Senescence-associated secretory phenotype (SASP) (Homo sapiens)

From WikiPathways

Revision as of 21:17, 15 February 2018 by Vjlynch (Talk | contribs)
Jump to: navigation, search
3-5, 8, 18...81954312, 23, 46, 73, 89...8, 34, 103624, 5, 8, 9, 49...5, 8, 30, 34, 49...342, 7, 40, 61, 7817, 6284, 5, 8, 30, 34...12, 23, 46, 73, 89...25, 625, 8, 9, 49, 59...62, 63956234, 82628cytosolnucleoplasmANAPC10 H2AFV p-T160-CDK2 IL8 gene INK4CDC26 HIST1H2BN ANAPC7 FZR1 HIST2H2AA3 UBE2D1 ANAPC7 CDK4 HIST1H2BJ HIST2H2BE HIST1H2AC UBB(153-228) CDC16 EHMT1:EHMT2CDKN2D p-T235, S321-CEBPB ANAPC7 UBE2E1 CDKN2B DNA Damage/TelomereStress InducedSenescenceUBE2D1 HIST1H2BD UBC(305-380) HIST1H2BB HIST1H2BC HIST3H2BB HIST1H2BA CDC23 p-FZR1 H2AFJ CCNA2 ANAPC1 HIST1H2AB UBB(77-152) CDKN2D Oncogenic MAPKsignalingUBC(533-608) AdoHcyp-T202,Y204-MAPK3 p-S63,S73-JUN CDKN2B gene p-T235,S321-CEBPB:NF-kB:IL6 geneANAPC16 UBA52(1-76) HIST1H2BB HIST2H2AA3 HIST1H2BM UBC(533-608) ANAPC4 p-4S,T359,T573-RPS6KA1 UBB(153-228) HIST1H2BK p-2S-JUN:p-2S,2T-FOS:IGFBP7 GeneHIST2H2BE HIST1H2BN p-T235,S321-CEBPB:NF-kB:IL8 GeneH2AFV p-T185,Y187-MAPK1 HIST1H2AJ HIST1H4 HIST1H4 ANAPC15 ANAPC1 ANAPC4 IL6 gene EHMT1 CDC16 ANAPC2 ADPIL8gene:Nucleosome-H3K9Me2NFKB1(1-433) 2xMyri-IL1AANAPC2 CyclinA:Cdk2:p21/p27complexH2AFZ ANAPC1 UBE2E1 p-T235, S321-CEBPB CCNA1 HIST1H2AJ CDC16 H2BFS H2AFB1 UBE2C Me2K-10-HIST2H3A UBB(1-76) HIST1H2BO IL8 gene Interleukin-6 familysignalingUBE2D1 p-2S-JUN:p-2S,2T-FOS:IL1A geneHIST2H3A CyclinA:phospho-Cdk(Thr160):Cdh1:phosho-APC/C complexHIST1H2AB RELA ANAPC15 ANAPC10 ADPANAPC7 p-4S,T356,T570-RPS6KA2 UBB(77-152) UBC(1-76) H2AFV CDKN1A UBC(457-532) Interleukin-1 familysignalingANAPC11 ANAPC11 HIST2H2AC HIST1H2AJ HIST2H2AC HIST2H2BE CDC23 HIST1H2BL ANAPC16 p-T185,Y187-MAPK1 H3F3A FZR1 ANAPC5 HIST1H2BA HIST1H2BH HIST1H2BL CCNA1 p-2S-cJUN:p-2S,2T-cFOSHIST1H2BM p-4S,T231,T365-RPS6KA3 HIST1H2BH HIST1H2BJ UBE2C HIST1H4 EHMT1 CDK2 ANAPC16 RPS27A(1-76) CDK6 UBC(153-228) CDC26 IL8 gene HIST1H2BD H2AFB1 ATPHIST1H2AD IL8 gene:NucleosomeH2AFZ H2AFX UBC(153-228) HIST2H2AC RELA CyclinA:phospho-Cdk2(Thr160):phospho-Cdh1:phospho-APC/C complexUBC(229-304) ANAPC10 ADPATPOxidative StressInduced SenescenceNFKB1(1-433) p16-INK4a HIST1H2AD ANAPC2 CDC27 HIST3H2BB HIST1H3A UbEHMT2 Cdh1:phospho-APC/CcomplexH2AFX HIST1H2AC CDC23 HIST1H4 ANAPC1 CDC26 CDC16 HIST1H2AB ANAPC2 HIST1H2BH ANAPC15 RPS6KA1 ANAPC10 RPS6KA3 p-T235,S321-CEBPBhomodimerMe2K-10-HIST2H3A CDC23 CDKN2B geneHIST1H2BM UBE2E1 Ribosomal protein S6kinaseH2BFS ATPHIST1H2BN ANAPC11 HIST1H2BC H2AFZ p-T325,T331,S362,S374-FOS CDKN2B p-T325,T331,S362,S374-FOS HIST1H2BO ANAPC5 Me2K10-HIST1H3A HIST1H2BL CDKN2C H2AFV HIST1H2BM HIST1H2BA IGFBP7 gene UBC(229-304) HIST1H2BA CEBPBUBC(381-456) HIST1H2BH HIST1H2BN CDKN1B ANAPC4 ANAPC2 UBC(1-76) CDK6 HIST1H2BL H2BFS IL8CEBPB geneFZR1 HIST1H2AC UBC(609-684) IL6 gene UBC(381-456) Oncogene InducedSenescenceHIST1H2BJ CDC27 H2AFJ CDC26 H2AFB1 UBC(609-684) HIST1H2AD H2AFJ ANAPC16 IL1A gene HIST1H2BC UBC(305-380) HIST1H2BK UBB(1-76) NFKB1(1-433) RELA p-T325,T331,S362,S374-FOS CDK4,CDK6:INK4HIST1H2BD IL6 gene CDC27 HIST1H2AC CDC26 IL1A geneCCNA1 ANAPC5 IL6 gene:Nucleosomep-S63,S73-JUN CCNA2 HIST1H2BD HIST1H2AB HIST1H2BC ANAPC5 p-T,Y MAPK dimersp-T202,Y204-MAPK3 CDC27 UBE2C NF-kB complexHIST3H2BB H2AFZ HIST1H2BB HIST1H2BK EHMT2 HIST1H2BK CDK4,CDK6CDC16 Ub-EHMT1:Ub-EHMT2:Cdh1:p-APC/CIL6 geneHIST3H2BB HIST1H2BO ANAPC15 HIST1H2AD EHMT2 CCNA:p-T160-CDK2FZR1 Me2K-10-H3F3A CDKN2C HIST1H2AJ EHMT1:EHMT2:Cdh1:p-APC/CHIST2H2AA3 ADPUBC(77-152) HIST2H2BE p-Y705-STAT3 dimerIGFBP7ANAPC11 p16-INK4a IL8 geneH2BFS RPS27A(1-76) p-S63,S73-JUN AdoMetANAPC5 CDC23 UBE2E1 p-T235,S321-CEBPB:CDKN2B GeneUBE2D1 Me2K-10-H3F3A ANAPC10 HIST1H2BJ ANAPC16 p-T160-CDK2 HIST1H3A ANAPC15 HIST1H2BB UBC(77-152) H2AFB1 ANAPC7 ATPUBE2D1 HIST2H2AA3 p-T235, S321-CEBPB p-T235, S321-CEBPB EHMT1 ANAPC4 HIST2H2AC H2AFJ RPS6KA2 CDKN2BANAPC1 p-ERK1/2/5Phospho-Ribosomalprotein S6 kinaseCCNA1 CDK4 HIST2H3A Me2K10-HIST1H3A UBE2E1 CDC27 p-T218,Y220-MAPK7 p-Y705-STAT3 ANAPC11 HIST1H2BO IL6gene:Nucleosome-H3K9Me2ANAPC4 UBA52(1-76) UBE2C CCNA2 CCNA2 p-T235, S321-CEBPBH2AFX UBE2C IGFBP7 geneUBC(457-532) IL6H3F3A H2AFX p-T160-CDK2 p-T235-CEBPB8626219, 58, 64, 69, 116...834, 8295633215, 41, 44, 50, 71...6, 21, 22, 26, 28...39, 4514, 37, 67, 77, 80...1, 10, 11, 13, 16...


Description

The culture medium of senescent cells in enriched in secreted proteins when compared with the culture medium of quiescent i.e. presenescent cells and these secreted proteins constitute the so-called senescence-associated secretory phenotype (SASP), also known as the senescence messaging secretome (SMS). SASP components include inflammatory and immune-modulatory cytokines (e.g. IL6 and IL8), growth factors (e.g. IGFBPs), shed cell surface molecules (e.g. TNF receptors) and survival factors. While the SASP exhibits a wide ranging profile, it is not significantly affected by the type of senescence trigger (oncogenic signalling, oxidative stress or DNA damage) or the cell type (epithelial vs. mesenchymal) (Coppe et al. 2008). However, as both oxidative stress and oncogenic signaling induce DNA damage, the persistent DNA damage may be a deciding SASP initiator (Rodier et al. 2009). SASP components function in an autocrine manner, reinforcing the senescent phenotype (Kuilman et al. 2008, Acosta et al. 2008), and in the paracrine manner, where they may promote epithelial-to-mesenchymal transition (EMT) and malignancy in the nearby premalignant or malignant cells (Coppe et al. 2008). Interleukin-1-alpha (IL1A), a minor SASP component whose transcription is stimulated by the AP-1 (FOS:JUN) complex (Bailly et al. 1996), can cause paracrine senescence through IL1 and inflammasome signaling (Acosta et al. 2013).

Here, transcriptional regulatory processes that mediate the SASP are annotated. DNA damage triggers ATM-mediated activation of TP53, resulting in the increased level of CDKN1A (p21). CDKN1A-mediated inhibition of CDK2 prevents phosphorylation and inactivation of the Cdh1:APC/C complex, allowing it to ubiquitinate and target for degradation EHMT1 and EHMT2 histone methyltransferases. As EHMT1 and EHMT2 methylate and silence the promoters of IL6 and IL8 genes, degradation of these methyltransferases relieves the inhibition of IL6 and IL8 transcription (Takahashi et al. 2012). In addition, oncogenic RAS signaling activates the CEBPB (C/EBP-beta) transcription factor (Nakajima et al. 1993, Lee et al. 2010), which binds promoters of IL6 and IL8 genes and stimulates their transcription (Kuilman et al. 2008, Lee et al. 2010). CEBPB also stimulates the transcription of CDKN2B (p15-INK4B), reinforcing the cell cycle arrest (Kuilman et al. 2008). CEBPB transcription factor has three isoforms, due to three alternative translation start sites. The CEBPB-1 isoform (C/EBP-beta-1) seems to be exclusively involved in growth arrest and senescence, while the CEBPB-2 (C/EBP-beta-2) isoform may promote cellular proliferation (Atwood and Sealy 2010 and 2011). IL6 signaling stimulates the transcription of CEBPB (Niehof et al. 2001), creating a positive feedback loop (Kuilman et al. 2009, Lee et al. 2010). NF-kappa-B transcription factor is also activated in senescence (Chien et al. 2011) through IL1 signaling (Jimi et al. 1996, Hartupee et al. 2008, Orjalo et al. 2009). NF-kappa-B binds IL6 and IL8 promoters and cooperates with CEBPB transcription factor in the induction of IL6 and IL8 transcription (Matsusaka et al. 1993, Acosta et al. 2008). Besides IL6 and IL8, their receptors are also upregulated in senescence (Kuilman et al. 2008, Acosta et al. 2008) and IL6 and IL8 may be master regulators of the SASP.<p>IGFBP7 is also an SASP component that is upregulated in response to oncogenic RAS-RAF-MAPK signaling and oxidative stress, as its transcription is directly stimulated by the AP-1 (JUN:FOS) transcription factor. IGFBP7 negatively regulates RAS-RAF (BRAF)-MAPK signaling and is important for the establishment of senescence in melanocytes (Wajapeyee et al. 2008).<p>Please refer to Young and Narita 2009 for a recent review. View original pathway at:Reactome.</div>

Comments

Reactome-Converter 
Pathway is converted from Reactome ID: 2559582
Reactome-version 
Reactome version: 61
Reactome Author 
Reactome Author: Orlic-Milacic, Marija

Try the New WikiPathways

View approved pathways at the new wikipathways.org.

Quality Tags

Ontology Terms

 

Bibliography

View all...
  1. Gao H, Le Y, Wu X, Silberstein LE, Giese RW, Zhu Z.; ''VentX, a novel lymphoid-enhancing factor/T-cell factor-associated transcription repressor, is a putative tumor suppressor.''; PubMed Europe PMC Scholia
  2. Canman CE, Lim DS, Cimprich KA, Taya Y, Tamai K, Sakaguchi K, Appella E, Kastan MB, Siliciano JD.; ''Activation of the ATM kinase by ionizing radiation and phosphorylation of p53.''; PubMed Europe PMC Scholia
  3. Gao Z, Zhang J, Bonasio R, Strino F, Sawai A, Parisi F, Kluger Y, Reinberg D.; ''PCGF homologs, CBX proteins, and RYBP define functionally distinct PRC1 family complexes.''; PubMed Europe PMC Scholia
  4. Rawat VP, Arseni N, Ahmed F, Mulaw MA, Thoene S, Heilmeier B, Sadlon T, D'Andrea RJ, Hiddemann W, Bohlander SK, Buske C, Feuring-Buske M.; ''The vent-like homeobox gene VENTX promotes human myeloid differentiation and is highly expressed in acute myeloid leukemia.''; PubMed Europe PMC Scholia
  5. Niehof M, Streetz K, Rakemann T, Bischoff SC, Manns MP, Horn F, Trautwein C.; ''Interleukin-6-induced tethering of STAT3 to the LAP/C/EBPbeta promoter suggests a new mechanism of transcriptional regulation by STAT3.''; PubMed Europe PMC Scholia
  6. Taga T, Kishimoto T.; ''Gp130 and the interleukin-6 family of cytokines.''; PubMed Europe PMC Scholia
  7. Moynagh PN.; ''The Pellino family: IRAK E3 ligases with emerging roles in innate immune signalling.''; PubMed Europe PMC Scholia
  8. McLaughlin MM, Kumar S, McDonnell PC, Van Horn S, Lee JC, Livi GP, Young PR.; ''Identification of mitogen-activated protein (MAP) kinase-activated protein kinase-3, a novel substrate of CSBP p38 MAP kinase.''; PubMed Europe PMC Scholia
  9. Ainbinder E, Bergelson S, Pinkus R, Daniel V.; ''Regulatory mechanisms involved in activator-protein-1 (AP-1)-mediated activation of glutathione-S-transferase gene expression by chemical agents.''; PubMed Europe PMC Scholia
  10. Parry D, Bates S, Mann DJ, Peters G.; ''Lack of cyclin D-Cdk complexes in Rb-negative cells correlates with high levels of p16INK4/MTS1 tumour suppressor gene product.''; PubMed Europe PMC Scholia
  11. Huang TT, Vinci JM, Lan L, Jeffrey JJ, Wilcox BD.; ''Serotonin-inducible transcription of interleukin-1alpha in uterine smooth muscle cells requires an AP-1 site: cloning and partial characterization of the rat IL-1alpha promoter.''; PubMed Europe PMC Scholia
  12. To KH, Pajovic S, Gallie BL, Thériault BL.; ''Regulation of p14ARF expression by miR-24: a potential mechanism compromising the p53 response during retinoblastoma development.''; PubMed Europe PMC Scholia
  13. Ranganathan A, Pearson GW, Chrestensen CA, Sturgill TW, Cobb MH.; ''The MAP kinase ERK5 binds to and phosphorylates p90 RSK.''; PubMed Europe PMC Scholia
  14. Agger K, Cloos PA, Rudkjaer L, Williams K, Andersen G, Christensen J, Helin K.; ''The H3K27me3 demethylase JMJD3 contributes to the activation of the INK4A-ARF locus in response to oncogene- and stress-induced senescence.''; PubMed Europe PMC Scholia
  15. Lavoie H, Therrien M.; ''Regulation of RAF protein kinases in ERK signalling.''; PubMed Europe PMC Scholia
  16. Sharpless NE, Sherr CJ.; ''Forging a signature of in vivo senescence.''; PubMed Europe PMC Scholia
  17. Guan KL, Jenkins CW, Li Y, O'Keefe CL, Noh S, Wu X, Zariwala M, Matera AG, Xiong Y.; ''Isolation and characterization of p19INK4d, a p16-related inhibitor specific to CDK6 and CDK4.''; PubMed Europe PMC Scholia
  18. Meng W, Swenson LL, Fitzgibbon MJ, Hayakawa K, Ter Haar E, Behrens AE, Fulghum JR, Lippke JA.; ''Structure of mitogen-activated protein kinase-activated protein (MAPKAP) kinase 2 suggests a bifunctional switch that couples kinase activation with nuclear export.''; PubMed Europe PMC Scholia
  19. Libermann TA, Baltimore D.; ''Activation of interleukin-6 gene expression through the NF-kappa B transcription factor.''; PubMed Europe PMC Scholia
  20. Baek KH, Ryeom S.; ''Detection of Oncogene-Induced Senescence In Vivo.''; PubMed Europe PMC Scholia
  21. Palomo J, Dietrich D, Martin P, Palmer G, Gabay C.; ''The interleukin (IL)-1 cytokine family--Balance between agonists and antagonists in inflammatory diseases.''; PubMed Europe PMC Scholia
  22. el-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent JM, Lin D, Mercer WE, Kinzler KW, Vogelstein B.; ''WAF1, a potential mediator of p53 tumor suppression.''; PubMed Europe PMC Scholia
  23. Fang S, Jensen JP, Ludwig RL, Vousden KH, Weissman AM.; ''Mdm2 is a RING finger-dependent ubiquitin protein ligase for itself and p53.''; PubMed Europe PMC Scholia
  24. Senturk S, Mumcuoglu M, Gursoy-Yuzugullu O, Cingoz B, Akcali KC, Ozturk M.; ''Transforming growth factor-beta induces senescence in hepatocellular carcinoma cells and inhibits tumor growth.''; PubMed Europe PMC Scholia
  25. Umair Z, Kumar S, Kim DH, Rafiq K, Kumar V, Kim S, Park JB, Lee JY, Lee U, Kim J.; ''Ventx1.1 as a Direct Repressor of Early Neural Gene zic3 in Xenopus laevis.''; PubMed Europe PMC Scholia
  26. Tachibana M, Ueda J, Fukuda M, Takeda N, Ohta T, Iwanari H, Sakihama T, Kodama T, Hamakubo T, Shinkai Y.; ''Histone methyltransferases G9a and GLP form heteromeric complexes and are both crucial for methylation of euchromatin at H3-K9.''; PubMed Europe PMC Scholia
  27. Weinmann AS, Bartley SM, Zhang T, Zhang MQ, Farnham PJ.; ''Use of chromatin immunoprecipitation to clone novel E2F target promoters.''; PubMed Europe PMC Scholia
  28. Atwood AA, Sealy LJ.; ''C/EBPβ's role in determining Ras-induced senescence or transformation.''; PubMed Europe PMC Scholia
  29. Sun P, Yoshizuka N, New L, Moser BA, Li Y, Liao R, Xie C, Chen J, Deng Q, Yamout M, Dong MQ, Frangou CG, Yates JR, Wright PE, Han J.; ''PRAK is essential for ras-induced senescence and tumor suppression.''; PubMed Europe PMC Scholia
  30. Heinrich PC, Behrmann I, Haan S, Hermanns HM, Müller-Newen G, Schaper F.; ''Principles of interleukin (IL)-6-type cytokine signalling and its regulation.''; PubMed Europe PMC Scholia
  31. Ben-Levy R, Leighton IA, Doza YN, Attwood P, Morrice N, Marshall CJ, Cohen P.; ''Identification of novel phosphorylation sites required for activation of MAPKAP kinase-2.''; PubMed Europe PMC Scholia
  32. Sithanandam G, Latif F, Duh FM, Bernal R, Smola U, Li H, Kuzmin I, Wixler V, Geil L, Shrestha S.; ''3pK, a new mitogen-activated protein kinase-activated protein kinase located in the small cell lung cancer tumor suppressor gene region.''; PubMed Europe PMC Scholia
  33. Moretti PA, Davidson AJ, Baker E, Lilley B, Zon LI, D'Andrea RJ.; ''Molecular cloning of a human Vent-like homeobox gene.''; PubMed Europe PMC Scholia
  34. Kotake Y, Cao R, Viatour P, Sage J, Zhang Y, Xiong Y.; ''pRB family proteins are required for H3K27 trimethylation and Polycomb repression complexes binding to and silencing p16INK4alpha tumor suppressor gene.''; PubMed Europe PMC Scholia
  35. Onichtchouk D, Gawantka V, Dosch R, Delius H, Hirschfeld K, Blumenstock C, Niehrs C.; ''The Xvent-2 homeobox gene is part of the BMP-4 signalling pathway controlling [correction of controling] dorsoventral patterning of Xenopus mesoderm.''; PubMed Europe PMC Scholia
  36. Atwood AA, Sealy L.; ''Regulation of C/EBPbeta1 by Ras in mammary epithelial cells and the role of C/EBPbeta1 in oncogene-induced senescence.''; PubMed Europe PMC Scholia
  37. Barradas M, Anderton E, Acosta JC, Li S, Banito A, Rodriguez-Niedenführ M, Maertens G, Banck M, Zhou MM, Walsh MJ, Peters G, Gil J.; ''Histone demethylase JMJD3 contributes to epigenetic control of INK4a/ARF by oncogenic RAS.''; PubMed Europe PMC Scholia
  38. Harley CB, Futcher AB, Greider CW.; ''Telomeres shorten during ageing of human fibroblasts.''; PubMed Europe PMC Scholia
  39. Chien Y, Scuoppo C, Wang X, Fang X, Balgley B, Bolden JE, Premsrirut P, Luo W, Chicas A, Lee CS, Kogan SC, Lowe SW.; ''Control of the senescence-associated secretory phenotype by NF-κB promotes senescence and enhances chemosensitivity.''; PubMed Europe PMC Scholia
  40. Kuzmichev A, Nishioka K, Erdjument-Bromage H, Tempst P, Reinberg D.; ''Histone methyltransferase activity associated with a human multiprotein complex containing the Enhancer of Zeste protein.''; PubMed Europe PMC Scholia
  41. New L, Jiang Y, Han J.; ''Regulation of PRAK subcellular location by p38 MAP kinases.''; PubMed Europe PMC Scholia
  42. Bracken AP, Pasini D, Capra M, Prosperini E, Colli E, Helin K.; ''EZH2 is downstream of the pRB-E2F pathway, essential for proliferation and amplified in cancer.''; PubMed Europe PMC Scholia
  43. Acosta JC, O'Loghlen A, Banito A, Guijarro MV, Augert A, Raguz S, Fumagalli M, Da Costa M, Brown C, Popov N, Takatsu Y, Melamed J, d'Adda di Fagagna F, Bernard D, Hernando E, Gil J.; ''Chemokine signaling via the CXCR2 receptor reinforces senescence.''; PubMed Europe PMC Scholia
  44. Wu X, Gao H, Ke W, Giese RW, Zhu Z.; ''The homeobox transcription factor VentX controls human macrophage terminal differentiation and proinflammatory activation.''; PubMed Europe PMC Scholia
  45. Moiseeva O, Mallette FA, Mukhopadhyay UK, Moores A, Ferbeyre G.; ''DNA damage signaling and p53-dependent senescence after prolonged beta-interferon stimulation.''; PubMed Europe PMC Scholia
  46. Davidson AJ, Zon LI.; ''Turning mesoderm into blood: the formation of hematopoietic stem cells during embryogenesis.''; PubMed Europe PMC Scholia
  47. Rodier F, Coppé JP, Patil CK, Hoeijmakers WA, Muñoz DP, Raza SR, Freund A, Campeau E, Davalos AR, Campisi J.; ''Persistent DNA damage signalling triggers senescence-associated inflammatory cytokine secretion.''; PubMed Europe PMC Scholia
  48. Nakajima T, Kinoshita S, Sasagawa T, Sasaki K, Naruto M, Kishimoto T, Akira S.; ''Phosphorylation at threonine-235 by a ras-dependent mitogen-activated protein kinase cascade is essential for transcription factor NF-IL6.''; PubMed Europe PMC Scholia
  49. Jimi E, Ikebe T, Takahashi N, Hirata M, Suda T, Koga T.; ''Interleukin-1 alpha activates an NF-kappaB-like factor in osteoclast-like cells.''; PubMed Europe PMC Scholia
  50. de Lange T.; ''Shelterin: the protein complex that shapes and safeguards human telomeres.''; PubMed Europe PMC Scholia
  51. Saitoh M, Nishitoh H, Fujii M, Takeda K, Tobiume K, Sawada Y, Kawabata M, Miyazono K, Ichijo H.; ''Mammalian thioredoxin is a direct inhibitor of apoptosis signal-regulating kinase (ASK) 1.''; PubMed Europe PMC Scholia
  52. Sgouras DN, Athanasiou MA, Beal GJ, Fisher RJ, Blair DG, Mavrothalassitis GJ.; ''ERF: an ETS domain protein with strong transcriptional repressor activity, can suppress ets-associated tumorigenesis and is regulated by phosphorylation during cell cycle and mitogenic stimulation.''; PubMed Europe PMC Scholia
  53. Scerbo P, Marchal L, Kodjabachian L.; ''Lineage commitment of embryonic cells involves MEK1-dependent clearance of pluripotency regulator Ventx2.''; PubMed Europe PMC Scholia
  54. Ichijo H, Nishida E, Irie K, ten Dijke P, Saitoh M, Moriguchi T, Takagi M, Matsumoto K, Miyazono K, Gotoh Y.; ''Induction of apoptosis by ASK1, a mammalian MAPKKK that activates SAPK/JNK and p38 signaling pathways.''; PubMed Europe PMC Scholia
  55. Bailly S, Fay M, Israël N, Gougerot-Pocidalo MA.; ''The transcription factor AP-1 binds to the human interleukin 1 alpha promoter.''; PubMed Europe PMC Scholia
  56. Matsuura H, Nishitoh H, Takeda K, Matsuzawa A, Amagasa T, Ito M, Yoshioka K, Ichijo H.; ''Phosphorylation-dependent scaffolding role of JSAP1/JIP3 in the ASK1-JNK signaling pathway. A new mode of regulation of the MAP kinase cascade.''; PubMed Europe PMC Scholia
  57. Fuchs SY, Adler V, Buschmann T, Wu X, Ronai Z.; ''Mdm2 association with p53 targets its ubiquitination.''; PubMed Europe PMC Scholia
  58. Nitz R, Lokau J, Aparicio-Siegmund S, Scheller J, Garbers C.; ''Modular organization of Interleukin-6 and Interleukin-11 α-receptors.''; PubMed Europe PMC Scholia
  59. Sørensen CS, Lukas C, Kramer ER, Peters JM, Bartek J, Lukas J.; ''A conserved cyclin-binding domain determines functional interplay between anaphase-promoting complex-Cdh1 and cyclin A-Cdk2 during cell cycle progression.''; PubMed Europe PMC Scholia
  60. Kunsch C, Rosen CA.; ''NF-kappa B subunit-specific regulation of the interleukin-8 promoter.''; PubMed Europe PMC Scholia
  61. Scerbo P, Girardot F, Vivien C, Markov GV, Luxardi G, Demeneix B, Kodjabachian L, Coen L.; ''Ventx factors function as Nanog-like guardians of developmental potential in Xenopus.''; PubMed Europe PMC Scholia
  62. Wu X, Gao H, Ke W, Hager M, Xiao S, Freeman MR, Zhu Z.; ''VentX trans-activates p53 and p16ink4a to regulate cellular senescence.''; PubMed Europe PMC Scholia
  63. Yu TW, Anderson D.; ''Reactive oxygen species-induced DNA damage and its modification: a chemical investigation.''; PubMed Europe PMC Scholia
  64. Serrano M, Hannon GJ, Beach D.; ''A new regulatory motif in cell-cycle control causing specific inhibition of cyclin D/CDK4.''; PubMed Europe PMC Scholia
  65. Foulds CE, Nelson ML, Blaszczak AG, Graves BJ.; ''Ras/mitogen-activated protein kinase signaling activates Ets-1 and Ets-2 by CBP/p300 recruitment.''; PubMed Europe PMC Scholia
  66. Nicke B, Bastien J, Khanna SJ, Warne PH, Cowling V, Cook SJ, Peters G, Delpuech O, Schulze A, Berns K, Mullenders J, Beijersbergen RL, Bernards R, Ganesan TS, Downward J, Hancock DC.; ''Involvement of MINK, a Ste20 family kinase, in Ras oncogene-induced growth arrest in human ovarian surface epithelial cells.''; PubMed Europe PMC Scholia
  67. Hartupee J, Li X, Hamilton T.; ''Interleukin 1alpha-induced NFkappaB activation and chemokine mRNA stabilization diverge at IRAK1.''; PubMed Europe PMC Scholia
  68. Stephen AG, Esposito D, Bagni RK, McCormick F.; ''Dragging ras back in the ring.''; PubMed Europe PMC Scholia
  69. Okazaki K, Sagata N.; ''The Mos/MAP kinase pathway stabilizes c-Fos by phosphorylation and augments its transforming activity in NIH 3T3 cells.''; PubMed Europe PMC Scholia
  70. Moerman-Herzog AM, Barger SW.; ''A polymorphism in the upstream regulatory region of the interleukin-1α gene confers differential binding by transcription factors of the AP-1 family.''; PubMed Europe PMC Scholia
  71. Arend WP, Palmer G, Gabay C.; ''IL-1, IL-18, and IL-33 families of cytokines.''; PubMed Europe PMC Scholia
  72. Takahashi A, Imai Y, Yamakoshi K, Kuninaka S, Ohtani N, Yoshimoto S, Hori S, Tachibana M, Anderton E, Takeuchi T, Shinkai Y, Peters G, Saya H, Hara E.; ''DNA damage signaling triggers degradation of histone methyltransferases through APC/C(Cdh1) in senescent cells.''; PubMed Europe PMC Scholia
  73. Lito P, Rosen N, Solit DB.; ''Tumor adaptation and resistance to RAF inhibitors.''; PubMed Europe PMC Scholia
  74. Imai Y, Gates MA, Melby AE, Kimelman D, Schier AF, Talbot WS.; ''The homeobox genes vox and vent are redundant repressors of dorsal fates in zebrafish.''; PubMed Europe PMC Scholia
  75. Gao H, Wu B, Le Y, Zhu Z.; ''Homeobox protein VentX induces p53-independent apoptosis in cancer cells.''; PubMed Europe PMC Scholia
  76. Takekawa M, Tatebayashi K, Saito H.; ''Conserved docking site is essential for activation of mammalian MAP kinase kinases by specific MAP kinase kinase kinases.''; PubMed Europe PMC Scholia
  77. Alheim K, McDowell TL, Symons JA, Duff GW, Bartfai T.; ''An AP-1 site is involved in the NGF induction of IL-1 alpha in PC12 cells.''; PubMed Europe PMC Scholia
  78. Agherbi H, Gaussmann-Wenger A, Verthuy C, Chasson L, Serrano M, Djabali M.; ''Polycomb mediated epigenetic silencing and replication timing at the INK4a/ARF locus during senescence.''; PubMed Europe PMC Scholia
  79. Guan KL, Jenkins CW, Li Y, Nichols MA, Wu X, O'Keefe CL, Matera AG, Xiong Y.; ''Growth suppression by p18, a p16INK4/MTS1- and p14INK4B/MTS2-related CDK6 inhibitor, correlates with wild-type pRb function.''; PubMed Europe PMC Scholia
  80. Lee S, Miller M, Shuman JD, Johnson PF.; ''CCAAT/Enhancer-binding protein beta DNA binding is auto-inhibited by multiple elements that also mediate association with p300/CREB-binding protein (CBP).''; PubMed Europe PMC Scholia
  81. Zhang H, Cohen SN.; ''Smurf2 up-regulation activates telomere-dependent senescence.''; PubMed Europe PMC Scholia
  82. Malumbres M, Pérez De Castro I, Hernández MI, Jiménez M, Corral T, Pellicer A.; ''Cellular response to oncogenic ras involves induction of the Cdk4 and Cdk6 inhibitor p15(INK4b).''; PubMed Europe PMC Scholia
  83. Deacon K, Blank JL.; ''Characterization of the mitogen-activated protein kinase kinase 4 (MKK4)/c-Jun NH2-terminal kinase 1 and MKK3/p38 pathways regulated by MEK kinases 2 and 3. MEK kinase 3 activates MKK3 but does not cause activation of p38 kinase in vivo.''; PubMed Europe PMC Scholia
  84. Yang BS, Hauser CA, Henkel G, Colman MS, Van Beveren C, Stacey KJ, Hume DA, Maki RA, Ostrowski MC.; ''Ras-mediated phosphorylation of a conserved threonine residue enhances the transactivation activities of c-Ets1 and c-Ets2.''; PubMed Europe PMC Scholia
  85. Pylayeva-Gupta Y, Grabocka E, Bar-Sagi D.; ''RAS oncogenes: weaving a tumorigenic web.''; PubMed Europe PMC Scholia
  86. Hastie ND, Dempster M, Dunlop MG, Thompson AM, Green DK, Allshire RC.; ''Telomere reduction in human colorectal carcinoma and with ageing.''; PubMed Europe PMC Scholia
  87. Quelle DE, Zindy F, Ashmun RA, Sherr CJ.; ''Alternative reading frames of the INK4a tumor suppressor gene encode two unrelated proteins capable of inducing cell cycle arrest.''; PubMed Europe PMC Scholia
  88. Heinrich PC, Behrmann I, Müller-Newen G, Schaper F, Graeve L.; ''Interleukin-6-type cytokine signalling through the gp130/Jak/STAT pathway.''; PubMed Europe PMC Scholia
  89. Lee S, Shuman JD, Guszczynski T, Sakchaisri K, Sebastian T, Copeland TD, Miller M, Cohen MS, Taunton J, Smart RC, Xiao Z, Yu LR, Veenstra TD, Johnson PF.; ''RSK-mediated phosphorylation in the C/EBP{beta} leucine zipper regulates DNA binding, dimerization, and growth arrest activity.''; PubMed Europe PMC Scholia
  90. Lee JH, Paull TT.; ''ATM activation by DNA double-strand breaks through the Mre11-Rad50-Nbs1 complex.''; PubMed Europe PMC Scholia
  91. Fleming Y, Armstrong CG, Morrice N, Paterson A, Goedert M, Cohen P.; ''Synergistic activation of stress-activated protein kinase 1/c-Jun N-terminal kinase (SAPK1/JNK) isoforms by mitogen-activated protein kinase kinase 4 (MKK4) and MKK7.''; PubMed Europe PMC Scholia
  92. Coppé JP, Patil CK, Rodier F, Sun Y, Muñoz DP, Goldstein J, Nelson PS, Desprez PY, Campisi J.; ''Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor.''; PubMed Europe PMC Scholia
  93. Smogorzewska A, van Steensel B, Bianchi A, Oelmann S, Schaefer MR, Schnapp G, de Lange T.; ''Control of human telomere length by TRF1 and TRF2.''; PubMed Europe PMC Scholia
  94. Karlseder J, Broccoli D, Dai Y, Hardy S, de Lange T.; ''p53- and ATM-dependent apoptosis induced by telomeres lacking TRF2.''; PubMed Europe PMC Scholia
  95. Kuilman T, Michaloglou C, Vredeveld LC, Douma S, van Doorn R, Desmet CJ, Aarden LA, Mooi WJ, Peeper DS.; ''Oncogene-induced senescence relayed by an interleukin-dependent inflammatory network.''; PubMed Europe PMC Scholia
  96. Bembenek J, Yu H.; ''Regulation of the anaphase-promoting complex by the dual specificity phosphatase human Cdc14a.''; PubMed Europe PMC Scholia
  97. Samatar AA, Poulikakos PI.; ''Targeting RAS-ERK signalling in cancer: promises and challenges.''; PubMed Europe PMC Scholia
  98. Rogers CD, Archer TC, Cunningham DD, Grammer TC, Casey EM.; ''Sox3 expression is maintained by FGF signaling and restricted to the neural plate by Vent proteins in the Xenopus embryo.''; PubMed Europe PMC Scholia
  99. Wu Y, Xiao S, Zhu XD.; ''MRE11-RAD50-NBS1 and ATM function as co-mediators of TRF1 in telomere length control.''; PubMed Europe PMC Scholia
  100. Ohtani N, Zebedee Z, Huot TJ, Stinson JA, Sugimoto M, Ohashi Y, Sharrocks AD, Peters G, Hara E.; ''Opposing effects of Ets and Id proteins on p16INK4a expression during cellular senescence.''; PubMed Europe PMC Scholia
  101. Roux PP, Richards SA, Blenis J.; ''Phosphorylation of p90 ribosomal S6 kinase (RSK) regulates extracellular signal-regulated kinase docking and RSK activity.''; PubMed Europe PMC Scholia
  102. White A, Pargellis CA, Studts JM, Werneburg BG, Farmer BT.; ''Molecular basis of MAPK-activated protein kinase 2:p38 assembly.''; PubMed Europe PMC Scholia
  103. Stein B, Baldwin AS.; ''Distinct mechanisms for regulation of the interleukin-8 gene involve synergism and cooperativity between C/EBP and NF-kappa B.''; PubMed Europe PMC Scholia
  104. Acosta JC, Banito A, Wuestefeld T, Georgilis A, Janich P, Morton JP, Athineos D, Kang TW, Lasitschka F, Andrulis M, Pascual G, Morris KJ, Khan S, Jin H, Dharmalingam G, Snijders AP, Carroll T, Capper D, Pritchard C, Inman GJ, Longerich T, Sansom OJ, Benitah SA, Zender L, Gil J.; ''A complex secretory program orchestrated by the inflammasome controls paracrine senescence.''; PubMed Europe PMC Scholia
  105. Erickson S, Sangfelt O, Heyman M, Castro J, Einhorn S, Grandér D.; ''Involvement of the Ink4 proteins p16 and p15 in T-lymphocyte senescence.''; PubMed Europe PMC Scholia
  106. Wajapeyee N, Serra RW, Zhu X, Mahalingam M, Green MR.; ''Oncogenic BRAF induces senescence and apoptosis through pathways mediated by the secreted protein IGFBP7.''; PubMed Europe PMC Scholia
  107. Dinarello CA.; ''Immunological and inflammatory functions of the interleukin-1 family.''; PubMed Europe PMC Scholia
  108. Lal A, Kim HH, Abdelmohsen K, Kuwano Y, Pullmann R, Srikantan S, Subrahmanyam R, Martindale JL, Yang X, Ahmed F, Navarro F, Dykxhoorn D, Lieberman J, Gorospe M.; ''p16(INK4a) translation suppressed by miR-24.''; PubMed Europe PMC Scholia
  109. Prior IA, Lewis PD, Mattos C.; ''A comprehensive survey of Ras mutations in cancer.''; PubMed Europe PMC Scholia
  110. Lukas C, Sørensen CS, Kramer E, Santoni-Rugiu E, Lindeneg C, Peters JM, Bartek J, Lukas J.; ''Accumulation of cyclin B1 requires E2F and cyclin-A-dependent rearrangement of the anaphase-promoting complex.''; PubMed Europe PMC Scholia
  111. Orjalo AV, Bhaumik D, Gengler BK, Scott GK, Campisi J.; ''Cell surface-bound IL-1alpha is an upstream regulator of the senescence-associated IL-6/IL-8 cytokine network.''; PubMed Europe PMC Scholia
  112. Khanna KK, Keating KE, Kozlov S, Scott S, Gatei M, Hobson K, Taya Y, Gabrielli B, Chan D, Lees-Miller SP, Lavin MF.; ''ATM associates with and phosphorylates p53: mapping the region of interaction.''; PubMed Europe PMC Scholia
  113. Baron VT, Pio R, Jia Z, Mercola D.; ''Early Growth Response 3 regulates genes of inflammation and directly activates IL6 and IL8 expression in prostate cancer.''; PubMed Europe PMC Scholia
  114. Murakami Y, Watari K, Shibata T, Uba M, Ureshino H, Kawahara A, Abe H, Izumi H, Mukaida N, Kuwano M, Ono M.; ''N-myc downstream-regulated gene 1 promotes tumor inflammatory angiogenesis through JNK activation and autocrine loop of interleukin-1α by human gastric cancer cells.''; PubMed Europe PMC Scholia
  115. Garbers C, Hermanns HM, Schaper F, Müller-Newen G, Grötzinger J, Rose-John S, Scheller J.; ''Plasticity and cross-talk of interleukin 6-type cytokines.''; PubMed Europe PMC Scholia
  116. Bracken AP, Kleine-Kohlbrecher D, Dietrich N, Pasini D, Gargiulo G, Beekman C, Theilgaard-Mönch K, Minucci S, Porse BT, Marine JC, Hansen KH, Helin K.; ''The Polycomb group proteins bind throughout the INK4A-ARF locus and are disassociated in senescent cells.''; PubMed Europe PMC Scholia
  117. Taga T.; ''Gp130, a shared signal transducing receptor component for hematopoietic and neuropoietic cytokines.''; PubMed Europe PMC Scholia
  118. Kishimoto T, Akira S, Narazaki M, Taga T.; ''Interleukin-6 family of cytokines and gp130.''; PubMed Europe PMC Scholia
  119. Burleson FG, Simeonova PP, Germolec DR, Luster MI.; ''Dermatotoxic chemical stimulate of c-jun and c-fos transcription and AP-1 DNA binding in human keratinocytes.''; PubMed Europe PMC Scholia
  120. Vijayachandra K, Lee J, Glick AB.; ''Smad3 regulates senescence and malignant conversion in a mouse multistage skin carcinogenesis model.''; PubMed Europe PMC Scholia
  121. Hannon GJ, Beach D.; ''p15INK4B is a potential effector of TGF-beta-induced cell cycle arrest.''; PubMed Europe PMC Scholia
  122. Nakashima K, Taga T.; ''gp130 and the IL-6 family of cytokines: signaling mechanisms and thrombopoietic activities.''; PubMed Europe PMC Scholia
  123. Niu J, Li Z, Peng B, Chiao PJ.; ''Identification of an autoregulatory feedback pathway involving interleukin-1alpha in induction of constitutive NF-kappaB activation in pancreatic cancer cells.''; PubMed Europe PMC Scholia
  124. Murphy LO, Smith S, Chen RH, Fingar DC, Blenis J.; ''Molecular interpretation of ERK signal duration by immediate early gene products.''; PubMed Europe PMC Scholia
  125. Clifton AD, Young PR, Cohen P.; ''A comparison of the substrate specificity of MAPKAP kinase-2 and MAPKAP kinase-3 and their activation by cytokines and cellular stress.''; PubMed Europe PMC Scholia
  126. Dietrich N, Bracken AP, Trinh E, Schjerling CK, Koseki H, Rappsilber J, Helin K, Hansen KH.; ''Bypass of senescence by the polycomb group protein CBX8 through direct binding to the INK4A-ARF locus.''; PubMed Europe PMC Scholia
  127. Le Gallic L, Virgilio L, Cohen P, Biteau B, Mavrothalassitis G.; ''ERF nuclear shuttling, a continuous monitor of Erk activity that links it to cell cycle progression.''; PubMed Europe PMC Scholia
  128. Lukas SM, Kroe RR, Wildeson J, Peet GW, Frego L, Davidson W, Ingraham RH, Pargellis CA, Labadia ME, Werneburg BG.; ''Catalysis and function of the p38 alpha.MK2a signaling complex.''; PubMed Europe PMC Scholia
  129. Maertens O, Cichowski K.; ''An expanding role for RAS GTPase activating proteins (RAS GAPs) in cancer.''; PubMed Europe PMC Scholia
  130. Raingeaud J, Whitmarsh AJ, Barrett T, Dérijard B, Davis RJ.; ''MKK3- and MKK6-regulated gene expression is mediated by the p38 mitogen-activated protein kinase signal transduction pathway.''; PubMed Europe PMC Scholia
  131. Harper JW, Adami GR, Wei N, Keyomarsi K, Elledge SJ.; ''The p21 Cdk-interacting protein Cip1 is a potent inhibitor of G1 cyclin-dependent kinases.''; PubMed Europe PMC Scholia
  132. Shimizu H, Mitomo K, Watanabe T, Okamoto S, Yamamoto K.; ''Involvement of a NF-kappa B-like transcription factor in the activation of the interleukin-6 gene by inflammatory lymphokines.''; PubMed Europe PMC Scholia
  133. Parisi T, Pollice A, Di Cristofano A, Calabrò V, La Mantia G.; ''Transcriptional regulation of the human tumor suppressor p14(ARF) by E2F1, E2F2, E2F3, and Sp1-like factors.''; PubMed Europe PMC Scholia
  134. Mizukami Y, Yoshioka K, Morimoto S, Yoshida Ki.; ''A novel mechanism of JNK1 activation. Nuclear translocation and activation of JNK1 during ischemia and reperfusion.''; PubMed Europe PMC Scholia
  135. Zhong YF, Holland PW.; ''The dynamics of vertebrate homeobox gene evolution: gain and loss of genes in mouse and human lineages.''; PubMed Europe PMC Scholia
  136. Zhang Y, Xiong Y, Yarbrough WG.; ''ARF promotes MDM2 degradation and stabilizes p53: ARF-INK4a locus deletion impairs both the Rb and p53 tumor suppression pathways.''; PubMed Europe PMC Scholia
  137. Trinh BQ, Barengo N, Kim SB, Lee JS, Zweidler-McKay PA, Naora H.; ''The homeobox gene DLX4 regulates erythro-megakaryocytic differentiation by stimulating IL-1β and NF-κB signaling.''; PubMed Europe PMC Scholia
  138. Glover JN, Harrison SC.; ''Crystal structure of the heterodimeric bZIP transcription factor c-Fos-c-Jun bound to DNA.''; PubMed Europe PMC Scholia
  139. Lin TY, Cheng YC, Yang HC, Lin WC, Wang CC, Lai PL, Shieh SY.; ''Loss of the candidate tumor suppressor BTG3 triggers acute cellular senescence via the ERK-JMJD3-p16(INK4a) signaling axis.''; PubMed Europe PMC Scholia
  140. Wu X, Gao H, Bleday R, Zhu Z.; ''Homeobox transcription factor VentX regulates differentiation and maturation of human dendritic cells.''; PubMed Europe PMC Scholia
  141. Young AR, Narita M.; ''SASP reflects senescence.''; PubMed Europe PMC Scholia
  142. Nelson ML, Kang HS, Lee GM, Blaszczak AG, Lau DK, McIntosh LP, Graves BJ.; ''Ras signaling requires dynamic properties of Ets1 for phosphorylation-enhanced binding to coactivator CBP.''; PubMed Europe PMC Scholia
  143. Moiseeva O, Bourdeau V, Roux A, Deschênes-Simard X, Ferbeyre G.; ''Mitochondrial dysfunction contributes to oncogene-induced senescence.''; PubMed Europe PMC Scholia
  144. Wu X, Bayle JH, Olson D, Levine AJ.; ''The p53-mdm-2 autoregulatory feedback loop.''; PubMed Europe PMC Scholia
  145. Seidel JJ, Graves BJ.; ''An ERK2 docking site in the Pointed domain distinguishes a subset of ETS transcription factors.''; PubMed Europe PMC Scholia
  146. Le Y, Gao H, Bleday R, Zhu Z.; ''The homeobox protein VentX reverts immune suppression in the tumor microenvironment.''; PubMed Europe PMC Scholia
  147. Voncken JW, Niessen H, Neufeld B, Rennefahrt U, Dahlmans V, Kubben N, Holzer B, Ludwig S, Rapp UR.; ''MAPKAP kinase 3pK phosphorylates and regulates chromatin association of the polycomb group protein Bmi1.''; PubMed Europe PMC Scholia
  148. Matsusaka T, Fujikawa K, Nishio Y, Mukaida N, Matsushima K, Kishimoto T, Akira S.; ''Transcription factors NF-IL6 and NF-kappa B synergistically activate transcription of the inflammatory cytokines, interleukin 6 and interleukin 8.''; PubMed Europe PMC Scholia
  149. New L, Jiang Y, Zhao M, Liu K, Zhu W, Flood LJ, Kato Y, Parry GC, Han J.; ''PRAK, a novel protein kinase regulated by the p38 MAP kinase.''; PubMed Europe PMC Scholia
  150. Kishimoto T.; ''IL-6: from its discovery to clinical applications.''; PubMed Europe PMC Scholia
  151. Banin S, Moyal L, Shieh S, Taya Y, Anderson CW, Chessa L, Smorodinsky NI, Prives C, Reiss Y, Shiloh Y, Ziv Y.; ''Enhanced phosphorylation of p53 by ATM in response to DNA damage.''; PubMed Europe PMC Scholia

History

View all...
CompareRevisionActionTimeUserComment
129524view01:02, 9 May 2024EweitzModified title
114857view16:36, 25 January 2021ReactomeTeamReactome version 75
113303view11:37, 2 November 2020ReactomeTeamReactome version 74
112515view15:47, 9 October 2020ReactomeTeamReactome version 73
101427view11:30, 1 November 2018ReactomeTeamreactome version 66
100965view21:07, 31 October 2018ReactomeTeamreactome version 65
100502view19:42, 31 October 2018ReactomeTeamreactome version 64
100048view16:25, 31 October 2018ReactomeTeamreactome version 63
99600view14:59, 31 October 2018ReactomeTeamreactome version 62 (2nd attempt)
99216view12:44, 31 October 2018ReactomeTeamreactome version 62
96095view21:17, 15 February 2018VjlynchAdded IGFBP genes
93504view11:25, 9 August 2017ReactomeTeamreactome version 61
86599view09:21, 11 July 2016ReactomeTeamreactome version 56
83199view10:21, 18 November 2015ReactomeTeamVersion54
81577view13:07, 21 August 2015ReactomeTeamNew pathway

External references

DataNodes

View all...
NameTypeDatabase referenceComment
2xMyri-IL1AProteinP01583 (Uniprot-TrEMBL)
ADPMetaboliteCHEBI:16761 (ChEBI)
ANAPC1 ProteinQ9H1A4 (Uniprot-TrEMBL)
ANAPC10 ProteinQ9UM13 (Uniprot-TrEMBL)
ANAPC11 ProteinQ9NYG5 (Uniprot-TrEMBL)
ANAPC15 ProteinP60006 (Uniprot-TrEMBL)
ANAPC16 ProteinQ96DE5 (Uniprot-TrEMBL)
ANAPC2 ProteinQ9UJX6 (Uniprot-TrEMBL)
ANAPC4 ProteinQ9UJX5 (Uniprot-TrEMBL)
ANAPC5 ProteinQ9UJX4 (Uniprot-TrEMBL)
ANAPC7 ProteinQ9UJX3 (Uniprot-TrEMBL)
ATPMetaboliteCHEBI:15422 (ChEBI)
AdoHcyMetaboliteCHEBI:16680 (ChEBI)
AdoMetMetaboliteCHEBI:15414 (ChEBI)
CCNA1 ProteinP78396 (Uniprot-TrEMBL)
CCNA2 ProteinP20248 (Uniprot-TrEMBL)
CCNA:p-T160-CDK2ComplexR-HSA-187952 (Reactome)
CDC16 ProteinQ13042 (Uniprot-TrEMBL)
CDC23 ProteinQ9UJX2 (Uniprot-TrEMBL)
CDC26 ProteinQ8NHZ8 (Uniprot-TrEMBL)
CDC27 ProteinP30260 (Uniprot-TrEMBL)
CDK2 ProteinP24941 (Uniprot-TrEMBL)
CDK4 ProteinP11802 (Uniprot-TrEMBL)
CDK4,CDK6:INK4ComplexR-HSA-182579 (Reactome)
CDK4,CDK6ComplexR-HSA-69209 (Reactome)
CDK6 ProteinQ00534 (Uniprot-TrEMBL)
CDKN1A ProteinP38936 (Uniprot-TrEMBL)
CDKN1B ProteinP46527 (Uniprot-TrEMBL)
CDKN2B ProteinP42772 (Uniprot-TrEMBL)
CDKN2B gene ProteinENSG00000147883 (Ensembl)
CDKN2B geneGeneProductENSG00000147883 (Ensembl)
CDKN2BProteinP42772 (Uniprot-TrEMBL)
CDKN2C ProteinP42773 (Uniprot-TrEMBL)
CDKN2D ProteinP55273 (Uniprot-TrEMBL)
CEBPB geneGeneProductENSG00000172216 (Ensembl)
CEBPBProteinP17676 (Uniprot-TrEMBL)
Cdh1:phospho-APC/C complexComplexR-HSA-174250 (Reactome)
Cyclin

A:Cdk2:p21/p27

complex
ComplexR-HSA-187926 (Reactome)
Cyclin

A:phospho-Cdk(Thr

160):Cdh1:phosho-APC/C complex
ComplexR-HSA-188374 (Reactome)
Cyclin

A:phospho-Cdk2(Thr

160):phospho-Cdh1:phospho-APC/C complex
ComplexR-HSA-188387 (Reactome)
DNA Damage/Telomere

Stress Induced

Senescence
PathwayR-HSA-2559586 (Reactome) Reactive oxygen species (ROS), whose concentration increases in senescent cells due to oncogenic RAS-induced mitochondrial dysfunction (Moiseeva et al. 2009) or due to environmental stress, cause DNA damage in the form of double strand breaks (DSBs) (Yu and Anderson 1997). In addition, persistent cell division fueled by oncogenic signaling leads to replicative exhaustion, manifested in critically short telomeres (Harley et al. 1990, Hastie et al. 1990). Shortened telomeres are no longer able to bind the protective shelterin complex (Smogorzewska et al. 2000, de Lange 2005) and are recognized as damaged DNA.

The evolutionarily conserved MRN complex, consisting of MRE11A (MRE11), RAD50 and NBN (NBS1) subunits, binds DSBs (Lee and Paull 2005) and shortened telomeres that are no longer protected by shelterin (Wu et al. 2007). Once bound to the DNA, the MRN complex recruits and activates ATM kinase (Lee and Paull 2005, Wu et al. 2007), leading to phosphorylation of ATM targets, including TP53 (p53) (Banin et al. 1998, Canman et al. 1998, Khanna et al. 1998). TP53, phosphorylated on serine S15 by ATM, binds the CDKN1A (also known as p21, CIP1 or WAF1) promoter and induces CDKN1A transcription (El-Deiry et al. 1993, Karlseder et al. 1999). CDKN1A inhibits the activity of CDK2, leading to G1/S cell cycle arrest (Harper et al. 1993, El-Deiry et al. 1993).

SMURF2 is upregulated in response to telomere attrition in human fibroblasts and induces senecscent phenotype through RB1 and TP53, independently of its role in TGF-beta-1 signaling (Zhang and Cohen 2004). The exact mechanism of SMURF2 involvement is senescence has not been elucidated.

EHMT1 ProteinQ9H9B1 (Uniprot-TrEMBL)
EHMT1:EHMT2:Cdh1:p-APC/CComplexR-HSA-3788733 (Reactome)
EHMT1:EHMT2ComplexR-HSA-3788728 (Reactome)
EHMT2 ProteinQ96KQ7 (Uniprot-TrEMBL)
FZR1 ProteinQ9UM11 (Uniprot-TrEMBL)
H2AFB1 ProteinP0C5Y9 (Uniprot-TrEMBL)
H2AFJ ProteinQ9BTM1 (Uniprot-TrEMBL)
H2AFV ProteinQ71UI9 (Uniprot-TrEMBL)
H2AFX ProteinP16104 (Uniprot-TrEMBL)
H2AFZ ProteinP0C0S5 (Uniprot-TrEMBL)
H2BFS ProteinP57053 (Uniprot-TrEMBL)
H3F3A ProteinP84243 (Uniprot-TrEMBL)
HIST1H2AB ProteinP04908 (Uniprot-TrEMBL)
HIST1H2AC ProteinQ93077 (Uniprot-TrEMBL)
HIST1H2AD ProteinP20671 (Uniprot-TrEMBL)
HIST1H2AJ ProteinQ99878 (Uniprot-TrEMBL)
HIST1H2BA ProteinQ96A08 (Uniprot-TrEMBL)
HIST1H2BB ProteinP33778 (Uniprot-TrEMBL)
HIST1H2BC ProteinP62807 (Uniprot-TrEMBL)
HIST1H2BD ProteinP58876 (Uniprot-TrEMBL)
HIST1H2BH ProteinQ93079 (Uniprot-TrEMBL)
HIST1H2BJ ProteinP06899 (Uniprot-TrEMBL)
HIST1H2BK ProteinO60814 (Uniprot-TrEMBL)
HIST1H2BL ProteinQ99880 (Uniprot-TrEMBL)
HIST1H2BM ProteinQ99879 (Uniprot-TrEMBL)
HIST1H2BN ProteinQ99877 (Uniprot-TrEMBL)
HIST1H2BO ProteinP23527 (Uniprot-TrEMBL)
HIST1H3A ProteinP68431 (Uniprot-TrEMBL)
HIST1H4 ProteinP62805 (Uniprot-TrEMBL)
HIST2H2AA3 ProteinQ6FI13 (Uniprot-TrEMBL)
HIST2H2AC ProteinQ16777 (Uniprot-TrEMBL)
HIST2H2BE ProteinQ16778 (Uniprot-TrEMBL)
HIST2H3A ProteinQ71DI3 (Uniprot-TrEMBL)
HIST3H2BB ProteinQ8N257 (Uniprot-TrEMBL)
IGFBP7 gene ProteinENSG00000163453 (Ensembl)
IGFBP7 geneGeneProductENSG00000163453 (Ensembl)
IGFBP7ProteinQ16270 (Uniprot-TrEMBL)
IL1A gene ProteinENSG00000115008 (Ensembl)
IL1A geneGeneProductENSG00000115008 (Ensembl)
IL6 gene:Nucleosome-H3K9Me2ComplexR-HSA-3788744 (Reactome)
IL6 gene ProteinENSG00000136244 (Ensembl)
IL6 gene:NucleosomeComplexR-HSA-3788741 (Reactome)
IL6 geneGeneProductENSG00000136244 (Ensembl)
IL6ProteinP05231 (Uniprot-TrEMBL)
IL8 gene:Nucleosome-H3K9Me2ComplexR-HSA-3788746 (Reactome)
IL8 gene ProteinENSG00000169429 (Ensembl)
IL8 gene:NucleosomeComplexR-HSA-3788743 (Reactome)
IL8 geneGeneProductENSG00000169429 (Ensembl)
IL8ProteinP10145 (Uniprot-TrEMBL)
INK4ComplexR-HSA-182588 (Reactome)
Interleukin-1 family signalingPathwayR-HSA-446652 (Reactome) Interleukin 1 (IL1) signals via Interleukin 1 receptor 1 (IL1R1), the only signaling-capable IL1 receptor. This is a single chain type 1 transmembrane protein comprising an extracellular ligand binding domain and an intracellular region called the Toll/Interleukin-1 receptor (TIR) domain that is structurally conserved and shared by other members of the two families of receptors (Xu et al. 2000). This domain is also shared by the downstream adapter molecule MyD88. IL1 binding to IL1R1 leads to the recruitment of a second receptor chain termed the IL1 receptor accessory protein (IL1RAP or IL1RAcP) enabling the formation of a high-affinity ligand-receptor complex that is capable of signal transduction. Intracellular signaling is initiated by the recruitment of MyD88 to the IL-1R1/IL1RAP complex. IL1RAP is only recruited to IL1R1 when IL1 is present; it is believed that a TIR domain signaling complex is formed between the receptor and the adapter TIR domains. The recruitment of MyD88 leads to the recruitment of Interleukin-1 receptor-associated kinase (IRAK)-1 and -4, probably via their death domains. IRAK4 then activates IRAK1, allowing IRAK1 to autophosphorylate. Both IRAK1 and IRAK4 then dissociate from MyD88 (Brikos et al. 2007) which remains stably complexed with IL-1R1 and IL1RAP. They in turn interact with Tumor Necrosis Factor Receptor (TNFR)-Associated Factor 6 (TRAF6), which is an E3 ubiquitin ligase (Deng et al. 2000). TRAF6 is then thought to auto-ubiquinate, attaching K63-polyubiquitin to itself with the assistance of the E2 conjugating complex Ubc13/Uev1a. K63-pUb-TRAF6 recruits Transforming Growth Factor (TGF) beta-activated protein kinase 1 (TAK1) in a complex with TAK1-binding protein 2 (TAB2) and TAB3, which both contain nuclear zinc finger motifs that interact with K63-polyubiquitin chains (Ninomiya-Tsuji et al. 1999). This activates TAK1, which then activates inhibitor of NF-kappaB (IkappaB) kinase 2 (IKK2 or IKKB) within the IKK complex, the kinase responsible for phosphorylation of IkappaB. The IKK complex also contains the scaffold protein NF-kappa B essential modulator (NEMO). TAK1 also couples to the upstream kinases for p38 and c-jun N-terminal kinase (JNK). IRAK1 undergoes K63-linked polyubiquination; Pellino E3 ligases are important in this process. (Butler et al. 2007; Ordureau et al. 2008). The activity of these proteins is greatly enhanced by IRAK phosphorylation (Schauvliege et al. 2006), leading to K63-linked polyubiquitination of IRAK1. This recruits NEMO to IRAK1, with NEMO binding to polyubiquitin (Conze et al. 2008).

TAK1 activates IKKB (and IKK), resulting in phosphorylation of the inhibitory IkB proteins and enabling translocation of NFkB to the nucleus; IKKB also phosphorylates NFkB p105, leading to its degradation and the subsequent release of active TPL2 that triggers the extracellular-signal regulated kinase (ERK)1/2 MAPK cascade. TAK1 can also trigger the p38 and JNK MAPK pathways via activating the upstream MKKs3, 4 and 6. The MAPK pathways activate a number of downstream kinases and transcription factors that co-operate with NFkB to induce the expression of a range of TLR/IL-1R-responsive genes. There are reports suggesting that IL1 stimulation increases nuclear localization of IRAK1 (Bol et al. 2000) and that nuclear IRAK1 binds to the promoter of NFkB-regulated gene and IkBa, enhancing binding of the NFkB p65 subunit to NFkB responsive elements within the IkBa promoter. IRAK1 is required for IL1-induced Ser-10 phosphorylation of histone H3 in vivo (Liu et al. 2008). However, details of this aspect of IRAK1 signaling mechanisms remain unclear.
Interleukin-6 family signalingPathwayR-HSA-6783589 (Reactome) The interleukin-6 (IL6) family of cytokines includes IL6, IL11, IL27, leukemia inhibitory factor (LIF), oncostatin M (OSM), ciliary neurotrophic factor (CNTF), cardiotrophin 1 and 2 (CT-1) and cardiotrophin-like cytokine (CLC) (Heinrich et al. 2003, Pflanz et al. 2002). The latest addition to this family is IL31, discovered in 2004 (Dillon et al. 2004). The family is defined largely by the shared use of the common signal transducing receptor Interleukin-6 receptor subunit beta (IL6ST, gp130). The IL31 receptor uniquely does not include this subunit, instead it uses the related IL31RA. The members of the IL6 family share very low sequence homology but are structurally highly related, forming anti-parallel four-helix bundles with a characteristic “up-up-down-down� topology (Rozwarski et al. 1994, Cornelissen et al. 2012).

Although each member of the IL6 family signals through a distinct receptor complex, their underlying signaling mechanisms are similar. Assembly of the receptor complex is followed by activation of receptor-associated Janus kinases (JAKs), believed to be constitutively associated with the receptor subunits.Activation of JAKs initiates downstream cytoplasmic signaling cascades that involve recruitment and phosphorylation of transcription factors of the Signal transducer and activator of transcription (STAT) family, which dimerize and translocate to the nucleus where they bind enhancer elements of target genes leading to transcriptional activation (Nakashima & Taga 1998).

Negative regulators of IL-6 signaling include Suppressor of cytokine signals (SOCS) family members and PTPN11 (SHP-2).

IL6 is a pleiotropic cytokine with roles in processes including immune regulation, hematopoiesis, inflammation, oncogenesis, metabolic control and sleep.

IL6 and IL11 bind their corresponding specific receptors IL6R and IL11R respectively, resulting in dimeric complexes that associate with IL6ST, leading to IL6ST homodimer formation and signal initiation. IL6R alpha exists in transmembrane and soluble forms. The transmembrane form is mainly expressed by hepatocytes, neutrophils, monocytes/macrophages, and some lymphocytes. Soluble forms of IL6R (sIL6R) are also expressed by these cells. Two major mechanisms for the production of sIL6R have been proposed. Alternative splicing generates a transcript lacking the transmembrane domain by using splicing donor and acceptor sites that flank the transmembrane domain coding region. This also introduces a frameshift leading to the incorporation of 10 additional amino acids at the C terminus of sIL6R.A second mechanism for the generation of sIL6R is the proteolytic cleavage or 'shedding' of membrane-bound IL-6R. Two proteases ADAM10 and ADAM17 are thought to contribute to this (Briso et al. 2008). sIL6R can bind IL6 and stimulate cells that express gp130 but not IL6R alpha, a process that is termed trans-signaling. This explains why many cells, including hematopoietic progenitor cells, neuronal cells, endothelial cells, smooth muscle cells, and embryonic stem cells, do not respond to IL6 alone, but show a remarkable response to IL6/sIL6R. It is clear that the trans-signaling pathway is responsible for the pro-inflammatory activities of IL6 whereas the membrane bound receptor governs regenerative and anti-inflammatory IL6 activities

LIF, CNTF, OSM, CTF1, CRLF1 and CLCF1 signal via IL6ST:LIFR heterodimeric receptor complexes (Taga & Kishimoto 1997, Mousa & Bakhiet 2013). OSM signals via a receptor complex consisting of IL6ST and OSMR. These cytokines play important roles in the regulation of complex cellular processes such as gene activation, proliferation and differentiation (Heinrich et al. 1998).

Antibodies have been developed to inhibit IL6 activity for the treatment of inflammatory diseases (Kopf et al. 2010).
Me2K-10-H3F3A ProteinP84243 (Uniprot-TrEMBL)
Me2K-10-HIST2H3A ProteinQ71DI3 (Uniprot-TrEMBL)
Me2K10-HIST1H3A ProteinP68431 (Uniprot-TrEMBL)
NF-kB complexComplexR-HSA-194047 (Reactome)
NFKB1(1-433) ProteinP19838 (Uniprot-TrEMBL)
Oncogene Induced SenescencePathwayR-HSA-2559585 (Reactome) Oncogene-induced senescence is triggered by high level of RAS/RAF/MAPK signaling that can be caused, for example, by oncogenic mutations in RAS or RAF proteins, or by oncogenic mutations in growth factor receptors, such as EGFR, that act upstream of RAS/RAF/MAPK cascade. Oncogene-induced senescence can also be triggered by high transcriptional activity of E2F1, E2F2 or E2F3 which can be caused, for example, by the loss-of-function of RB1 tumor suppressor.

Oncogenic signals trigger transcription of CDKN2A locus tumor suppressor genes: p16-INK4A and p14-ARF. p16-INK4A and p14-ARF share exons 2 and 3, but are expressed from different promoters and use different reading frames (Quelle et al. 1995). Therefore, while their mRNAs are homologous and are both translationally inhibited by miR-24 microRNA (Lal et al. 2008, To et al. 2012), they share no similarity at the amino acid sequence level and perform distinct functions in the cell. p16-INK4A acts as the inhibitor of cyclin-dependent kinases CDK4 and CDK6 which phosphorylate and inhibit RB1 protein thereby promoting G1 to S transition and cell cycle progression (Serrano et al. 1993). Increased p16-INK4A level leads to hypophosphorylation of RB1, allowing RB1 to inhibit transcription of E2F1, E2F2 and E2F3-target genes that are needed for cell cycle progression, which results in cell cycle arrest in G1 phase. p14-ARF binds and destabilizes MDM2 ubiquitin ligase (Zhang et al. 1998), responsible for ubiquitination and degradation of TP53 (p53) tumor suppressor protein (Wu et al. 1993, Fuchs et al. 1998, Fang et al. 2000). Therefore, increased p14-ARF level leads to increased level of TP53 and increased expression of TP53 target genes, such as p21, which triggers p53-mediated cell cycle arrest and, depending on other factors, may also lead to p53-mediated apoptosis. CDKN2B locus, which encodes an inhibitor of CDK4 and CDK6, p15-INK4B, is located in the vicinity of CDKN2A locus, at the chromosome band 9p21. p15-INK4B, together with p16-INK4A, contributes to senescence of human T-lymphocytes (Erickson et al. 1998) and mouse fibroblasts (Malumbres et al. 2000). SMAD3, activated by TGF-beta-1 signaling, controls senescence in the mouse multistage carcinogenesis model through regulation of MYC and p15-INK4B gene expression (Vijayachandra et al. 2003). TGF-beta-induced p15-INK4B expression is also important for the senescence of hepatocellular carcinoma cell lines (Senturk et al. 2010).

MAP kinases MAPK1 (ERK2) and MAPK3 (ERK1), which are activated by RAS signaling, phosphorylate ETS1 and ETS2 transcription factors in the nucleus (Yang et al. 1996, Seidel et al. 2002, Foulds et al. 2004, Nelson et al. 2010). Phosphorylated ETS1 and ETS2 are able to bind RAS response elements (RREs) in the CDKN2A locus and stimulate p16-INK4A transcription (Ohtani et al. 2004). At the same time, activated ERKs (MAPK1 i.e. ERK2 and MAPK3 i.e. ERK1) phosphorylate ERF, the repressor of ETS2 transcription, which leads to translocation of ERF to the cytosol and increased transcription of ETS2 (Sgouras et al. 1995, Le Gallic et al. 2004). ETS2 can be sequestered and inhibited by binding to ID1, resulting in inhibition of p16-INK4A transcription (Ohtani et al. 2004).

Transcription of p14-ARF is stimulated by binding of E2F transcription factors (E2F1, E2F2 or E2F3) in complex with SP1 to p14-ARF promoter (Parisi et al. 2002).

Oncogenic RAS signaling affects mitochondrial metabolism through an unknown mechanism, leading to increased generation of reactive oxygen species (ROS), which triggers oxidative stress induced senescence pathway. In addition, increased rate of cell division that is one of the consequences of oncogenic signaling, leads to telomere shortening which acts as another senescence trigger.

Oncogenic MAPK signalingPathwayR-HSA-6802957 (Reactome) The importance of the RAS/RAF/MAPK cascade in regulating cellular proliferation, differentiation and survival is highlighted by the fact that components of the 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. RAS pathway activation is also achieved in a smaller subset of cancers by loss-of-function mutations in negative regulators of RAS signaling, such as the RAS GAP NF1(reviewed in Prior et al, 2012; Pylayeva-Gupta et al, 2011; Stephen et al, 2014; Lavoie and Therrien, 2015; Lito et al, 2013; Samatar and Poulikakos, 2014; Maertens and Cichowski, 2014).
Oxidative Stress Induced SenescencePathwayR-HSA-2559580 (Reactome) Oxidative stress, caused by increased concentration of reactive oxygen species (ROS) in the cell, can happen as a consequence of mitochondrial dysfunction induced by the oncogenic RAS (Moiseeva et al. 2009) or independent of oncogenic signaling. Prolonged exposure to interferon-beta (IFNB, IFN-beta) also results in ROS increase (Moiseeva et al. 2006). ROS oxidize thioredoxin (TXN), which causes TXN to dissociate from the N-terminus of MAP3K5 (ASK1), enabling MAP3K5 to become catalytically active (Saitoh et al. 1998). ROS also stimulate expression of Ste20 family kinases MINK1 (MINK) and TNIK through an unknown mechanism, and MINK1 and TNIK positively regulate MAP3K5 activation (Nicke et al. 2005).


MAP3K5 phosphorylates and activates MAP2K3 (MKK3) and MAP2K6 (MKK6) (Ichijo et al. 1997, Takekawa et al. 2005), which act as p38 MAPK kinases, as well as MAP2K4 (SEK1) (Ichijo et al. 1997, Matsuura et al. 2002), which, together with MAP2K7 (MKK7), acts as a JNK kinase.


MKK3 and MKK6 phosphorylate and activate p38 MAPK alpha (MAPK14) and beta (MAPK11) (Raingeaud et al. 1996), enabling p38 MAPKs to phosphorylate and activate MAPKAPK2 (MK2) and MAPKAPK3 (MK3) (Ben-Levy et al. 1995, Clifton et al. 1996, McLaughlin et al. 1996, Sithanandam et al. 1996, Meng et al. 2002, Lukas et al. 2004, White et al. 2007), as well as MAPKAPK5 (PRAK) (New et al. 1998 and 2003, Sun et al. 2007).


Phosphorylation of JNKs (MAPK8, MAPK9 and MAPK10) by MAP3K5-activated MAP2K4 (Deacon and Blank 1997, Fleming et al. 2000) allows JNKs to migrate to the nucleus (Mizukami et al. 1997) where they phosphorylate JUN. Phosphorylated JUN binds FOS phosphorylated by ERK1 or ERK2, downstream of activated RAS (Okazaki and Sagata 1995, Murphy et al. 2002), forming the activated protein 1 (AP-1) complex (FOS:JUN heterodimer) (Glover and Harrison 1995, Ainbinder et al. 1997).


Activation of p38 MAPKs and JNKs downstream of MAP3K5 (ASK1) ultimately converges on transcriptional regulation of CDKN2A locus. In dividing cells, nucleosomes bound to the CDKN2A locus are trimethylated on lysine residue 28 of histone H3 (HIST1H3A) by the Polycomb repressor complex 2 (PRC2), creating the H3K27Me3 (Me3K-28-HIST1H3A) mark (Bracken et al. 2007, Kotake et al. 2007). The expression of Polycomb constituents of PRC2 (Kuzmichev et al. 2002) - EZH2, EED and SUZ12 - and thereby formation of the PRC2, is positively regulated in growing cells by E2F1, E2F2 and E2F3 (Weinmann et al. 2001, Bracken et al. 2003). H3K27Me3 mark serves as a docking site for the Polycomb repressor complex 1 (PRC1) that contains BMI1 (PCGF4) and is therefore named PRC1.4, leading to the repression of transcription of p16-INK4A and p14-ARF from the CDKN2A locus, where PCR1.4 mediated repression of p14-ARF transcription in humans may be context dependent (Voncken et al. 2005, Dietrich et al. 2007, Agherbi et al. 2009, Gao et al. 2012). MAPKAPK2 and MAPKAPK3, activated downstream of the MAP3K5-p38 MAPK cascade, phosphorylate BMI1 of the PRC1.4 complex, leading to dissociation of PRC1.4 complex from the CDKN2A locus and upregulation of p14-ARF transcription (Voncken et al. 2005). AP-1 transcription factor, formed as a result of MAP3K5-JNK signaling, as well as RAS signaling, binds the promoter of KDM6B (JMJD3) gene and stimulates KDM6B expression. KDM6B is a histone demethylase that removes H3K27Me3 mark i.e. demethylates lysine K28 of HIST1H3A, thereby preventing PRC1.4 binding to the CDKN2A locus and allowing transcription of p16-INK4A (Agger et al. 2009, Barradas et al. 2009, Lin et al. 2012).


p16-INK4A inhibits phosphorylation-mediated inactivation of RB family members by CDK4 and CDK6, leading to cell cycle arrest (Serrano et al. 1993). p14-ARF inhibits MDM2-mediated degradation of TP53 (p53) (Zhang et al. 1998), which also contributes to cell cycle arrest in cells undergoing oxidative stress. In addition, phosphorylation of TP53 by MAPKAPK5 (PRAK) activated downstream of MAP3K5-p38 MAPK signaling, activates TP53 and contributes to cellular senescence (Sun et al. 2007).

Phospho-Ribosomal protein S6 kinaseComplexR-HSA-199849 (Reactome)
RELA ProteinQ04206 (Uniprot-TrEMBL)
RPS27A(1-76) ProteinP62979 (Uniprot-TrEMBL)
RPS6KA1 ProteinQ15418 (Uniprot-TrEMBL)
RPS6KA2 ProteinQ15349 (Uniprot-TrEMBL)
RPS6KA3 ProteinP51812 (Uniprot-TrEMBL)
Ribosomal protein S6 kinaseComplexR-HSA-199858 (Reactome)
UBA52(1-76) ProteinP62987 (Uniprot-TrEMBL)
UBB(1-76) ProteinP0CG47 (Uniprot-TrEMBL)
UBB(153-228) ProteinP0CG47 (Uniprot-TrEMBL)
UBB(77-152) ProteinP0CG47 (Uniprot-TrEMBL)
UBC(1-76) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(153-228) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(229-304) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(305-380) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(381-456) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(457-532) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(533-608) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(609-684) ProteinP0CG48 (Uniprot-TrEMBL)
UBC(77-152) ProteinP0CG48 (Uniprot-TrEMBL)
UBE2C ProteinO00762 (Uniprot-TrEMBL)
UBE2D1 ProteinP51668 (Uniprot-TrEMBL)
UBE2E1 ProteinP51965 (Uniprot-TrEMBL)
Ub-EHMT1:Ub-EHMT2:Cdh1:p-APC/CComplexR-HSA-3788739 (Reactome)
UbComplexR-HSA-68524 (Reactome)
p-2S-JUN:p-2S,2T-FOS:IGFBP7 GeneComplexR-HSA-3797203 (Reactome)
p-2S-JUN:p-2S,2T-FOS:IL1A geneComplexR-HSA-4568738 (Reactome)
p-2S-cJUN:p-2S,2T-cFOSComplexR-HSA-450327 (Reactome)
p-4S,T231,T365-RPS6KA3 ProteinP51812 (Uniprot-TrEMBL)
p-4S,T356,T570-RPS6KA2 ProteinQ15349 (Uniprot-TrEMBL)
p-4S,T359,T573-RPS6KA1 ProteinQ15418 (Uniprot-TrEMBL)
p-ERK1/2/5ComplexR-HSA-199878 (Reactome)
p-FZR1 ProteinQ9UM11 (Uniprot-TrEMBL)
p-S63,S73-JUN ProteinP05412 (Uniprot-TrEMBL)
p-T,Y MAPK dimersComplexR-HSA-198701 (Reactome)
p-T160-CDK2 ProteinP24941 (Uniprot-TrEMBL)
p-T185,Y187-MAPK1 ProteinP28482 (Uniprot-TrEMBL)
p-T202,Y204-MAPK3 ProteinP27361 (Uniprot-TrEMBL)
p-T218,Y220-MAPK7 ProteinQ13164 (Uniprot-TrEMBL)
p-T235, S321-CEBPB ProteinP17676 (Uniprot-TrEMBL)
p-T235, S321-CEBPBProteinP17676 (Uniprot-TrEMBL)
p-T235,S321-CEBPB homodimerComplexR-HSA-3857334 (Reactome)
p-T235,S321-CEBPB:CDKN2B GeneComplexR-HSA-3857347 (Reactome)
p-T235,S321-CEBPB:NF-kB:IL6 geneComplexR-HSA-3857324 (Reactome)
p-T235,S321-CEBPB:NF-kB:IL8 GeneComplexR-HSA-3857319 (Reactome)
p-T235-CEBPBProteinP17676 (Uniprot-TrEMBL)
p-T325,T331,S362,S374-FOS ProteinP01100 (Uniprot-TrEMBL)
p-Y705-STAT3 ProteinP40763 (Uniprot-TrEMBL)
p-Y705-STAT3 dimerComplexR-HSA-1112525 (Reactome)
p16-INK4a ProteinP42771 (Uniprot-TrEMBL)

Annotated Interactions

View all...
SourceTargetTypeDatabase referenceComment
2xMyri-IL1AArrowR-HSA-4568740 (Reactome)
ADPArrowR-HSA-198746 (Reactome)
ADPArrowR-HSA-3788705 (Reactome)
ADPArrowR-HSA-3857328 (Reactome)
ADPArrowR-HSA-3857329 (Reactome)
ATPR-HSA-198746 (Reactome)
ATPR-HSA-3788705 (Reactome)
ATPR-HSA-3857328 (Reactome)
ATPR-HSA-3857329 (Reactome)
AdoHcyArrowR-HSA-3788745 (Reactome)
AdoHcyArrowR-HSA-3788748 (Reactome)
AdoMetR-HSA-3788745 (Reactome)
AdoMetR-HSA-3788748 (Reactome)
ArrowR-HSA-3790130 (Reactome)
ArrowR-HSA-3790137 (Reactome)
CCNA:p-T160-CDK2R-HSA-3788708 (Reactome)
CDK4,CDK6:INK4ArrowR-HSA-182594 (Reactome)
CDK4,CDK6R-HSA-182594 (Reactome)
CDKN2B geneR-HSA-3857345 (Reactome)
CDKN2B geneR-HSA-3857348 (Reactome)
CDKN2BArrowR-HSA-3857348 (Reactome)
CEBPB geneR-HSA-3858387 (Reactome)
CEBPBArrowR-HSA-3858387 (Reactome)
CEBPBR-HSA-3857329 (Reactome)
Cdh1:phospho-APC/C complexR-HSA-3788708 (Reactome)
Cdh1:phospho-APC/C complexR-HSA-3788725 (Reactome)
Cyclin

A:Cdk2:p21/p27

complex
TBarR-HSA-3788705 (Reactome)
Cyclin

A:Cdk2:p21/p27

complex
TBarR-HSA-3788708 (Reactome)
Cyclin

A:phospho-Cdk(Thr

160):Cdh1:phosho-APC/C complex
ArrowR-HSA-3788708 (Reactome)
Cyclin

A:phospho-Cdk(Thr

160):Cdh1:phosho-APC/C complex
R-HSA-3788705 (Reactome)
Cyclin

A:phospho-Cdk(Thr

160):Cdh1:phosho-APC/C complex
mim-catalysisR-HSA-3788705 (Reactome)
Cyclin

A:phospho-Cdk2(Thr

160):phospho-Cdh1:phospho-APC/C complex
ArrowR-HSA-3788705 (Reactome)
EHMT1:EHMT2:Cdh1:p-APC/CArrowR-HSA-3788725 (Reactome)
EHMT1:EHMT2:Cdh1:p-APC/CR-HSA-3788724 (Reactome)
EHMT1:EHMT2:Cdh1:p-APC/Cmim-catalysisR-HSA-3788724 (Reactome)
EHMT1:EHMT2R-HSA-3788725 (Reactome)
EHMT1:EHMT2TBarR-HSA-3790130 (Reactome)
EHMT1:EHMT2TBarR-HSA-3790137 (Reactome)
EHMT1:EHMT2mim-catalysisR-HSA-3788745 (Reactome)
EHMT1:EHMT2mim-catalysisR-HSA-3788748 (Reactome)
IGFBP7 geneR-HSA-3797196 (Reactome)
IGFBP7 geneR-HSA-3797202 (Reactome)
IGFBP7ArrowR-HSA-3797202 (Reactome)
IL1A geneR-HSA-4568737 (Reactome)
IL1A geneR-HSA-4568740 (Reactome)
IL6 gene:Nucleosome-H3K9Me2ArrowR-HSA-3788748 (Reactome)
IL6 gene:NucleosomeR-HSA-3788748 (Reactome)
IL6 geneR-HSA-3790130 (Reactome)
IL6 geneR-HSA-3857305 (Reactome)
IL6ArrowR-HSA-3790130 (Reactome)
IL8 gene:Nucleosome-H3K9Me2ArrowR-HSA-3788745 (Reactome)
IL8 gene:NucleosomeR-HSA-3788745 (Reactome)
IL8 geneR-HSA-3790137 (Reactome)
IL8 geneR-HSA-3857308 (Reactome)
IL8ArrowR-HSA-3790137 (Reactome)
INK4R-HSA-182594 (Reactome)
NF-kB complexR-HSA-3857305 (Reactome)
NF-kB complexR-HSA-3857308 (Reactome)
Phospho-Ribosomal protein S6 kinaseArrowR-HSA-198746 (Reactome)
Phospho-Ribosomal protein S6 kinasemim-catalysisR-HSA-3857328 (Reactome)
R-HSA-182594 (Reactome) Prior to mitogen activation, the inhibitory proteins of the INK4 family (p15, p16, p18, and p19) associate with the catalytic domains of free CDK4 and CDK6, preventing their association with D type cyclins (CCND1, CCND2 and CCND3), and thus their activation (Serrano et al. 1993, Hannon and Beach 1994, Guan et al. 1994, Guan et al. 1996, Parry et al. 1995). Inactivation and defects of RB1 strongly upregulate p16-INK4A (Parry et al. 1995).
R-HSA-198746 (Reactome) The p90 ribosomal S6 kinases (RSK1-4) comprise a family of serine/threonine kinases that lie at the terminus of the ERK pathway. RSK family members are unusual among serine/threonine kinases in that they contain two distinct kinase domains, both of which are catalytically functional . The C-terminal kinase domain is believed to be involved in autophosphorylation, a critical step in RSK activation, whereas the N-terminal kinase domain, which is homologous to members of the AGC superfamily of kinases, is responsible for the phosphorylation of all known exogenous substrates of RSK.
RSKs can be activated by the ERKs (ERK1, 2, 5) in the cytoplasm as well as in the nucleus, they both have cytoplasmic and nuclear substrates, and they are able to move from nucleus to cytoplasm. Efficient RSK activation by ERKs requires its interaction through a docking site located near the RSK C terminus. The mechanism of RSK activation has been studied mainly with regard to ERK1 and ERK2. RSK activation leads to the phosphorylation of four essential residues Ser239, Ser381, Ser398, and Thr590, and two additional sites, Thr377 and Ser749 (the amino acid numbering refers to RSK1). ERK is thought to play at least two roles in RSK1 activation. First, activated ERK phosphorylates RSK1 on Thr590, and possibly on Thr377 and Ser381, and second, ERK brings RSK1 into close proximity to membrane-associated kinases that may phosphorylate RSK1 on Ser381 and Ser398.
Moreover, RSKs and ERK1/2 form a complex that transiently dissociates upon growth factor signalling. Complex dissociation requires phosphorylation of RSK1 serine 749, a growth factor regulated phosphorylation site located near the ERK docking site. Serine 749 is phosphorylated by the N-terminal kinase domain of RSK1 itself. ERK1/2 docking to RSK2 and RSK3 is also regulated in a similar way. The length of RSK activation following growth factor stimulation depends on the duration of the RSK/ERK complex, which, in turn, differs among the different RSK isoforms. RSK1 and RSK2 readily dissociate from ERK1/2 following growth factor stimulation stimulation, but RSK3 remains associated with active ERK1/2 longer, and also remains active longer than RSK1 and RSK2.

R-HSA-3788705 (Reactome) At the G1/S transition, the Cdh1 (FZR1) subunit of the APC/C:Cdh1 complex is phosphorylated by Cyclin A:Cdk2 (CCNA:CDK2) and dissociates from APC/C. This inactivates APC/C and permits the accumulation of cell cycle proteins required for DNA synthesis and entry into mitosis (Lukas et al. 1999). Activation of the ATM kinase by DNA damage in the form of double strand breaks results in TP53-mediated induction of CDKN1A (p21) expression. CDKN1A binds CCNA:CDK2 complex and prevents it from phosphorylating Cdh1 (Takahashi et al. 2012).
R-HSA-3788708 (Reactome) Cyclin A-Cdk2 (CCNA:CDK2) prevents unscheduled APC reactivation during S phase by binding and subsequently phosphorylating FZR1 (Cdh1). Phosphorylation-dependent dissociation of the Cdh1-activating subunit inhibits the APC/C (Sorensen et al. 2001). DNA damage activates ATM kinase, resulting in TP53-mediated induction of CDKN1A (p21) expression. CDKN1A binds CCNA:CDK2 complex and prevents its association with Cdh1 (Takahashi et al. 2012).
R-HSA-3788724 (Reactome) Cdh1:APC/C complex, stabilized by the DNA damage-induced ATM-TP53-CDKN1A axis, ubiquitinates EHMT1 (GLP) and EHMT2 (G9a) histone methyltransferases, targeting them for degradation (Takahashi et al. 2012).
R-HSA-3788725 (Reactome) Cdh1 (FZR1) is able to bind both G9a (EHMT2) and GLP (EHMT1) (Takahashi et al. 2012). EHMT1 and EHMT2 histone methyltransferases were shown to function as a heterodimer in vivo (Tachibana et al. 2005).
R-HSA-3788745 (Reactome) EHMT1 (GLP) and EHMT2 (G9a) histone methyltransferases dimethylate histone H3 (HIST1H3A) on lysine residue 10, creating an H3K9Me2 mark on nucleosomes associated with the IL8 promoter (Takahashi et al. 2012).
R-HSA-3788748 (Reactome) EHMT1 (GLP) and EHMT2 (G9a) histone methyltransferases dimethylate histone H3 (HIST1H3A) on lysine residue 10, creating an H3K9Me2 mark on nucleosomes associated with the IL6 promoter (Takahashi et al. 2012).
R-HSA-3790130 (Reactome) Methylation of the IL6 promoter by EHMT1:EHMT2 (GLP:G9a) histone methyltransferases inhibits IL6 transcription, while Cdh1:APC/C-mediated degradation of EHTM1:EHTM2 downstream of the ATM-TP53-CDKN1A axis stimulates IL6 transcription (Takahashi et al. 2012). Oncogenic RAS signaling stimulates activation of the CEBPB transcription factor (C/EBP-beta) which binds IL6 promoter and stimulates IL6 transcription (Kuilman et al. 2008, Lee et al. 2010). NF kappa B transcription factor is also activated in senescent cells (Chien et al. 2011) through interleukin-1-alpha (IL1A) signaling (Jimi et al. 1996, Hartupee et al. 2008, Orjalo et al. 2009), and it cooperates with CEBPB in the activation of IL6 transcription (Shimizu et al. 1990, Libermann and Baltimore 1990, Matsusaka et al. 1993, Acosta et al. 2008). Autocrine IL6 signaling stimulates CEBPB expression (Kuilman et al. 2008), creating a positive feedback loop. STAT3, activated by IL6 signaling cascade is necessary for CEBPB transcription, but the direct binding of STAT3 to the CEBPB promoter has not been demonstrated (Niehof et al. 2001).
R-HSA-3790137 (Reactome) Methylation of the IL8 promoter by EHMT1:EHMT2 (GLP:G9a) histone methyltransferases inhibits IL8 transcription, while Cdh1:APC/C-mediated degradation of EHTM1:EHTM2 downstream of the ATM-TP53-CDKN1A axis stimulates IL8 transcription (Takahashi et al. 2012). CEBPB transcription factor, activated by oncogenic RAS signaling, binds IL8 promoter and stimulates IL8 transcription (Kuilman et al. 2008). The NF-kappa-B transcription factor is also activated in senescent cells (Chien et al. 2011) through interleukin-1-alpha (IL1A) signaling (Jimi et al. 1996, Hartupee et al. 2008, Orjalo et al. 2009), and it cooperates with CEBPB in the activation of IL8 transcription (Kunsch and Rosen 1993, Stein and Baldwin 1993, Matsusaka et al. 1993, Acosta et al. 2008).
R-HSA-3797196 (Reactome) FOS:JUN (AP-1) transcription factor, formed in response to oncogenic RAF-MAPK signaling which also triggers oxidative stress, binds the promoter of IGFBP7 gene (Wajapeyee et al. 2008).
R-HSA-3797202 (Reactome) FOS:JUN (AP-1) transcription factor stimulates the transcription of IGFBP7 gene. IGFBP7 is a component of the senescence-associated secretory phenotype (SASP) and is secreted by senescent melanocytes in which the senescence is induced by the expression of oncogenic BRAF V600E. The BRAF V600E-mediated induction of IGFBP7 expression is AP-1 dependent. The conditioned medium harvested from BRAF V600E senescent melanocytes is able to inhibit cellular proliferation and induce senescence of naive melanocytes only when IGFBP7 is present in the medium (Wajapeyee et al. 2008).
R-HSA-3857305 (Reactome) RSK6A1/2/3-mediated phosphorylation of CEBPB downstream of activated RAS stimulates CEBPB homodimerization and DNA binding (Lee, Shuman et al. 2010) and, specifically, RAS-induced CEBPB activation stimulates CEBPB binding to the IL6 promoter (Kuilman et al. 2008; Lee, Shuman et al. 2010). RAS-activated CEBPB is able to recruit additional transcription activators, such as EP300, to the IL6 promoter (Lee, Miller et al. 2010). NFKB transcription complex, activated by interleukin-1-alpha (IL1A) signaling (Jimi et al. 1996, Hartupee et al. 2008, Orjalo et al. 2009), also binds the promoter of the IL6 gene (Shimizu et al. 1990, Libermann and Baltimore 1990) and cooperates with CEBPB in the activation of IL6 transcription (Matsusaka et al. 1993).
R-HSA-3857308 (Reactome) RAS-mediated activation of CEBPB (C/EBP-beta) stimulates CEBPB binding to the IL8 promoter (Kuilman et al. 2008). NFKB transcription complex, activated by interleukin-1-alpha (IL1A) signaling (Jimi et al. 1996, Hartupee et al. 2008, Orjalo et al. 2009), also binds the promoter of the IL8 gene (Kunsch and Rosen 1993) and cooperates with CEBPB in the activation of IL8 transcription (Matsusaka et al. 1993, Stein and Baldwin 1993).
R-HSA-3857328 (Reactome) Phosphorylation of CEBPB (C/EBP-beta) serine residue S321 by ERK1/2-activated RSK1, RSK2 or RSK3, downstream of activated RAS, is necessary for the relief of CEBPB autoinhibiton (Lee et al. 2010). Phosphorylation on other sites may also be involved in CEBPB activation.
R-HSA-3857329 (Reactome) Phosphorylation of CEBPB (C/EBP-beta) transcription factor on threonine residue T235 happens downstream of activated RAS, is mediated by MAPKs - likely MAPK3 (ERK1) and MAPK1 (ERK2), and positively affects CEBPB-mediated transcription of IL6 (Nakajima et al. 1993).
R-HSA-3857336 (Reactome) RSK1/2/3-mediated phosphorylation of CEBPB promotes the formation of CEBPB homodimers which are active as transcription factors (Lee, Miller et al. 2010; Lee, Shuman et al. 2010).
R-HSA-3857345 (Reactome) The CEBPB transcription factor, activated by oncogenic RAS signaling, binds the CDKN2B promoter (Kuilman et al. 2008).
R-HSA-3857348 (Reactome) Once bound to the CDKN2B promoter, CEBPB stimulates CKDN2B transcription, contributing to cell cycle arrest in oncogene induced senescence (Kuilman et al. 2008). In addition, since CEBPB expression is stimulated by IL6 signaling, with IL6 itself being a transcriptional target of CEBPB (Neihof et al. 2001, Kuilman et al. 2008, Lee et al. 2010), IL6-CEBPB-CDKN2B axis provides an autocrine SASP-mediated growth arrest mechanism.
R-HSA-3858387 (Reactome) STAT3, activated by IL6 signaling cascade, is necessary for CEBPB transcription, but direct binding of STAT3 to the CEBPB promoter has not been demonstrated (Niehof et al. 2001). As CEBPB activates IL6 transcription in response to oncogenic RAS signaling, and IL6 activates CEBPB transcription (Niehof et al. 2001, Kuilman et al. 2008, Lee et al. 2010), a positive feedback loop exists between CEBPB and IL6.
R-HSA-4568737 (Reactome) The promoter of the interleukin 1-alpha (IL1A) gene contains AP-1 binding sites which are occupied by the AP-1 (FOS:JUN) complex, resulting in the stimulation of IL1A transcription (Bailly et al. 1996 Alheim et al. 1996, Burleson et al. 1996, Huang et al. 1999, Niu et al. 2004, Moerman-Herzog and Barger 2012, Murakami et al. 2013).
R-HSA-4568740 (Reactome) The AP-1 complex (FOS:JUN) binds IL1A promoter and stimulates IL1A transcription (Bailly et al. 1996 Alheim et al. 1996, Burleson et al. 1996, Huang et al. 1999, Niu et al. 2004, Moerman-Herzog and Barger 2012, Murakami et al. 2013).
Ribosomal protein S6 kinaseR-HSA-198746 (Reactome)
Ub-EHMT1:Ub-EHMT2:Cdh1:p-APC/CArrowR-HSA-3788724 (Reactome)
UbR-HSA-3788724 (Reactome)
p-2S-JUN:p-2S,2T-FOS:IGFBP7 GeneArrowR-HSA-3797196 (Reactome)
p-2S-JUN:p-2S,2T-FOS:IGFBP7 GeneArrowR-HSA-3797202 (Reactome)
p-2S-JUN:p-2S,2T-FOS:IL1A geneArrowR-HSA-4568737 (Reactome)
p-2S-JUN:p-2S,2T-FOS:IL1A geneArrowR-HSA-4568740 (Reactome)
p-2S-cJUN:p-2S,2T-cFOSR-HSA-3797196 (Reactome)
p-2S-cJUN:p-2S,2T-cFOSR-HSA-4568737 (Reactome)
p-ERK1/2/5mim-catalysisR-HSA-198746 (Reactome)
p-T,Y MAPK dimersmim-catalysisR-HSA-3857329 (Reactome)
p-T235, S321-CEBPBArrowR-HSA-3857328 (Reactome)
p-T235, S321-CEBPBR-HSA-3857336 (Reactome)
p-T235,S321-CEBPB homodimerArrowR-HSA-3857336 (Reactome)
p-T235,S321-CEBPB homodimerR-HSA-3857305 (Reactome)
p-T235,S321-CEBPB homodimerR-HSA-3857308 (Reactome)
p-T235,S321-CEBPB homodimerR-HSA-3857345 (Reactome)
p-T235,S321-CEBPB:CDKN2B GeneArrowR-HSA-3857345 (Reactome)
p-T235,S321-CEBPB:CDKN2B GeneArrowR-HSA-3857348 (Reactome)
p-T235,S321-CEBPB:NF-kB:IL6 geneArrowR-HSA-3790130 (Reactome)
p-T235,S321-CEBPB:NF-kB:IL6 geneArrowR-HSA-3857305 (Reactome)
p-T235,S321-CEBPB:NF-kB:IL8 GeneArrowR-HSA-3790137 (Reactome)
p-T235,S321-CEBPB:NF-kB:IL8 GeneArrowR-HSA-3857308 (Reactome)
p-T235-CEBPBArrowR-HSA-3857329 (Reactome)
p-T235-CEBPBR-HSA-3857328 (Reactome)
p-Y705-STAT3 dimerArrowR-HSA-3858387 (Reactome)

Personal tools