Hedgehog 'off' state (Homo sapiens)

From WikiPathways

Jump to: navigation, search
9, 18, 20, 441, 3, 4, 8, 12...2, 22, 6034, 74, 783, 10, 38, 40, 50...9420, 839453, 70, 72, 927, 21, 24, 48, 819, 35, 4127, 9427, 45, 945, 6, 9, 11, 13...2, 38, 46, 60, 73...24, 3570, 7227, 70, 72, 92, 947, 9, 21, 24, 35...2, 19, 30, 60, 8727, 942, 22, 60, 8770, 7224, 35741, 3, 4, 8, 12...34, 74, 789, 35, 4134, 7834, 7814, 15, 32, 36, 37, 42...27, 28, 53, 70, 7227, 45, 70, 74cytosolendocytic vesicle lumenciliumnucleoplasmUBC(381-456) UBB(1-76) PSMA7 PSMB1 PKA regulatorysubunits:cAMPPSME2 PSMD12 PSMB7 PSMD6 UBC(381-456) IFT172 SUFU PSMD10 p6S-GLI3:SUFUGLI2UBC(1-76) PSMA4 GLI1 UBC(153-228) RBX1 ADPINTU IFT122 ATPPSMD9 SKP1 PSMD7 PSMD10 SMO UBC(533-608) PSMD14 PSMB5 CSNK1A1pS-GLI1:SUFUp11S-GLI2:SUFUPSMD14 ADPUbGTP UBC(533-608) GLI2 gene:GLI3RPSMA5 PSMB2 CUL1 ADCY2 PSMA3 ATPGLI2:SUFUPSMC2 ADPPSMA6 PTCH1 gene PSMB4 GLI1 gene ATPNUMB:ITCH:ub-pS-GLI1:SUFUIFT140 WDR19 UBB(1-76) PSMF1 p8S-GLI2:SUFUSHFM1 TTC21B OFD1 UBB(153-228) ADCY3 SUFU PSMB11 BTRC p10S-GLI3 IFT88 ADCY9 GLI1 geneGLI2 gene PSME4 adenylatecyclases:Mg2+UBB(153-228) GLI3:SUFUSKP1 UBC(77-152) UBC(609-684) ub-pS-GLI1 GLI1 anterograde IFTregulators of HhPSMC3 PSMA3 GNAS2 PSMC6 PSMA2 IFT122 DYNC2H1 ciliary basal bodyregulators of HhPSME3 retrograde IFTregulators of HhUBC(229-304) SMO dimerPSMB8 PRKAR2B WDR19 SUFU GLI2 SUFU p4S-GLI2 PRKAR2A PSMD5 GPR161PSMD3 GLI2 KIF3A UBC(153-228) KIF7:microtubuleGLI1 p11S-GLI2 GLI3R PRKACB GLI3 SUFU PSMD4 PSMC2 IFT52 SUFU SUFU SMO IFT57 SUFU ITCHub-pS-GLI1 UBB(77-152) PSMD1 NUMB UBC(1-76) SMO dimerADCY8 PRKAR2B G alpha (s):GTPIFT140 RPGRIP1L p4S-GLI2:SUFUPSME2 BTRC:CUL1:RBX1:SKP1ADCY5 PSMB10 UBC(305-380) IFT122 SUFU TTC21B PTCH1 geneGLI3 ATPWDR35 TTC21B KIF7 p13S-GLI3:SUFUPSMB1 GLI3R SHFM1 IFT-A complex:TULP3GLI3 PSMD1 WDR35 ATPGLI1:SUFUPSMB8 PPiPSMD13 UBB(153-228) ADCY10 PCP regulators of HhTULP3RPS27A(1-76) PSMB3 p6S-GLI3 PSMC4 PSMD8 p13S-GLI3 GLI3RPSMD2 UBC(1-76) UBC(457-532) PSMB7 UBC(229-304) PSMA8 NUMB cAMPADCY6 PSMC5 PRKACG PSMA4 GLI3 PRKAR1A GPR161:IFT-A:TULP3GPR161 p8S-GLI2 GLI2 geneGLI1ADCY1 PRKAR2A SUFU p10S-GLI3:SUFUSUFU PSMC1 UbBTRC CUL1 UBC(609-684) PSMC3 PSMD11 TULP3 SUFU GLI2 ub-p13S-GLI3 PKA tetramerPSMB9 FUZ UBC(457-532) UBB(77-152) PSMB4 UBC(305-380) PKA catalyticsubunitWDR35 UBA52(1-76) SUFU RBX1 ADCY4 PSMA8 PRKACG ub-p11S-GLI2:SUFUGSK3BPTCH1 gene:GLI3RPSMB9 IFT122 GLI3 PSMC4 PRKACA GLI1 PSMA5 UBB(1-76) GLI1,2,3microtubule Mg2+ SUFUPSMD9 PSMD5 PTCH1PSME4 NUMBKIF7SUFU PRKAR1B GLI3RPSMA6 PSMB2 26S proteasomeSUFU 26S proteasomePSMD7 PSMB6 ADCY7 PSMC5 PSMD2 UBC(77-152) RPS27A(1-76) ADPUBA52(1-76) PSMB10 UBC(153-228) SUFU UBC(305-380) UBC(533-608) PSMD4 RPS27A(1-76) PSMD3 PSMB5 ATPPSMD11 PSME1 GLI:SUFUGLI2 PRKAR1B pS-GLI1 GLI:SUFUADPGLI2 PSMD6 GNAS1 UbGLI:SUFUPSMB3 IFT140 microtubuleTTC21B UBC(609-684) PSMB6 ITCH PSMA1 PRKAR1A UBC(77-152) PSME1 PSMA7 cAMP MKS1 ub-p11S-GLI2 ATPGLI1 gene:GLI3RADPIFT-A complexUBC(381-456) PSMF1 ADPPSMC6 ub-pS-GLI1:SUFUNUMB:ITCHGLI3R 4xub-p13S-Gli3:SUFUGLI1 UBC(457-532) PSMB11 PSME3 PSMA2 PRKACA SUFU PSMD12 PRKACB UBB(77-152) PSMD13 TULP3 PSMC1 BTRC:CUL1:RBX1:SKP1ITCH ATPPSMA1 WDR19 PSMD8 UBA52(1-76) UBC(229-304) 17, 797, 2170, 723575871717, 7970, 7270, 721770, 728770, 7235


Description

Hedgehog is a secreted morphogen that has evolutionarily conserved roles in body organization by regulating the activity of the Ci/Gli transcription factor family. In Drosophila in the absence of Hh signaling, full-length Ci is partially degraded by the proteasome to generate a truncated repressor form that translocates to the nucleus to represses Hh-responsive genes. Binding of Hh ligand to the Patched (PTC) receptor allows the 7-pass transmembrane protein Smoothened (SMO) to be activated in an unknown manner, disrupting the partial proteolysis of Ci and allowing the full length activator form to accumulate (reviewed in Ingham et al, 2011; Briscoe and Therond, 2013).
While many of the core components of Hh signaling are conserved from flies to humans, the pathways do show points of significant divergence. Notably, the human genome encodes three Ci homologues, GLI1, 2 and 3 that each play slightly different roles in regulating Hh responsive genes. GLI3 is the primary repressor of Hh signaling in vertebrates, and is converted to the truncated GLI3R repressor form in the absence of Hh. GLI2 is a potent activator of transcription in the presence of Hh but contributes only minimally to the repression function. While a minor fraction of GLI2 protein is processed into the repressor form in the absence of Hh, the majority is either fully degraded by the proteasome or sequestered in the full-length form in the cytosol by protein-protein interactions. GLI1 lacks the repression domain and appears to be an obligate transcriptional activator (reviewed in Briscoe and Therond, 2013).
Vertebrate but not fly Hh signaling also depends on the movement of pathway components through the primary cilium. The primary cilium is a non-motile microtubule based structure whose construction and maintenance depends on intraflagellar transport (IFT). Anterograde IFT moves molecules from the ciliary base along the axoneme to the ciliary tip in a manner that requires the microtubule-plus-end directed kinesin KIF3 motor complex and the IFT-B protein complex, while retrograde IFT back to the ciliary base depends on the minus-end directed dynein motor and the IFT-A complex. Genetic screens have identified a number of cilia-related proteins that are required both to maintain Hh in the 'off' state and to transduce the signal when the pathway is activated (reviewed in Hui and Angers, 2011; Goetz and Anderson, 2010). View original pathway at Reactome.

Comments

Reactome-Converter 
Pathway is converted from Reactome ID: 5610787
Reactome-version 
Reactome version: 75
Reactome Author 
Reactome Author: Rothfels, Karen

Try the New WikiPathways

View approved pathways at the new wikipathways.org.

Quality Tags

Ontology Terms

 

Bibliography

View all...
  1. Houde C, Dickinson RJ, Houtzager VM, Cullum R, Montpetit R, Metzler M, Simpson EM, Roy S, Hayden MR, Hoodless PA, Nicholson DW.; ''Hippi is essential for node cilia assembly and Sonic hedgehog signaling.''; PubMed Europe PMC Scholia
  2. Mukhopadhyay S, Rohatgi R.; ''G-protein-coupled receptors, Hedgehog signaling and primary cilia.''; PubMed Europe PMC Scholia
  3. He M, Subramanian R, Bangs F, Omelchenko T, Liem KF, Kapoor TM, Anderson KV.; ''The kinesin-4 protein Kif7 regulates mammalian Hedgehog signalling by organizing the cilium tip compartment.''; PubMed Europe PMC Scholia
  4. Zeng H, Hoover AN, Liu A.; ''PCP effector gene Inturned is an important regulator of cilia formation and embryonic development in mammals.''; PubMed Europe PMC Scholia
  5. Rohatgi R, Milenkovic L, Scott MP.; ''Patched1 regulates hedgehog signaling at the primary cilium.''; PubMed Europe PMC Scholia
  6. Denef N, Neubüser D, Perez L, Cohen SM.; ''Hedgehog induces opposite changes in turnover and subcellular localization of patched and smoothened.''; PubMed Europe PMC Scholia
  7. Vokes SA, Ji H, Wong WH, McMahon AP.; ''A genome-scale analysis of the cis-regulatory circuitry underlying sonic hedgehog-mediated patterning of the mammalian limb.''; PubMed Europe PMC Scholia
  8. Heydeck W, Zeng H, Liu A.; ''Planar cell polarity effector gene Fuzzy regulates cilia formation and Hedgehog signal transduction in mouse.''; PubMed Europe PMC Scholia
  9. Hui CC, Angers S.; ''Gli proteins in development and disease.''; PubMed Europe PMC Scholia
  10. Maurya AK, Ben J, Zhao Z, Lee RT, Niah W, Ng AS, Iyu A, Yu W, Elworthy S, van Eeden FJ, Ingham PW.; ''Positive and negative regulation of Gli activity by Kif7 in the zebrafish embryo.''; PubMed Europe PMC Scholia
  11. Huang S, Zhang Z, Zhang C, Lv X, Zheng X, Chen Z, Sun L, Wang H, Zhu Y, Zhang J, Yang S, Lu Y, Sun Q, Tao Y, Liu F, Zhao Y, Chen D.; ''Activation of Smurf E3 ligase promoted by smoothened regulates hedgehog signaling through targeting patched turnover.''; PubMed Europe PMC Scholia
  12. Park TJ, Haigo SL, Wallingford JB.; ''Ciliogenesis defects in embryos lacking inturned or fuzzy function are associated with failure of planar cell polarity and Hedgehog signaling.''; PubMed Europe PMC Scholia
  13. Corbit KC, Aanstad P, Singla V, Norman AR, Stainier DY, Reiter JF.; ''Vertebrate Smoothened functions at the primary cilium.''; PubMed Europe PMC Scholia
  14. Stone DM, Murone M, Luoh S, Ye W, Armanini MP, Gurney A, Phillips H, Brush J, Goddard A, de Sauvage FJ, Rosenthal A.; ''Characterization of the human suppressor of fused, a negative regulator of the zinc-finger transcription factor Gli.''; PubMed Europe PMC Scholia
  15. Szczepny A, Wagstaff KM, Dias M, Gajewska K, Wang C, Davies RG, Kaur G, Ly-Huynh J, Loveland KL, Jans DA.; ''Overlapping binding sites for importin β1 and suppressor of fused (SuFu) on glioma-associated oncogene homologue 1 (Gli1) regulate its nuclear localization.''; PubMed Europe PMC Scholia
  16. Rohatgi R, Scott MP.; ''Patching the gaps in Hedgehog signalling.''; PubMed Europe PMC Scholia
  17. 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
  18. Ingham PW, Nakano Y, Seger C.; ''Mechanisms and functions of Hedgehog signalling across the metazoa.''; PubMed Europe PMC Scholia
  19. Patterson VL, Damrau C, Paudyal A, Reeve B, Grimes DT, Stewart ME, Williams DJ, Siggers P, Greenfield A, Murdoch JN.; ''Mouse hitchhiker mutants have spina bifida, dorso-ventral patterning defects and polydactyly: identification of Tulp3 as a novel negative regulator of the Sonic hedgehog pathway.''; PubMed Europe PMC Scholia
  20. Briscoe J, Thérond PP.; ''The mechanisms of Hedgehog signalling and its roles in development and disease.''; PubMed Europe PMC Scholia
  21. Agren M, Kogerman P, Kleman MI, Wessling M, Toftgård R.; ''Expression of the PTCH1 tumor suppressor gene is regulated by alternative promoters and a single functional Gli-binding site.''; PubMed Europe PMC Scholia
  22. Sassone-Corsi P.; ''The cyclic AMP pathway.''; PubMed Europe PMC Scholia
  23. Tuson M, He M, Anderson KV.; ''Protein kinase A acts at the basal body of the primary cilium to prevent Gli2 activation and ventralization of the mouse neural tube.''; PubMed Europe PMC Scholia
  24. Dai P, Shinagawa T, Nomura T, Harada J, Kaul SC, Wadhwa R, Khan MM, Akimaru H, Sasaki H, Colmenares C, Ishii S.; ''Ski is involved in transcriptional regulation by the repressor and full-length forms of Gli3.''; PubMed Europe PMC Scholia
  25. Weatherbee SD, Niswander LA, Anderson KV.; ''A mouse model for Meckel syndrome reveals Mks1 is required for ciliogenesis and Hedgehog signaling.''; PubMed Europe PMC Scholia
  26. Chen Y, Sasai N, Ma G, Yue T, Jia J, Briscoe J, Jiang J.; ''Sonic Hedgehog dependent phosphorylation by CK1α and GRK2 is required for ciliary accumulation and activation of smoothened.''; PubMed Europe PMC Scholia
  27. Pan Y, Wang B.; ''A novel protein-processing domain in Gli2 and Gli3 differentially blocks complete protein degradation by the proteasome.''; PubMed Europe PMC Scholia
  28. Wang B, Fallon JF, Beachy PA.; ''Hedgehog-regulated processing of Gli3 produces an anterior/posterior repressor gradient in the developing vertebrate limb.''; PubMed Europe PMC Scholia
  29. Wang Y, Zhou Z, Walsh CT, McMahon AP.; ''Selective translocation of intracellular Smoothened to the primary cilium in response to Hedgehog pathway modulation.''; PubMed Europe PMC Scholia
  30. Norman RX, Ko HW, Huang V, Eun CM, Abler LL, Zhang Z, Sun X, Eggenschwiler JT.; ''Tubby-like protein 3 (TULP3) regulates patterning in the mouse embryo through inhibition of Hedgehog signaling.''; PubMed Europe PMC Scholia
  31. Khaliullina H, Panáková D, Eugster C, Riedel F, Carvalho M, Eaton S.; ''Patched regulates Smoothened trafficking using lipoprotein-derived lipids.''; PubMed Europe PMC Scholia
  32. Pearse RV, Collier LS, Scott MP, Tabin CJ.; ''Vertebrate homologs of Drosophila suppressor of fused interact with the gli family of transcriptional regulators.''; PubMed Europe PMC Scholia
  33. Yue S, Tang LY, Tang Y, Tang Y, Shen QH, Ding J, Chen Y, Zhang Z, Yu TT, Zhang YE, Cheng SY.; ''Requirement of Smurf-mediated endocytosis of Patched1 in sonic hedgehog signal reception.''; PubMed Europe PMC Scholia
  34. Di Marcotullio L, Greco A, Mazzà D, Canettieri G, Pietrosanti L, Infante P, Coni S, Moretti M, De Smaele E, Ferretti E, Screpanti I, Gulino A.; ''Numb activates the E3 ligase Itch to control Gli1 function through a novel degradation signal.''; PubMed Europe PMC Scholia
  35. Hu MC, Mo R, Bhella S, Wilson CW, Chuang PT, Hui CC, Rosenblum ND.; ''GLI3-dependent transcriptional repression of Gli1, Gli2 and kidney patterning genes disrupts renal morphogenesis.''; PubMed Europe PMC Scholia
  36. Kise Y, Morinaka A, Teglund S, Miki H.; ''Sufu recruits GSK3beta for efficient processing of Gli3.''; PubMed Europe PMC Scholia
  37. Humke EW, Dorn KV, Milenkovic L, Scott MP, Rohatgi R.; ''The output of Hedgehog signaling is controlled by the dynamic association between Suppressor of Fused and the Gli proteins.''; PubMed Europe PMC Scholia
  38. Qin J, Lin Y, Norman RX, Ko HW, Eggenschwiler JT.; ''Intraflagellar transport protein 122 antagonizes Sonic Hedgehog signaling and controls ciliary localization of pathway components.''; PubMed Europe PMC Scholia
  39. Cortellino S, Wang C, Wang B, Bassi MR, Caretti E, Champeval D, Calmont A, Jarnik M, Burch J, Zaret KS, Larue L, Bellacosa A.; ''Defective ciliogenesis, embryonic lethality and severe impairment of the Sonic Hedgehog pathway caused by inactivation of the mouse complex A intraflagellar transport gene Ift122/Wdr10, partially overlapping with the DNA repair gene Med1/Mbd4.''; PubMed Europe PMC Scholia
  40. Cheung HO, Zhang X, Ribeiro A, Mo R, Makino S, Puviindran V, Law KK, Briscoe J, Hui CC.; ''The kinesin protein Kif7 is a critical regulator of Gli transcription factors in mammalian hedgehog signaling.''; PubMed Europe PMC Scholia
  41. Kinzler KW, Vogelstein B.; ''The GLI gene encodes a nuclear protein which binds specific sequences in the human genome.''; PubMed Europe PMC Scholia
  42. Paces-Fessy M, Boucher D, Petit E, Paute-Briand S, Blanchet-Tournier MF.; ''The negative regulator of Gli, Suppressor of fused (Sufu), interacts with SAP18, Galectin3 and other nuclear proteins.''; PubMed Europe PMC Scholia
  43. Dwyer JR, Sever N, Carlson M, Nelson SF, Beachy PA, Parhami F.; ''Oxysterols are novel activators of the hedgehog signaling pathway in pluripotent mesenchymal cells.''; PubMed Europe PMC Scholia
  44. Goetz SC, Anderson KV.; ''The primary cilium: a signalling centre during vertebrate development.''; PubMed Europe PMC Scholia
  45. Pan Y, Wang C, Wang B.; ''Phosphorylation of Gli2 by protein kinase A is required for Gli2 processing and degradation and the Sonic Hedgehog-regulated mouse development.''; PubMed Europe PMC Scholia
  46. Hwang SH, Mukhopadhyay S.; ''G-protein-coupled receptors and localized signaling in the primary cilium during ventral neural tube patterning.''; PubMed Europe PMC Scholia
  47. Mas C, Ruiz i Altaba A.; ''Small molecule modulation of HH-GLI signaling: current leads, trials and tribulations.''; PubMed Europe PMC Scholia
  48. Lee EY, Ji H, Ouyang Z, Zhou B, Ma W, Vokes SA, McMahon AP, Wong WH, Scott MP.; ''Hedgehog pathway-regulated gene networks in cerebellum development and tumorigenesis.''; PubMed Europe PMC Scholia
  49. Chen Y, Li S, Tong C, Zhao Y, Wang B, Liu Y, Jia J, Jiang J.; ''G protein-coupled receptor kinase 2 promotes high-level Hedgehog signaling by regulating the active state of Smo through kinase-dependent and kinase-independent mechanisms in Drosophila.''; PubMed Europe PMC Scholia
  50. Liem KF, He M, Ocbina PJ, Anderson KV.; ''Mouse Kif7/Costal2 is a cilia-associated protein that regulates Sonic hedgehog signaling.''; PubMed Europe PMC Scholia
  51. Svärd J, Heby-Henricson K, Persson-Lek M, Rozell B, Lauth M, Bergström A, Ericson J, Toftgård R, Teglund S.; ''Genetic elimination of Suppressor of fused reveals an essential repressor function in the mammalian Hedgehog signaling pathway.''; PubMed Europe PMC Scholia
  52. Huangfu D, Anderson KV.; ''Cilia and Hedgehog responsiveness in the mouse.''; PubMed Europe PMC Scholia
  53. Schrader EK, Harstad KG, Holmgren RA, Matouschek A.; ''A three-part signal governs differential processing of Gli1 and Gli3 proteins by the proteasome.''; PubMed Europe PMC Scholia
  54. Cheng SY, Bishop JM.; ''Suppressor of Fused represses Gli-mediated transcription by recruiting the SAP18-mSin3 corepressor complex.''; PubMed Europe PMC Scholia
  55. Tran PV, Haycraft CJ, Besschetnova TY, Turbe-Doan A, Stottmann RW, Herron BJ, Chesebro AL, Qiu H, Scherz PJ, Shah JV, Yoder BK, Beier DR.; ''THM1 negatively modulates mouse sonic hedgehog signal transduction and affects retrograde intraflagellar transport in cilia.''; PubMed Europe PMC Scholia
  56. Kim J, Kato M, Beachy PA.; ''Gli2 trafficking links Hedgehog-dependent activation of Smoothened in the primary cilium to transcriptional activation in the nucleus.''; PubMed Europe PMC Scholia
  57. Vierkotten J, Dildrop R, Peters T, Wang B, Rüther U.; ''Ftm is a novel basal body protein of cilia involved in Shh signalling.''; PubMed Europe PMC Scholia
  58. Huangfu D, Anderson KV.; ''Signaling from Smo to Ci/Gli: conservation and divergence of Hedgehog pathways from Drosophila to vertebrates.''; PubMed Europe PMC Scholia
  59. Tukachinsky H, Lopez LV, Salic A.; ''A mechanism for vertebrate Hedgehog signaling: recruitment to cilia and dissociation of SuFu-Gli protein complexes.''; PubMed Europe PMC Scholia
  60. Mukhopadhyay S, Wen X, Ratti N, Loktev A, Rangell L, Scales SJ, Jackson PK.; ''The ciliary G-protein-coupled receptor Gpr161 negatively regulates the Sonic hedgehog pathway via cAMP signaling.''; PubMed Europe PMC Scholia
  61. Haycraft CJ, Banizs B, Aydin-Son Y, Zhang Q, Michaud EJ, Yoder BK.; ''Gli2 and Gli3 localize to cilia and require the intraflagellar transport protein polaris for processing and function.''; PubMed Europe PMC Scholia
  62. Liu A, Wang B, Niswander LA.; ''Mouse intraflagellar transport proteins regulate both the activator and repressor functions of Gli transcription factors.''; PubMed Europe PMC Scholia
  63. Varjosalo M, Björklund M, Cheng F, Syvänen H, Kivioja T, Kilpinen S, Sun Z, Kallioniemi O, Stunnenberg HG, He WW, Ojala P, Taipale J.; ''Application of active and kinase-deficient kinome collection for identification of kinases regulating hedgehog signaling.''; PubMed Europe PMC Scholia
  64. Endoh-Yamagami S, Evangelista M, Wilson D, Wen X, Theunissen JW, Phamluong K, Davis M, Scales SJ, Solloway MJ, de Sauvage FJ, Peterson AS.; ''The mammalian Cos2 homolog Kif7 plays an essential role in modulating Hh signal transduction during development.''; PubMed Europe PMC Scholia
  65. Chen MH, Wilson CW, Li YJ, Law KK, Lu CS, Gacayan R, Zhang X, Hui CC, Chuang PT.; ''Cilium-independent regulation of Gli protein function by Sufu in Hedgehog signaling is evolutionarily conserved.''; PubMed Europe PMC Scholia
  66. Ayers KL, Thérond PP.; ''Evaluating Smoothened as a G-protein-coupled receptor for Hedgehog signalling.''; PubMed Europe PMC Scholia
  67. Cooper AF, Yu KP, Brueckner M, Brailey LL, Johnson L, McGrath JM, Bale AE.; ''Cardiac and CNS defects in a mouse with targeted disruption of suppressor of fused.''; PubMed Europe PMC Scholia
  68. Ferrante MI, Zullo A, Barra A, Bimonte S, Messaddeq N, Studer M, Dollé P, Franco B.; ''Oral-facial-digital type I protein is required for primary cilia formation and left-right axis specification.''; PubMed Europe PMC Scholia
  69. Corcoran RB, Scott MP.; ''Oxysterols stimulate Sonic hedgehog signal transduction and proliferation of medulloblastoma cells.''; PubMed Europe PMC Scholia
  70. Tempé D, Casas M, Karaz S, Blanchet-Tournier MF, Concordet JP.; ''Multisite protein kinase A and glycogen synthase kinase 3beta phosphorylation leads to Gli3 ubiquitination by SCFbetaTrCP.''; PubMed Europe PMC Scholia
  71. Pastorino L, Ghiorzo P, Nasti S, Battistuzzi L, Cusano R, Marzocchi C, Garrè ML, Clementi M, Scarrà GB.; ''Identification of a SUFU germline mutation in a family with Gorlin syndrome.''; PubMed Europe PMC Scholia
  72. Wang B, Li Y.; ''Evidence for the direct involvement of {beta}TrCP in Gli3 protein processing.''; PubMed Europe PMC Scholia
  73. Santagata S, Boggon TJ, Baird CL, Gomez CA, Zhao J, Shan WS, Myszka DG, Shapiro L.; ''G-protein signaling through tubby proteins.''; PubMed Europe PMC Scholia
  74. Huntzicker EG, Estay IS, Zhen H, Lokteva LA, Jackson PK, Oro AE.; ''Dual degradation signals control Gli protein stability and tumor formation.''; PubMed Europe PMC Scholia
  75. Zhao Y, Tong C, Jiang J.; ''Hedgehog regulates smoothened activity by inducing a conformational switch.''; PubMed Europe PMC Scholia
  76. Gray RS, Abitua PB, Wlodarczyk BJ, Szabo-Rogers HL, Blanchard O, Lee I, Weiss GS, Liu KJ, Marcotte EM, Wallingford JB, Finnell RH.; ''The planar cell polarity effector Fuz is essential for targeted membrane trafficking, ciliogenesis and mouse embryonic development.''; PubMed Europe PMC Scholia
  77. Taipale J, Cooper MK, Maiti T, Beachy PA.; ''Patched acts catalytically to suppress the activity of Smoothened.''; PubMed Europe PMC Scholia
  78. Di Marcotullio L, Ferretti E, Greco A, De Smaele E, Po A, Sico MA, Alimandi M, Giannini G, Maroder M, Screpanti I, Gulino A.; ''Numb is a suppressor of Hedgehog signalling and targets Gli1 for Itch-dependent ubiquitination.''; PubMed Europe PMC Scholia
  79. Voges D, Zwickl P, Baumeister W.; ''The 26S proteasome: a molecular machine designed for controlled proteolysis.''; PubMed Europe PMC Scholia
  80. Pal K, Mukhopadhyay S.; ''Primary cilium and sonic hedgehog signaling during neural tube patterning: role of GPCRs and second messengers.''; PubMed Europe PMC Scholia
  81. Vokes SA, Ji H, McCuine S, Tenzen T, Giles S, Zhong S, Longabaugh WJ, Davidson EH, Wong WH, McMahon AP.; ''Genomic characterization of Gli-activator targets in sonic hedgehog-mediated neural patterning.''; PubMed Europe PMC Scholia
  82. Milenkovic L, Scott MP, Rohatgi R.; ''Lateral transport of Smoothened from the plasma membrane to the membrane of the cilium.''; PubMed Europe PMC Scholia
  83. Hatayama M, Aruga J.; ''Gli protein nuclear localization signal.''; PubMed Europe PMC Scholia
  84. Wilson CW, Chen MH, Chuang PT.; ''Smoothened adopts multiple active and inactive conformations capable of trafficking to the primary cilium.''; PubMed Europe PMC Scholia
  85. Dunaeva M, Michelson P, Kogerman P, Toftgard R.; ''Characterization of the physical interaction of Gli proteins with SUFU proteins.''; PubMed Europe PMC Scholia
  86. Monnier V, Dussillol F, Alves G, Lamour-Isnard C, Plessis A.; ''Suppressor of fused links fused and Cubitus interruptus on the hedgehog signalling pathway.''; PubMed Europe PMC Scholia
  87. Mukhopadhyay S, Wen X, Chih B, Nelson CD, Lane WS, Scales SJ, Jackson PK.; ''TULP3 bridges the IFT-A complex and membrane phosphoinositides to promote trafficking of G protein-coupled receptors into primary cilia.''; PubMed Europe PMC Scholia
  88. Huangfu D, Liu A, Rakeman AS, Murcia NS, Niswander L, Anderson KV.; ''Hedgehog signalling in the mouse requires intraflagellar transport proteins.''; PubMed Europe PMC Scholia
  89. May SR, Ashique AM, Karlen M, Wang B, Shen Y, Zarbalis K, Reiter J, Ericson J, Peterson AS.; ''Loss of the retrograde motor for IFT disrupts localization of Smo to cilia and prevents the expression of both activator and repressor functions of Gli.''; PubMed Europe PMC Scholia
  90. Taylor MD, Liu L, Raffel C, Hui CC, Mainprize TG, Zhang X, Agatep R, Chiappa S, Gao L, Lowrance A, Hao A, Goldstein AM, Stavrou T, Scherer SW, Dura WT, Wainwright B, Squire JA, Rutka JT, Hogg D.; ''Mutations in SUFU predispose to medulloblastoma.''; PubMed Europe PMC Scholia
  91. Nachtergaele S, Mydock LK, Krishnan K, Rammohan J, Schlesinger PH, Covey DF, Rohatgi R.; ''Oxysterols are allosteric activators of the oncoprotein Smoothened.''; PubMed Europe PMC Scholia
  92. Wen X, Lai CK, Evangelista M, Hongo JA, de Sauvage FJ, Scales SJ.; ''Kinetics of hedgehog-dependent full-length Gli3 accumulation in primary cilia and subsequent degradation.''; PubMed Europe PMC Scholia
  93. Jia J, Kolterud A, Zeng H, Hoover A, Teglund S, Toftgård R, Liu A.; ''Suppressor of Fused inhibits mammalian Hedgehog signaling in the absence of cilia.''; PubMed Europe PMC Scholia
  94. Pan Y, Bai CB, Joyner AL, Wang B.; ''Sonic hedgehog signaling regulates Gli2 transcriptional activity by suppressing its processing and degradation.''; PubMed Europe PMC Scholia

History

View all...
CompareRevisionActionTimeUserComment
114910view16:42, 25 January 2021ReactomeTeamReactome version 75
113355view11:42, 2 November 2020ReactomeTeamReactome version 74
112564view15:53, 9 October 2020ReactomeTeamReactome version 73
101477view11:34, 1 November 2018ReactomeTeamreactome version 66
101015view21:14, 31 October 2018ReactomeTeamreactome version 65
100551view19:48, 31 October 2018ReactomeTeamreactome version 64
100099view16:32, 31 October 2018ReactomeTeamreactome version 63
99649view15:04, 31 October 2018ReactomeTeamreactome version 62 (2nd attempt)
99251view12:45, 31 October 2018ReactomeTeamreactome version 62
93840view13:40, 16 August 2017ReactomeTeamreactome version 61
93396view11:22, 9 August 2017ReactomeTeamreactome version 61
87141view18:52, 18 July 2016MkutmonOntology Term : 'Hedgehog signaling pathway' added !
86481view09:19, 11 July 2016ReactomeTeamreactome version 56
83587view07:34, 26 November 2015EgonwAdded missing ENSG parts of the Ensembl IDs.
83062view09:50, 18 November 2015ReactomeTeamVersion54
81377view12:54, 21 August 2015ReactomeTeamNew pathway

External references

DataNodes

View all...
NameTypeDatabase referenceComment
26S proteasomeComplexR-HSA-68819 (Reactome)
4xub-p13S-Gli3:SUFUComplexR-HSA-5610526 (Reactome)
ADCY1 ProteinQ08828 (Uniprot-TrEMBL)
ADCY10 ProteinQ96PN6 (Uniprot-TrEMBL)
ADCY2 ProteinQ08462 (Uniprot-TrEMBL)
ADCY3 ProteinO60266 (Uniprot-TrEMBL)
ADCY4 ProteinQ8NFM4 (Uniprot-TrEMBL)
ADCY5 ProteinO95622 (Uniprot-TrEMBL)
ADCY6 ProteinO43306 (Uniprot-TrEMBL)
ADCY7 ProteinP51828 (Uniprot-TrEMBL)
ADCY8 ProteinP40145 (Uniprot-TrEMBL)
ADCY9 ProteinO60503 (Uniprot-TrEMBL)
ADPMetaboliteCHEBI:456216 (ChEBI)
ATPMetaboliteCHEBI:30616 (ChEBI)
BTRC ProteinQ9Y297 (Uniprot-TrEMBL)
BTRC:CUL1:RBX1:SKP1ComplexR-HSA-206748 (Reactome)
CSNK1A1ProteinP48729 (Uniprot-TrEMBL)
CUL1 ProteinQ13616 (Uniprot-TrEMBL)
DYNC2H1 ProteinQ8NCM8 (Uniprot-TrEMBL)
FUZ ProteinQ9BT04 (Uniprot-TrEMBL)
G alpha (s):GTPComplexR-HSA-164358 (Reactome)
GLI1 ProteinP08151 (Uniprot-TrEMBL)
GLI1 gene ProteinENSG00000111087 (Ensembl)
GLI1 gene:GLI3RComplexR-HSA-5617398 (Reactome)
GLI1 geneGeneProductENSG00000111087 (Ensembl)
GLI1,2,3ComplexR-HSA-5610616 (Reactome)
GLI1:SUFUComplexR-HSA-5610531 (Reactome)
GLI1ProteinP08151 (Uniprot-TrEMBL)
GLI2 ProteinP10070 (Uniprot-TrEMBL)
GLI2 gene ProteinENSG00000074047 (Ensembl)
GLI2 gene:GLI3RComplexR-HSA-5617399 (Reactome)
GLI2 geneGeneProductENSG00000074047 (Ensembl)
GLI2:SUFUComplexR-HSA-5610536 (Reactome)
GLI2ProteinP10070 (Uniprot-TrEMBL)
GLI3 ProteinP10071 (Uniprot-TrEMBL)
GLI3:SUFUComplexR-HSA-5610542 (Reactome)
GLI3R ProteinP10071 (Uniprot-TrEMBL)
GLI3RProteinP10071 (Uniprot-TrEMBL)
GLI:SUFUComplexR-HSA-5621780 (Reactome)
GLI:SUFUComplexR-HSA-5621783 (Reactome)
GLI:SUFUComplexR-HSA-5621787 (Reactome)
GNAS1 ProteinQ5JWF2 (Uniprot-TrEMBL)
GNAS2 ProteinP63092 (Uniprot-TrEMBL)
GPR161 ProteinQ8N6U8 (Uniprot-TrEMBL)
GPR161:IFT-A:TULP3ComplexR-HSA-5610579 (Reactome)
GPR161ProteinQ8N6U8 (Uniprot-TrEMBL)
GSK3BProteinP49841 (Uniprot-TrEMBL)
GTP MetaboliteCHEBI:15996 (ChEBI)
IFT-A complex:TULP3ComplexR-HSA-5610556 (Reactome)
IFT-A complexComplexR-HSA-5610555 (Reactome)
IFT122 ProteinQ9HBG6 (Uniprot-TrEMBL)
IFT140 ProteinQ96RY7 (Uniprot-TrEMBL)
IFT172 ProteinQ9UG01 (Uniprot-TrEMBL)
IFT52 ProteinQ9Y366 (Uniprot-TrEMBL)
IFT57 ProteinQ9NWB7 (Uniprot-TrEMBL)
IFT88 ProteinQ13099 (Uniprot-TrEMBL)
INTU ProteinQ9ULD6 (Uniprot-TrEMBL)
ITCH ProteinQ96J02 (Uniprot-TrEMBL)
ITCHProteinQ96J02 (Uniprot-TrEMBL)
KIF3A ProteinQ9Y496 (Uniprot-TrEMBL)
KIF7 ProteinQ2M1P5 (Uniprot-TrEMBL)
KIF7:microtubuleComplexR-HSA-5610559 (Reactome)
KIF7ProteinQ2M1P5 (Uniprot-TrEMBL)
MKS1 ProteinQ9NXB0 (Uniprot-TrEMBL)
Mg2+ MetaboliteCHEBI:18420 (ChEBI)
NUMB ProteinP49757 (Uniprot-TrEMBL)
NUMB:ITCH:ub-pS-GLI1:SUFUComplexR-HSA-5610565 (Reactome)
NUMB:ITCHComplexR-HSA-5610562 (Reactome)
NUMBProteinP49757 (Uniprot-TrEMBL)
OFD1 ProteinO75665 (Uniprot-TrEMBL)
PCP regulators of HhComplexR-HSA-5610624 (Reactome)
PKA catalytic subunitComplexR-HSA-5610569 (Reactome)
PKA regulatory subunits:cAMPComplexR-HSA-5610566 (Reactome)
PKA tetramerComplexR-HSA-5610571 (Reactome)
PPiMetaboliteCHEBI:29888 (ChEBI)
PRKACA ProteinP17612 (Uniprot-TrEMBL)
PRKACB ProteinP22694 (Uniprot-TrEMBL)
PRKACG ProteinP22612 (Uniprot-TrEMBL)
PRKAR1A ProteinP10644 (Uniprot-TrEMBL)
PRKAR1B ProteinP31321 (Uniprot-TrEMBL)
PRKAR2A ProteinP13861 (Uniprot-TrEMBL)
PRKAR2B ProteinP31323 (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)
PTCH1 gene ProteinENSG00000185920 (Ensembl)
PTCH1 gene:GLI3RComplexR-HSA-5612507 (Reactome)
PTCH1 geneGeneProductENSG00000185920 (Ensembl)
PTCH1ProteinQ13635 (Uniprot-TrEMBL)
RBX1 ProteinP62877 (Uniprot-TrEMBL)
RPGRIP1L ProteinQ68CZ1 (Uniprot-TrEMBL)
RPS27A(1-76) ProteinP62979 (Uniprot-TrEMBL)
SHFM1 ProteinP60896 (Uniprot-TrEMBL)
SKP1 ProteinP63208 (Uniprot-TrEMBL)
SMO ProteinQ99835 (Uniprot-TrEMBL)
SMO dimerComplexR-HSA-5610573 (Reactome)
SMO dimerComplexR-HSA-5610574 (Reactome)
SUFU ProteinQ9UMX1 (Uniprot-TrEMBL)
SUFUProteinQ9UMX1 (Uniprot-TrEMBL)
TTC21B ProteinQ7Z4L5 (Uniprot-TrEMBL)
TULP3 ProteinO75386 (Uniprot-TrEMBL)
TULP3ProteinO75386 (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)
WDR19 ProteinQ8NEZ3 (Uniprot-TrEMBL)
WDR35 ProteinQ9P2L0 (Uniprot-TrEMBL)
adenylate cyclases:Mg2+ComplexR-HSA-5610577 (Reactome)
anterograde IFT regulators of HhComplexR-HSA-5610625 (Reactome)
cAMP MetaboliteCHEBI:17489 (ChEBI)
cAMPMetaboliteCHEBI:17489 (ChEBI)
ciliary basal body regulators of HhComplexR-HSA-5610628 (Reactome)
microtubule R-HSA-5610520 (Reactome)
microtubuleR-HSA-5610520 (Reactome)
p10S-GLI3 ProteinP10071 (Uniprot-TrEMBL)
p10S-GLI3:SUFUComplexR-HSA-5610581 (Reactome)
p11S-GLI2 ProteinP10070 (Uniprot-TrEMBL)
p11S-GLI2:SUFUComplexR-HSA-5610584 (Reactome)
p13S-GLI3 ProteinP10071 (Uniprot-TrEMBL)
p13S-GLI3:SUFUComplexR-HSA-5610587 (Reactome)
p4S-GLI2 ProteinP10070 (Uniprot-TrEMBL)
p4S-GLI2:SUFUComplexR-HSA-5610589 (Reactome)
p6S-GLI3 ProteinP10071 (Uniprot-TrEMBL)
p6S-GLI3:SUFUComplexR-HSA-5610595 (Reactome)
p8S-GLI2 ProteinP10070 (Uniprot-TrEMBL)
p8S-GLI2:SUFUComplexR-HSA-5610598 (Reactome)
pS-GLI1 ProteinP08151 (Uniprot-TrEMBL)
pS-GLI1:SUFUComplexR-HSA-5610605 (Reactome)
retrograde IFT regulators of HhComplexR-HSA-5610630 (Reactome)
ub-p11S-GLI2 ProteinP10070 (Uniprot-TrEMBL)
ub-p11S-GLI2:SUFUComplexR-HSA-5610608 (Reactome)
ub-p13S-GLI3 ProteinP10071 (Uniprot-TrEMBL)
ub-pS-GLI1 ProteinP08151 (Uniprot-TrEMBL)
ub-pS-GLI1:SUFUComplexR-HSA-5610612 (Reactome)

Annotated Interactions

View all...
SourceTargetTypeDatabase referenceComment
26S proteasomemim-catalysisR-HSA-5610754 (Reactome)
26S proteasomemim-catalysisR-HSA-5610757 (Reactome)
26S proteasomemim-catalysisR-HSA-5610758 (Reactome)
26S proteasomemim-catalysisR-HSA-5610760 (Reactome)
4xub-p13S-Gli3:SUFUArrowR-HSA-5610746 (Reactome)
4xub-p13S-Gli3:SUFUR-HSA-5610754 (Reactome)
ADPArrowR-HSA-5610717 (Reactome)
ADPArrowR-HSA-5610718 (Reactome)
ADPArrowR-HSA-5610720 (Reactome)
ADPArrowR-HSA-5610722 (Reactome)
ADPArrowR-HSA-5610730 (Reactome)
ADPArrowR-HSA-5610732 (Reactome)
ADPArrowR-HSA-5610741 (Reactome)
ATPR-HSA-5610717 (Reactome)
ATPR-HSA-5610718 (Reactome)
ATPR-HSA-5610720 (Reactome)
ATPR-HSA-5610722 (Reactome)
ATPR-HSA-5610727 (Reactome)
ATPR-HSA-5610730 (Reactome)
ATPR-HSA-5610732 (Reactome)
ATPR-HSA-5610741 (Reactome)
BTRC:CUL1:RBX1:SKP1mim-catalysisR-HSA-5610742 (Reactome)
BTRC:CUL1:RBX1:SKP1mim-catalysisR-HSA-5610745 (Reactome)
BTRC:CUL1:RBX1:SKP1mim-catalysisR-HSA-5610746 (Reactome)
CSNK1A1mim-catalysisR-HSA-5610718 (Reactome)
CSNK1A1mim-catalysisR-HSA-5610722 (Reactome)
G alpha (s):GTPArrowR-HSA-5610727 (Reactome)
GLI1 gene:GLI3RArrowR-HSA-5617408 (Reactome)
GLI1 gene:GLI3RTBarR-HSA-5617412 (Reactome)
GLI1 geneR-HSA-5617408 (Reactome)
GLI1 geneR-HSA-5617412 (Reactome)
GLI1,2,3R-HSA-5610723 (Reactome)
GLI1:SUFUR-HSA-5610741 (Reactome)
GLI1ArrowR-HSA-5617412 (Reactome)
GLI2 gene:GLI3RArrowR-HSA-5617410 (Reactome)
GLI2 gene:GLI3RTBarR-HSA-5617413 (Reactome)
GLI2 geneR-HSA-5617410 (Reactome)
GLI2 geneR-HSA-5617413 (Reactome)
GLI2:SUFUR-HSA-5610717 (Reactome)
GLI2ArrowR-HSA-5617413 (Reactome)
GLI3:SUFUR-HSA-5610720 (Reactome)
GLI3RArrowR-HSA-5610752 (Reactome)
GLI3RArrowR-HSA-5610754 (Reactome)
GLI3RR-HSA-5610752 (Reactome)
GLI3RR-HSA-5612508 (Reactome)
GLI3RR-HSA-5617408 (Reactome)
GLI3RR-HSA-5617410 (Reactome)
GLI:SUFUArrowR-HSA-5610723 (Reactome)
GLI:SUFUArrowR-HSA-5610766 (Reactome)
GLI:SUFUArrowR-HSA-5610767 (Reactome)
GLI:SUFUR-HSA-5610766 (Reactome)
GLI:SUFUR-HSA-5610767 (Reactome)
GPR161:IFT-A:TULP3ArrowR-HSA-5610725 (Reactome)
GPR161:IFT-A:TULP3ArrowR-HSA-5610727 (Reactome)
GPR161R-HSA-5610725 (Reactome)
GSK3Bmim-catalysisR-HSA-5610730 (Reactome)
GSK3Bmim-catalysisR-HSA-5610732 (Reactome)
IFT-A complex:TULP3ArrowR-HSA-5610726 (Reactome)
IFT-A complex:TULP3R-HSA-5610725 (Reactome)
IFT-A complexR-HSA-5610726 (Reactome)
ITCHR-HSA-5610735 (Reactome)
KIF7:microtubuleArrowR-HSA-5610733 (Reactome)
KIF7:microtubuleArrowR-HSA-5610767 (Reactome)
KIF7R-HSA-5610733 (Reactome)
NUMB:ITCH:ub-pS-GLI1:SUFUArrowR-HSA-5610737 (Reactome)
NUMB:ITCH:ub-pS-GLI1:SUFUR-HSA-5610760 (Reactome)
NUMB:ITCHArrowR-HSA-5610735 (Reactome)
NUMB:ITCHArrowR-HSA-5610760 (Reactome)
NUMB:ITCHR-HSA-5610737 (Reactome)
NUMB:ITCHmim-catalysisR-HSA-5610737 (Reactome)
NUMBR-HSA-5610735 (Reactome)
PCP regulators of HhArrowR-HSA-5610766 (Reactome)
PCP regulators of HhArrowR-HSA-5610767 (Reactome)
PKA catalytic subunitArrowR-HSA-5610749 (Reactome)
PKA catalytic subunitmim-catalysisR-HSA-5610717 (Reactome)
PKA catalytic subunitmim-catalysisR-HSA-5610720 (Reactome)
PKA catalytic subunitmim-catalysisR-HSA-5610741 (Reactome)
PKA regulatory subunits:cAMPArrowR-HSA-5610749 (Reactome)
PKA tetramerR-HSA-5610749 (Reactome)
PPiArrowR-HSA-5610727 (Reactome)
PTCH1 gene:GLI3RArrowR-HSA-5612508 (Reactome)
PTCH1 gene:GLI3RTBarR-HSA-5612510 (Reactome)
PTCH1 geneR-HSA-5612508 (Reactome)
PTCH1 geneR-HSA-5612510 (Reactome)
PTCH1ArrowR-HSA-5612510 (Reactome)
PTCH1TBarR-HSA-5610763 (Reactome)
R-HSA-5610717 (Reactome) Despite sharing 44% amino acid identity with GLI3, only a small fraction of GLI2 appears to be processed to a repressor form in the absence of Hh signaling; the bulk of the protein is completely degraded in a phosphorylation- and proteasome-dependent manner (Pan et al, 2007; Pan et al, 2009; Pan and Wang, 2007). Degradation of GLI2 depends on phosphorylation of four consensus PKA sites in the C-terminal region. This phosphorylation primes GLI2 for subsequent phosphorylation by CK1 and GSK3, creating a binding site for betaTrCP and promoting its subsequent ubiquitination and degradation (Pan et al, 2006; Pan and Wang, 2007; Pan et al, 2009).
R-HSA-5610718 (Reactome) Phosphorylation by PKA primes GLI2 for subsequent phosphorylation at adjacent CK sites (Pan et al, 2006; Pan and Wang, 2007).
R-HSA-5610720 (Reactome) Phosphorylation of GLI3 by PKA on up to six sites in the C-terminal region primes the protein for subsequent phosphorylation by CK1 and GSK3 and is required for the ubiquitin-mediated processing by the proteasome to yield the truncated repressor form (Tempe et al, 2006; Pan et al, 2006; Pan and Wang, 2007; Wang and Li, 2006). Processing of GLI3 is regulated in part by movement through the primary cilia, and disruption of intraflagellar transport abrogates processing (Wen et al, 2010)
R-HSA-5610722 (Reactome) Phosphorylation by PKA primes GLI3 for subsequent phosphorylation by CK1 at four or more sites. These serial phosphorylations are required for the recruitment of beta-TrCP and subsequent ubiquitination and processing of GLI3 (Tempe et al, 2006; Wang and Li, 2006; Wen et al, 2010; Schrader et al, 2011)
R-HSA-5610723 (Reactome) Vertebrate SUFU plays a critical role in the negative regulation of Hh signaling in the absence of ligand. Disruption of SUFU causes constitutive activation of the pathway, and is associated with the development of medulloblastoma in humans (Cooper et al, 2005; Svard et al, 2006; Taylor et al, 2002; Pastorino et al, 2009). SUFU binds directly to all three GLI proteins (Pearse et al, 1999; Stone et al, 1999; Jia et al, 2009; Svard et al, 2006). Formation of a SUFU:GLI complex is required for the processing of GLI3 to the GLI3R repressor form, and the processing depends on transit through the primary cilia (Kise et al, 2009; Humke et al, 2010; Huangfu and Anderson, 2005). Despite this, primary cilia are not required for SUFU to inhibit GLI activity; SUFU may also serve in a cilia-independent manner to sequester the full-length protein in the cytoplasm in the absence of Hh signal (Chen et al, 2009; Humke et al, 2010; Jia et al, 2009; Tukachinsky et al, 2010). After processing, GLI3R dissociates from SUFU and its activity is SUFU-independent (Humke et al, 2010; Tukachinsky et al, 2010). Nuclear SUFU may also play a direct role as a transcriptional co-repressor through interaction with the N-terminal DNA-binding domain of GLI proteins, though this remains to be fully elaborated (Monnier et al, 1998; Pearse et al, 1999; Cheng and Bishop, 2002; Paces-Fessy et al, 2004; Dunaeva et al, 2003; Szczepny et al, 2014).
R-HSA-5610725 (Reactome) TULP3 and the retrograde complex IFT-A are required to recruit GPR161 to the cilium in the absence of Hh ligand (Mukhopadhyay et al, 2010; Mukhopadhyay et al, 2013; reviewed in Mukhopadhyay and Rohtagi, 2014). TULP3 is a negative regulator of Hh signaling and siRNA depletion of TULP3 reduces the ciliary accumulation of GPR161 (Norman et al, 2009; Patterson et al, 2009; Mukhopadhyay et al, 2010; Mukhopadhyay et al, 2013).
R-HSA-5610726 (Reactome) TULP3 is recruited to the primary cilium through a direct interaction with the retrograde transport IFT-A complex and with membrane phosphoinositides (Santagata et al, 2001; Mukhopadhyay et al, 2010; Qin et al, 2011; reviewed in Mukhopadhyay and Rohatgi, 2014). TULP3 facilitates GLI processing by recruiting the GPCR GPR161, which in turn activates PKA by increasing cAMP levels in a G alpha s-dependent manner (Mukhopadhyay et al, 2013, reviewed in Hwang and Mukhopadhyay, 2014; Pal and Mukhopadhyay, 2014).
R-HSA-5610727 (Reactome) cAMP is generated by the action of adenylate cyclases (reviewed in Sassone-Corsi, 2012). GPR161 is an orphan GPCR that has recently been identified as a negative regulator of Hh signaling that acts by increasing cellular cAMP levels in the absence of ligand. Overexpression of GPR161 increases cellular cAMP levels in a manner that depends on the G alpha s subunit, and depletion of GPR161 results in aberrant Hh signaling and a decrease in the ratio of processed GLI3R (Mukhopadhyay et al, 2013). These data suggest that GPR161 negatively regulates GLI processing in the absence of Hh signal by modulating PKA activity through cAMP levels (Mukhopadhyay et al, 2013; reviewed in Mukhopadhyay and Rohatgi, 2014).
R-HSA-5610730 (Reactome) Like GLI3, GLI2 has putative GSK3 sites that contribute to the proteasome-dependent degradation of the protein in the absence of Hh signal. Deletion of the GSK3 phosphorylation sites abrogates the interaction with beta-TrCP, stabilizes GLI2 protein and increases the expression of a GLI-dependent reporter, consistent with a role for GSK3 in promoting GLI2 degradation (Pan et al, 2006).
R-HSA-5610732 (Reactome) GSK3-mediated phosphorylation of GLI3 is primed by earlier phosphorylations by PKA and CK1 and is required for the subsequent recruitment of beta-TrCP (Tempe et al, 2006; Wang and Li, 2006).
R-HSA-5610733 (Reactome) KIF7, the human ortholog of Drosophila COS2, is a kinesin-4 motor protein that binds directly to the plus ends of axonemal microtubules and inhibits their growth in an ATP-dependent manner (He et al, 2014). KIF7 is required for the processing and activity of GLI in the absence of Hh signal, and KIF7 function depends on the primary cilium (Liem et al, 2009; Cheung et al, 2009; Endoh-Yamagami et al, 2009). KIF7 has been shown to bind to GLI3 and to SUFU and may act in part by promoting the transit of the GLI:SUFU complex through the primary cilium, which is required for GLI processing (Endoh-Yamagami et al, 2009; Maurya et al, 2013). How KIF7 itself is localized to the cilia tip is unknown, although localization depends on the KIF7 motor domain (Liem et al, 2009; He et al, 2014). KIF7 localization is further enhanced at the primary cilia tip in response to Hh signaling, as is also the case for GLI2, GLI3 and SUFU (He et al, 2014; Varjosalo et al, 2008; Haycraft et al, 2005; Wen et al, 2010; Qin et al, 2011; Tukachinsky et al, 2010).
R-HSA-5610735 (Reactome) NUMB is a negative regulator of Hh signaling that acts by promoting the ITCH-dependent ubiquitination of GLI1. ITCH is an E3 ligase that is kept in an inactive conformation by an intramolecular interaction between the HECT domain and a WW motif. Binding of the adaptor protein NUMB to the WW region of ITCH displaces the HECT domain and promotes the catalytic activity of the E3 ligase (di Marcotullio et al, 2006; 2011).
R-HSA-5610737 (Reactome) GLI1 is recruited to the NUMB:ITCH complex through a direct interaction with both proteins. Once recruited, GLI1 is ubiquitinated by ITCH and subsequently degraded by the proteasome. ITCH-mediated degradation of GLI1 does not depend on the Dc or Dn degrons required for interaction with beta-TrCP, but instead relies on a novel PPXYs/pSP degron of GLI1 (di Marcotullio et al, 2006, 2011; Huntzicker et al, 2006). How these two apparently parallel systems of GLI1 ubiquitination and degradation are coordinated is not yet clear.
R-HSA-5610741 (Reactome) Although direct phosphorylation of GLI1 by PKA has not been demonstrated, deletion of the putative PKA sites abrogates the interaction of GLI1 with beta-TrCP and stabilizes GLI1 protein levels; similarly, treatment of GLI1-expressing cells with PKA inhibitors delays the kinetics of GLI1 degradation (Huntzicker et al, 2006). These data are consistent with a role for PKA-mediated phosphorylation in promoting the proteasome-dependent degradation of GLI1 in the absence of Hh signal, as is the case for GLI2 and GLI3 (Huntzicker et al, 2006; Tempe et al, 2006; Pan and Wang, 2007; Pan et al, 2009). Potential roles for CK2 and GSK3 in promoting the phosphorylation-dependent degradation of GLI1 have not been investigated.
R-HSA-5610742 (Reactome) GLI1 protein is degraded by the proteasome in the absence of Hh signal. GLI1 levels are stabilized by treatment of cells with the proteasome inhibitor MG312, and GLI1 and beta-TrCP1 co-precipitate when expressed in NIH 3T3 cells.
Two SCF(beta-TrCP)-dependent degradation sites, Dn and Dc, have been identified in human GLI1. Removal of these sites abrogates the interaction with beta-TrCP, reduces the beta-TrCP-dependent ubiquitination of GLI1 and stabilizes the GLI1 protein levels. As is the case for GLI2 and GLI3, ubiquitination of GLI1 depends on the its prior phosphorylation by PKA, as GLI1 degradation is sensitive to PKA inhibitors and removal of the putative PKA sites abrogates the interaction with beta-TrCP and delays the kinetics of degradation (Huntzicker et al, 2006).
R-HSA-5610745 (Reactome) GLI2 interacts directly with beta-TrCP and is polyubiquitinated in a phosphorylation-dependent manner. Binding and ubiquitination by beta-TrCP depends on 2 motifs located in the region of GLI2 phosphorylated by PKA, CK1 and GSK3 (Pan et al, 2006).
R-HSA-5610746 (Reactome) Hyperphosphorylated GLI3 binds directly with beta-TrCP though at least three independent domains and is polyubiquitinated at lysines 773, 778, 784 and 800 (Tempe et al, 2006). After ubiquitination, GLI3 is processed to the truncated repressor form by the proteasome (Tempe et al, 2006; Wang and Li, 2006)
R-HSA-5610749 (Reactome) cAMP is a known regulator of PKA activity and works by binding to the regulatory subunits and promoting dissociation of the tetramer, freeing the active catalytic subunits (reviewed in Sassone-Corsi, 2012). In the Hh pathway in the absence of ligand, cAMP levels increase in response to the recruitment of GPR161 to the ciliary base by TULP3 and the IFT-A retrograde complex (Mukhopadhyay et al, 2010; Mukhopadhyay et al, 2013). Activated PKA then initiates the phosphorylation cascade that regulates processing and/or degradation of the GLI proteins (reviewed in Briscoe and Therond, 2013; Mukhopadhyay and Rohatgi, 2014).
R-HSA-5610752 (Reactome) After processing by the proteasome, the truncated GLI3 translocates into the nucleus where it acts as the primary repressor for Hh-responsive genes (reviewed in Briscoe and Therond, 2013). Based on sequence comparisons with Ci and GLI1, GLI3 is predicted to have a bipartite NLS signal near the zinc finger domain, and import to the nucleus may be mediated by Importin alpha3, although the details remain to be worked out (reviewed in Hatayama and Aruga, 2012).
R-HSA-5610754 (Reactome) After phosphorylation and ubiquitination, GLI3 is processed by the proteasome to an 83-kDa repressor form that lacks the C-terminal activation domain (Wang et al, 2000; Tempe et al, 2006; Wang and Li, 2006). Partial processing appears to rely on at least three features of the GLI3 protein: the folded N-terminal zinc finger domain, an adjacent simple linker sequence, and the degron in the C-terminus that contains the phosphorylation and ubiquitination target residues (Pan and Wang, 2007; Schrader et al, 2011). The C-terminal end of the processed repressor form is not precisely defined.
R-HSA-5610757 (Reactome) After being ubiquitinated, the bulk of GLI2 is fully degraded by the proteasome; a small fraction of GLI2 may be converted to the repressor form after ubiquitination (Pan et al, 2006; Pan and Wang, 2007).
R-HSA-5610758 (Reactome) In the absence of Hh signal, GLI1 is degraded by the proteasome. Degradation depends on GLI1 ubiquitination by SCF(beta-TrCP) and by the E3 ligase ITCH (Huntzicker et al, 2006; di Marcotullio et al, 2006, 2011).
R-HSA-5610760 (Reactome) In the absence of Hh signal, GLI1 is degraded by the proteasome. Degradation depends on GLI1 ubiquitination by SCF(beta-TrCP) and by the E3 ligase ITCH (Huntzicker et al, 2006; di Marcotullio et al, 2006, 2011).
R-HSA-5610763 (Reactome) In the absence of Hh ligand, the Hh receptor PTCH inhibits signaling by negatively regulating the activity of SMO, a candidate member of the GPCR superfamily that transduces the Hh signal to downstream pathway components (reviewed in Ayers and Therond, 2010; Briscoe and Therond, 2013). Neither the mechanism by which SMO activates Hh signaling nor the manner in which PTCH represses this activty are fully elucidated, but these may involve regulation of putative SMO ligand(s) or changes in cellular localization, protein conformation and phosphorylation status, among other possibilities (reviewed in Briscoe and Therond, 2013; Ayers and Therond; 2010).
PTCH is a 7 transmembrane protein that is localized to the primary cilium in the absence of Hh ligand (Rohatgi et al, 2007). PTCH regulates SMO in a non-stoichiometric manner and there is little evidence that endogenous PTCH and SMO interact directly (Taipale et al, 2002; reviewed in Huangfu and Anderson, 2006). PTCH has a sterol sensing domain (SSD) and structural similarity to bacterial RND transporters. Mutation in conserved motifs in the RND domain abrogate the ability of PTCH to negatively regulate SMO activity (Taipale et al, 2002). The transmembrane heptahelical domain of SMO has been shown to bind to a number of natural and synthetic molecules, many of which are structurally related to sterols, and this binding can activate or repress SMO activity (Mas et al, 2010; Dwyer et al, 2007; Nachtergaele et al, 2012; Corcoran et al, 2006). Together, these data suggest a speculative model where PTCH regulates SMO activity by controlling the flux of sterol-related SMO agonists and/or antagonists, although this has not been fully substantiated (Khaliullina et al, 2009; reviewed in Rohatgi and Scott, 2007; Briscoe and Therond, 2013).
In the absence of Hh signal, SMO is largely found in intracellular vesicles, with a fraction localized to the plasma membrane (Milenkovic et al, 2009; Huangfu et al, 2006; Corbit et al, 2005; Rohatgi et al, 2007; Wang et al, 2009; Wilson et al, 2009). Like GLI2, 3 and SUFU, however, SMO may traffic through the cilium in the absence of ligand (Wilson et al, 2009; Kim et al, 2009). SMO and PTCH appear to have opposing localizations in both the 'off' and 'on' state, with PTCH exiting and SMO entering the cilium upon Hh pathway activation (Denef et al, 2000; Rohatgi et al, 2007; reviewed in Goetz and Anderson, 2010; Hui and Angers, 2011). Clearance of PTCH from the ciliary membrane in the presence of Hh is promoted by its ubiquitination by the E3 ligase SMURF (Huang et al, 2013; Yue et al, 2014)
Like the Drosophila homologue, vertebrate SMO appears to exists as a constitutive dimer. Dimerization is mediated by the N-terminal Cys-rich domain (CRD) and is required for function (Zhao et al, 2007). The C-terminal tail of SMO has arginine-rich clusters that appear to regulate the conformation of the tails in the dimer, maintaining the SMO dimer in an inactive state. In Drosophila, the inhibitory effect of the arginine-rich region is counteracted upon Hh pathway activation by PKA-mediated phosphorylation of adjacent serine residues. This promotes an open tail conformation that is required for cell surface accumulation and signaling (Zhao et al, 2007; Chen et al, 2010). These consensus PKA motifs are not conserved in the vertebrate SMO C-terminal tail, and a role for PKA-mediated phosphorylation and direct activation of SMO appears not to hold true in mammalian cells (Zhao et al, 2007; Tuson et al, 2011). A similar activating phosphorylation of vertebrate SMO may be CK1 or GRK2-dependent (Chen et al, 2011).
R-HSA-5610766 (Reactome) Vertebrate hedgehog signaling depends on the passage and/or localization of many of the pathway components through the primary cilium (reviewed in Goetz and Anderson, 2010). Although GLI and SUFU proteins are not concentrated in the cilium in the absence of Hh signaling, processing and/or degradation of the transcription factors requires transit through the cilium and basal levels of these proteins can be detected there (Wen et al, 2010; Tukachinsky et al, 2010; Kim et al, 2006; Liu et al, 2005; Haycraft et 2005). Consistent with this, members of both the IFT-B and IFT-A complex, as well as components of the ciliary basal body and the kinesin-2 and dynein motor proteins have been identified as regulators of Hh signaling (Huangfu et al, 2003; Tran et al, 2008; Liu et al, 2005; Houde et al, 2006; Huangfu et al, 2005; May et al, 2005; Cortellino et al, 2009; Vierkotten et al, 2007; Ferrante et al, 2006; Weatherbee et al, 2009; Liem et al, 2012; Qin et al 2011). KIF7, a microtubule-associated kinesin-type motor that negatively regulates the length of axonemal microtubules, is also required for correct localization of GLI:SUFU (He et al, 2014). Finally, a number of PCP pathway effectors have recently been shown to be required for ciliogenesis, and mutations in these genes disrupt GLI processing (Zeng et al, 2010; Gray et al, 2009; Heydeck et al, 2009; Park et al, 2006).
R-HSA-5610767 (Reactome) Vertebrate hedgehog signaling depends on the passage and/or localization of many of the pathway components through the primary cilium (reviewed in Goetz and Anderson, 2010). Although GLI and SUFU proteins are not concentrated in the cilium in the absence of Hh signaling, processing and/or degradation of the transcription factors requires transit through the cilium and basal levels of these proteins can be detected there (Wen et al, 2010; Tukachinsky et al, 2010; Kim et al, 2006; Liu et al, 2005; Haycraft et 2005). Consistent with this, members of both the IFT-B and IFT-A complex, as well as components of the ciliary basal body and the kinesin-2 and dynein motor proteins have been identified as regulators of Hh signaling (Huangfu et al, 2003; Tran et al, 2008; Liu et al, 2005; Houde et al, 2006; Huangfu et al, 2005; May et al, 2005; Cortellino et al, 2009; Vierkotten et al, 2007; Ferrante et al, 2006; Weatherbee et al, 2009; Liem et al, 2012; Qin et al 2011). KIF7, a microtubule-associated kinesin-type motor that negatively regulates the length of axonemal microtubules, is also required for correct localization of GLI:SUFU (He et al, 2014). Finally, a number of PCP pathway effectors have recently been shown to be required for ciliogenesis, and mutations in these genes disrupt GLI processing (Zeng et al, 2010; Gray et al, 2009; Heydeck et al, 2009; Park et al, 2006).
R-HSA-5612508 (Reactome) GLI3R is a DNA-binding transcriptional repressor that recognizes consensus GLI sites 5'-GACCACCC-3' in the promoters of target genes (Kinzler and Vogelstein, 1990). DNA-binding is mediated through 5 C2H2 Kruppel zinc fingers in the N-terminal region of the protein, which remains intact after proteasome-mediated processing (reviewed in Hui and Angers, 2011). GLI-dependent target genes have been identified by a number of ChIP based screens, and one well established target of GLI3R is the Hh receptor, PTCH1 (Lee et al, 2010; Vokes et al, 2007; Vokes et al, 2008). GLI3R has been shown to bind to a GLI-consensus sequence in the PTCH1 promoter as assesed by electrophoretic mobility shift assay and the protein is able to repress expression of a reporter gene driven by this element (Agren et al, 2004). GLI3R may promote repressive complexes at the PTCH1 promoter by the SKI1-dependent recruitment of HDAC complexes (Dai et al, 2002). Other GLI3R transcriptional targets include GLI1 and GLI2 (Hu et al, 2006).
R-HSA-5612510 (Reactome) Expression of the PTCH1 gene is repressed in the absence of Hh signaling by GLI3R (Agren et al, 2004; Lee et al, 2010; Vokes et al, 2007; Vokes et al, 2008). GLI3R may exert its repression activity through the SKI-dependent recruitment of HDACs (Dai et al, 2002).
R-HSA-5617408 (Reactome) GLI3R is a DNA-binding transcriptional repressor that recognizes consensus GLI sites 5'-GACCACCC-3' in the promoters of target genes (Kinzler and Vogelstein, 1990). DNA-binding is mediated through 5 C2H2 Kruppel zinc fingers in the N-terminal region of the protein, which remains intact after proteasome-mediated processing (reviewed in Hui and Angers, 2011). In the absence of Hh signaling, GLI3R has been shown to bind to the promoters of the GLI1 and GLI2 genes as assesed by ChIP and to repress gene expression (Hu et al, 2006).
R-HSA-5617410 (Reactome) GLI3R is a DNA-binding transcriptional repressor that recognizes consensus GLI sites 5'-GACCACCC-3' in the promoters of target genes (Kinzler and Vogelstein, 1990). DNA-binding is mediated through 5 C2H2 Kruppel zinc fingers in the N-terminal region of the protein, which remains intact after proteasome-mediated processing (reviewed in Hui and Angers, 2011). In the absence of Hh signaling, GLI3R has been shown to bind to the promoters of the GLI1 and GLI2 genes as assesed by ChIP and to repress gene expression (Hu et al, 2006).
R-HSA-5617412 (Reactome) Expression of the GLI1 gene is repressed in the absence of Hh signaling by GLI3R (Hu et al, 2006). GLI3R may exert its repression activity through the SKI-dependent recruitment of HDACs (Dai et al, 2002).
R-HSA-5617413 (Reactome) Expression of the GLI2 gene is repressed in the absence of Hh signaling by GLI3R (Hu et al, 2006). GLI3R may exert its repression activity through the SKI-dependent recruitment of HDACs (Dai et al, 2002).
SMO dimerArrowR-HSA-5610763 (Reactome)
SMO dimerR-HSA-5610763 (Reactome)
SUFUArrowR-HSA-5610754 (Reactome)
SUFUArrowR-HSA-5610757 (Reactome)
SUFUArrowR-HSA-5610758 (Reactome)
SUFUArrowR-HSA-5610760 (Reactome)
SUFUR-HSA-5610723 (Reactome)
TULP3R-HSA-5610726 (Reactome)
UbR-HSA-5610742 (Reactome)
UbR-HSA-5610745 (Reactome)
UbR-HSA-5610746 (Reactome)
adenylate cyclases:Mg2+mim-catalysisR-HSA-5610727 (Reactome)
anterograde IFT regulators of HhArrowR-HSA-5610767 (Reactome)
cAMPArrowR-HSA-5610727 (Reactome)
cAMPR-HSA-5610749 (Reactome)
ciliary basal body regulators of HhArrowR-HSA-5610766 (Reactome)
ciliary basal body regulators of HhArrowR-HSA-5610767 (Reactome)
microtubuleR-HSA-5610733 (Reactome)
p10S-GLI3:SUFUArrowR-HSA-5610722 (Reactome)
p10S-GLI3:SUFUR-HSA-5610732 (Reactome)
p11S-GLI2:SUFUArrowR-HSA-5610730 (Reactome)
p11S-GLI2:SUFUR-HSA-5610745 (Reactome)
p13S-GLI3:SUFUArrowR-HSA-5610732 (Reactome)
p13S-GLI3:SUFUR-HSA-5610746 (Reactome)
p4S-GLI2:SUFUArrowR-HSA-5610717 (Reactome)
p4S-GLI2:SUFUR-HSA-5610718 (Reactome)
p6S-GLI3:SUFUArrowR-HSA-5610720 (Reactome)
p6S-GLI3:SUFUR-HSA-5610722 (Reactome)
p8S-GLI2:SUFUArrowR-HSA-5610718 (Reactome)
p8S-GLI2:SUFUR-HSA-5610730 (Reactome)
pS-GLI1:SUFUArrowR-HSA-5610741 (Reactome)
pS-GLI1:SUFUR-HSA-5610737 (Reactome)
pS-GLI1:SUFUR-HSA-5610742 (Reactome)
retrograde IFT regulators of HhArrowR-HSA-5610766 (Reactome)
ub-p11S-GLI2:SUFUArrowR-HSA-5610745 (Reactome)
ub-p11S-GLI2:SUFUR-HSA-5610757 (Reactome)
ub-pS-GLI1:SUFUArrowR-HSA-5610742 (Reactome)
ub-pS-GLI1:SUFUR-HSA-5610758 (Reactome)
Personal tools