HSA-5672954 (Reactome)
activated RAF:scaffold:MAP2K:MAPK complexArrowR-HSA-5672972 (Reactome)
activated RAF:scaffold:MAP2K:MAPK complexR-HSA-5672978 (Reactome)
activated RAF:scaffold:MAP2K:MAPK complexmim-catalysisR-HSA-5672978 (Reactome)
activated

RAF:scaffold:p-2S

MAP2K:MAPK complex
ArrowR-HSA-5672978 (Reactome)
activated

RAF:scaffold:p-2S

MAP2K:MAPK complex
R-HSA-5672973 (Reactome)
activated

RAF:scaffold:p-2S

MAP2K:MAPK complex
mim-catalysisR-HSA-5672973 (Reactome)
activated

RAF:scaffold:p-2S MAP2K:p-2T MAPK

complex
ArrowR-HSA-5672973 (Reactome)
activated

RAF:scaffold:p-2S MAP2K:p-2T MAPK

complex
R-HSA-5672980 (Reactome)
activated RAF homo/heterodimerR-HSA-5674132 (Reactome)
cytosolic MAPK DUSPsmim-catalysisR-HSA-5675376 (Reactome)
dephosphorylated "receiver" RAF/KSR1R-HSA-5672966 (Reactome)
dephosphorylated

inactive RAFS:YWHAB

dimer
ArrowR-HSA-5672961 (Reactome)
dephosphorylated

inactive RAFS:YWHAB

dimer
R-HSA-5672960 (Reactome)
dephosphorylated inactive RAFsArrowR-HSA-5672960 (Reactome)
hyperphosphorylated BRAFArrowR-HSA-5675198 (Reactome)
hyperphosphorylated RAF1ArrowR-HSA-5675194 (Reactome)
hyperphosphorylated RAF1ArrowR-HSA-5675431 (Reactome)
inactive RAFs:YWHAB dimerArrowR-HSA-5672951 (Reactome)
inactive RAFs:YWHAB dimerR-HSA-5672961 (Reactome)
inactive RAFsR-HSA-5672951 (Reactome)
inactive RAFsR-HSA-5674140 (Reactome)
nuclear MAPK DUSPsmim-catalysisR-HSA-5675373 (Reactome)
p-2S MAP2K homo/heterodimersArrowR-HSA-5672980 (Reactome)
p-2S MAP2K homo/heterodimersR-HSA-5674366 (Reactome)
p-2S MAP2K1:p-2S MAPK2K2 heterodimerR-HSA-5674496 (Reactome)
p-2S,T MAP2K1:p-2S MAPK2K2 heterodimerArrowR-HSA-5674496 (Reactome)
p-S311

KSR1:MARK3:YWHAB

dimer
ArrowR-HSA-5672957 (Reactome)
p-S311

KSR1:MARK3:YWHAB

dimer
R-HSA-5672958 (Reactome)
p-S311 KSR1:MARK3ArrowR-HSA-5672958 (Reactome)
p-S311,S406

KSR1:MARK3:YWHAB

dimer
ArrowR-HSA-5672954 (Reactome)
p-S311,S406

KSR1:MARK3:YWHAB

dimer
R-HSA-5672957 (Reactome)
p-S311,S406 KSR1:MARK3ArrowR-HSA-5672948 (Reactome)
p-S311,S406 KSR1:MARK3R-HSA-5672954 (Reactome)
p-S338,Y341,T491,S494,S621 RAF1R-HSA-5675194 (Reactome)
p-S445,T599,S602,S729 BRAFR-HSA-5675198 (Reactome)
p-T,Y MAPK dimersArrowR-HSA-5674385 (Reactome)
p-T,Y MAPK monomers and dimers:PEA15ArrowR-HSA-5675206 (Reactome)
p-T,Y MAPK monomers and dimers:PEA15TBarR-HSA-5674387 (Reactome)
p-T,Y MAPK monomers and dimersArrowR-HSA-5674387 (Reactome)
p-T,Y MAPK monomers and dimersR-HSA-5674387 (Reactome)
p-T,Y MAPK monomers and dimersR-HSA-5675206 (Reactome)
p-T,Y MAPK monomers and dimersR-HSA-5675373 (Reactome)
p-T,Y MAPK monomers and dimersR-HSA-5675376 (Reactome)
p-T,Y MAPK monomers and dimersmim-catalysisR-HSA-5674496 (Reactome)
p-T,Y MAPK monomers and dimersmim-catalysisR-HSA-5675194 (Reactome)
p-T,Y MAPK monomers and dimersmim-catalysisR-HSA-5675198 (Reactome)
p-T,Y MAPKsArrowR-HSA-5672980 (Reactome)
p-T,Y MAPKsR-HSA-5674385 (Reactome)
p21

RAS:GTP:'activator'

RAF:YWHAB dimer
ArrowR-HSA-5672950 (Reactome)
p21

RAS:GTP:'activator'

RAF:YWHAB dimer
R-HSA-5672966 (Reactome)
p21

RAS:GTP:activated RAF homo/heterodimer

complexes
ArrowR-HSA-5672969 (Reactome)
p21

RAS:GTP:activated RAF homo/heterodimer

complexes
R-HSA-5672972 (Reactome)
p21

RAS:GTP:activated RAF1

homo/heterodimer:PEBP1
ArrowR-HSA-5675417 (Reactome)
p21

RAS:GTP:activated RAF1

homo/heterodimer:PEBP1
TBarR-HSA-5672972 (Reactome)
p21 RAS:GTP:homo/heterodimerized RAF complexArrowR-HSA-5672966 (Reactome)
p21 RAS:GTP:homo/heterodimerized RAF complexR-HSA-5672969 (Reactome)
p21 RAS:GDPArrowR-HSA-5658231 (Reactome)
p21 RAS:GDPR-HSA-5672965 (Reactome)
p21 RAS:GTP:BRAPArrowR-HSA-5674018 (Reactome)
p21 RAS:GTP:BRAPR-HSA-5674022 (Reactome)
p21 RAS:GTP:BRAPmim-catalysisR-HSA-5674022 (Reactome)
p21 RAS:GTP:RAS GAPsArrowR-HSA-5658435 (Reactome)
p21 RAS:GTP:RAS GAPsR-HSA-5658231 (Reactome)
p21 RAS:GTP:RAS GAPsmim-catalysisR-HSA-5658231 (Reactome)
p21 RAS:GTP:ub-BRAPArrowR-HSA-5674022 (Reactome)
p21 RAS:GTPArrowR-HSA-5672965 (Reactome)
p21 RAS:GTPR-HSA-5658435 (Reactome)
p21 RAS:GTPR-HSA-5672950 (Reactome)
p21 RAS:GTPR-HSA-5674018 (Reactome)
p21RAS:GTP:activated

RAF

homo/heterodimers
ArrowR-HSA-5672980 (Reactome)
p21RAS:GTP:activated

RAF1

homo/heterodimer
R-HSA-5675417 (Reactome)
p21RAS:GTP:activated

RAF1

homo/heterodimer
R-HSA-5675433 (Reactome)
p21RAS:GTP:dephosphorylated RAF1 homo/heterodimerArrowR-HSA-5675433 (Reactome)
p21RAS:GTP:dephosphorylated RAF1 homo/heterodimerR-HSA-5675431 (Reactome)
p21RAS:GTP:p-S621

RAF1

homo/heterodimer
ArrowR-HSA-5675431 (Reactome)
Personal tools